DataPDF Available

Hormonal Regulation of TNF in Alzheimer's Disease

Authors:
ASSOCIATE EDITOR: DAVID R. SIBLEY
Tumor Necrosis Factor-Induced Cerebral Insulin
Resistance in Alzheimer’s Disease Links Numerous
Treatment Rationales
Ian Clark, Craig Atwood, Richard Bowen, Gilberto Paz-Filho, and Bryce Vissel
Research School of Biology, Australian National University, Canberra, Australia (I.C.); Department of Medicine, University of Wisconsin-
Madison School of Medicine and Public Health, Madison, Wisconsin (C.A.); School of Exercise, Biomedical and Health Sciences, Edith
Cowan University, Joondalup, Australia (C.A.); OTB Research, Charleston, South Carolina (R.B.); John Curtin School of Medical
Research, Australian National University, Canberra, Australia (G.P.-F.); and the Neurodegeneration Research Group, Garvan Institute of
Medical Research, Sydney, Australia (B.V.)
Abstract............................................................................... B
I. Introduction ........................................................................... B
II. Gonadotropins, sex steroids, tumor necrosis factor, and Alzheimer’s disease.................. B
III. Tumor necrosis factor and Alzheimer’s disease ............................................ C
IV. Tumor necrosis factor, amyloid
, and
.................................................. D
V. Insulin................................................................................ E
A. Insulin in basic biology and the brain................................................. E
B. Insulin resistance and Alzheimer’s disease ............................................ E
C. Tumor necrosis factor and insulin resistance .......................................... F
D. Functional links of glucagon-like peptide-1 to insulin resistance ......................... G
VI. Tumor necrosis factor and glycogen synthase kinase-3 ..................................... G
VII. Tumor necrosis factor and mitochondrial dysfunction ...................................... H
VIII. Tumor necrosis factor and progenitor cells................................................ H
A. Progenitor cells, tumor necrosis factor, and insulin resistance ........................... H
B. Clock genes, controlled by tumor necrosis factor, govern progenitor activity............... I
IX. Streptozotocin ......................................................................... J
A. Streptozotocin model for diabetes and Alzheimer’s disease .............................. J
B. Streptozotocin induces tumor necrosis factor........................................... J
X. The broader picture—stroke, traumatic brain injury, and infectious disease.................. K
XI. Therapeutic implications................................................................ K
A. Specific inhibition of tumor necrosis factor ............................................ K
B. Nonspecific inhibition of tumor necrosis factor ......................................... L
1. Thalidomide and curcumin........................................................ L
2. Minocycline ..................................................................... L
3. Erythropoietin ................................................................... L
C. Administering leptin as a counter to insulin resistance ................................. M
D. Administering insulin as a counter to insulin resistance ................................ N
E. Glucagon-like peptide-1 mimetics and dipeptidyl peptidase-4 inhibitors as counters to
insulin resistance ................................................................... N
F. Glycogen synthase kinase-3 antagonists............................................... N
H. Apolipoprotein E mimetics and bexarotene ............................................ O
XII. Conclusions ........................................................................... P
References ............................................................................ P
Address correspondence to: Prof. Ian Clark, Division of Medical Science and Biochemistry, Research School of Biology, Australian National
University, Canberra ACT 0200, Australia. E-mail: ian.clark@anu.edu.au
This article is available online at http://pharmrev.aspetjournals.org.
http://dx.doi.org/10.1124/pr.112.005850.
1521-0081/12/6404-A–W$25.00
PHARMACOLOGICAL REVIEWS Vol. 64, No. 4
Copyright © 2012 by The American Society for Pharmacology and Experimental Therapeutics 5850/3792099
Pharmacol Rev 64:A–W, 2012
A
Pharmrev Fast Forward. Published on September 10, 2012 as DOI:10.1124/pr.112.005850
Copyright 2012 by the American Society for Pharmacology and Experimental Therapeutics.
Abstract——The evident limitations of the amyloid
theory of the pathogenesis of Alzheimer’s disease are
increasingly putting alternatives in the spotlight. We
argue here that a number of independently developing
approaches to therapy—including specific and non-
specific anti-tumor necrosis factor (TNF) agents, apo-
lipoprotein E mimetics, leptin, intranasal insulin, the
glucagon-like peptide-1 mimetics and glycogen syn-
thase kinase-3 (GSK-3) antagonists—are all part of an
interlocking chain of events. All these approaches in-
form us that inflammation and thence cerebral insulin
resistance constitute the pathway on which to focus
for a successful clinical outcome in treating this dis-
ease. The key link in this chain presently absent is a
recognition by Alzheimer’s research community of the
long-neglected history of TNF induction of insulin re-
sistance. When this is incorporated into the bigger
picture, it becomes evident that the interventions we
discuss are not competing alternatives but equally
valid approaches to correcting different parts of the
same pathway to Alzheimer’s disease. These treat-
ments can be expected to be at least additive, and
conceivably synergistic, in effect. Thus the inflamma-
tion, insulin resistance, GSK-3, and mitochondrial dys-
function hypotheses are not opposing ideas but stages
of the same fundamental, overarching, pathway of Alz-
heimer’s disease pathogenesis. The insight this pro-
vides into progenitor cells, including those involved in
adult neurogenesis, is a key part of this approach. This
pathway also has therapeutic implications for other
circumstances in which brain TNF is pathologically
increased, such as stroke, traumatic brain injury, and
the infectious disease encephalopathies.
I. Introduction
Despite its increasingly high incidence, harmful ef-
fects on people and society, and the considerable funding
directed toward understanding its mechanism, differing
ideas on the driving force of Alzheimer’s disease (AD
1
)
remain unresolved. For decades, the bulk of the research
effort has been focused by the wealth of logic in the idea
that amyloid
(A
), the major neurohistological hall-
mark of this condition, triggers the onset of disease. This
approach was very encouraging in mouse studies
(Huang et al., 1999; Hung et al., 2008), but the negative
outcome of recent human trials, including when amyloid
was confirmed to have been reduced (Holmes et al.,
2008; Green et al., 2009; Salloway et al., 2009; Extance,
2010), has led to much reassessment and repositioning
that has led to lucid arguments for nonfailure of the
amyloid model itself (Karran et al., 2011; Sperling et al.,
2011). These negative human trials may have also led to
wider acceptance of AD research that has thrown the net
wider, taking into account the pathophysiology this dis-
ease shares with a range of conditions, both infectious
and noninfectious. This has allowed ideas such as the
cerebral insulin resistance model (de la Monte and
Wands, 2008) to gain warranted prominence (Correia et
al., 2011; McNay and Recknagel, 2011).
As discussed below, two therapeutic approaches al-
ready realized to be consistent with the model we are
proposing are intranasal insulin and parenteral gluca-
gon-like peptide-1 (GLP-1) mimetics. A major purpose of
this review is to summarize the large volume of pub-
lished evidence that, taking into account TNF and func-
tionally similar cytokines, dramatically reinforces the
likelihood that cerebral insulin resistance is indeed cen-
tral, albeit somewhat downstream, in the etiology of this
disease. The AD literature on leptin is also consistent
with this. Here we present the case that a number of
proposed treatments for AD are functionally linked, ei-
ther by their capacity to lower insulin resistance or to
deal with the consequences of this event (Fig. 1). These
treatments include leuprolide acetate, various ways to
reduce TNF levels (specific anti-TNF biological agents,
and nonspecific down-regulators of TNF production
(thalidomide, curcumin, and their derivatives; minocy-
cline; erythropoietin variants; and sex steroids), the
GLP-1 mimetics and dipeptidyl peptidase-4 (DPP-4) in-
hibitors, leptin, insulin itself, as well as glycogen syn-
thase kinase-3
(GSK-3
) inhibitors. All are under ac-
tive investigation by researchers presently coming from
different perspectives.
As we also discuss, not only are extensive links be-
tween TNF and AD now reported, but also between TNF
and gonadotropins as well as TNF and cell division,
insulin resistance, type 2 diabetes (T2DM), mitochon-
drial dysfunction, and the pathologic condition caused
by intracerebroventricular streptozotocin. These well
documented aspects of the repertoire of TNF activity,
which we suggest should become common currency in
AD research, are expanded upon in this review.
II. Gonadotropins, Sex Steroids, Tumor Necrosis
Factor, and Alzheimer’s Disease
Considerable evidence exists that elevated levels of
the gonadotropins luteinizing hormone (LH) and follicle-
stimulating hormone (FSH) are associated with neuro-
degenerative disease. For examples, total brain levels of
A
, a traditional histological marker for AD, are in-
creased by high LH levels [such as after ovariectomy
(Frye et al., 2007)], and decreased by the gonadotropin
superagonist leuprolide acetate (Bowen and Atwood,
2004; Casadesus et al., 2006; Berry et al., 2008). Cogni-
1
Abbreviations: A
, amyloid
;A
PP, amyloid
precursor protein;
ACT,
1-antichymotrypsin; AD, Alzheimer’s disease; Akt, protein ki-
nase B; apoE, apolipoprotein E; BBB, blood-brain barrier; CD14, cluster
determinant 14; CNS, central nervous system; CRP, C-reactive protein;
CSF, cerebrospinal fluid; DPP-4, dipeptidyl peptidase-4; EPO, erythro-
poietin; FSH, follicle-stimulating hormone; GLP-1, glucagon-like pep-
tide-1; GSK, glycogen synthase kinase; IFN, interferon; IL, interleukin;
LH, luteinizing hormone; NP12, 4-benzyl-2-methyl-1,2,4-thiadiazoli-
dine-3,5-dione; SB216763, 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-
3-yl)-1H-pyrrole-2,5-dione; SZT, streptozotocin; T2DM, type 2 diabetes;
TBI, traumatic brain injury; TNF, tumor necrosis factor.
BCLARK ET AL.
tive function follows the same pattern, with low LH
levels improving memory and cognition and high levels
making them worse, provided LH receptors are present
(Casadesus et al., 2006, 2007; Bryan et al., 2010; Ziegler
and Thornton, 2010; McConnell et al., 2012). We have
discussed previously the developing field of physiological
sex hormone replacement therapy for AD treatment
(Clark and Atwood, 2011). This is rationalized, at least
in part, by the capacity of both estradiol and progester-
one to reduce TNF expression in astrocytes (Kipp et al.,
2007). These hormones have been reported to protect
against AD (Honjo et al., 1989; Asthana et al., 2001).
Gonadotropins can regulate production of TNF, which
was shown to alter cell cycle dynamics by the group that
first described it (Darzynkiewicz et al., 1984). In brief,
FSH has been reported to induce TNF in vitro (Iqbal et
al., 2006), and high LH and FSH levels have allowed the
rationalization, through their association with high TNF
and IL-1
, of the onset or exacerbation of rheumatoid
arthritis in women at menopause (Kåss et al., 2010). As
we have noted (Clark and Atwood, 2011), the antigo-
nadotropic actions of leuprolide render it an anti-mitotic
and anti-inflammatory agent when used to treat endo-
metriosis. In this context, leuprolide has been reported
to reduce a number of inflammatory cytokines [e.g.,
IL-1
(Meresman et al., 2003), IL-6 (Ferreira et al.,
2010; Ficicioglu et al., 2010), and monocyte chemotactic
protein-1 (Khan et al., 2010)], all of which are induced by
TNF (Shalaby et al., 1989; Charles et al., 1999; Mueller
et al., 2010) and reduced by anti-TNF treatment (Bren-
nan et al., 1989; Redl et al., 1996; Charles et al., 1999).
Insulin resistance, commonly a TNF-induced state, and
now regarded to be central to AD (see section V.B), is
routinely seen in late pregnancy (Ryan et al., 1985). Late
pregnancy is also a time of physiological low-grade in-
flammation (de Castro et al., 2011) that is plausibly
regulated by the interactions of gonadotropins and TNF.
Not enough is yet known about the integration of
these reproductive hormones into broader physiology
and disease. Nevertheless, they already give an encour-
aging lead into how TNF might become excessive very
early in AD (Clark and Atwood, 2011). As was TNF for
years, in most minds these widely published hormones
are still in a nomenclature straightjacket arising from
their first description. This generates popular assump-
tions and limits enquiry into their relevance across
wider biology. The potential for their involvement is
there, because LH receptors, for example, are present on
an astonishing array of cell types, ranging from thymo-
cytes and peripheral lymphocytes (Rao et al., 2003) and
macrophages (Sonoda et al., 2005) through endothelial
cells (Tsampalas et al., 2010) to neurons and various
microglial cells (Rao et al., 2003), as well as where one
would expect them to be from the gonadotropin function
of LH.
III. Tumor Necrosis Factor and
Alzheimer’s Disease
The literature often gives the impression that TNF is
the only inflammatory cytokine, and most of this review,
for the sake of brevity, is no exception. TNF is at present
widely regarded, mainly from experience in the field of
rheumatology (Brennan et al., 1989; Charles et al.,
1999), as the master cytokine that starts the inflamma-
tory cascade. Nevertheless, mediators such as the inter-
FIG. 1. The overarching inflammatory pathway to Alzheimer’s disease that becomes evident once it is appreciated that TNF induces insulin
resistance (red arrow). Treatment concepts now in development (in blue) all address this pathway.
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD C
leukin-1s (of which IL-1
is the form released into ex-
tracellular fluids) are also inflammatory and may well
develop their own literature parallel to that described
here for TNF. IL-1 is the most advanced in this regard
(Griffin et al., 1989; Kitazawa et al., 2011). A mutual
dependence of these two cytokines is evident in the
brain, with reports of anti-TNF agents limiting the re-
lease of IL-1 (Terrando et al., 2010) and TNF levels
being reduced when IL-1 signaling is blocked (Kitazawa
et al., 2011). A number of higher numbered interleukins,
such as IL-12, IL-17, and IL-22, have become function-
ally linked with TNF but are, so far, little studied in the
brain. It also warrants noting, for clarity, that the term
TNF (Carswell et al., 1975) is identical to TNF-
, the
commonly seen suffix being a now-meaningless relic
from when lymphotoxin was, for a limited period some
years ago, referred to as TNF-
.
Although some still regard inflammation in AD as
solely a secondary downstream consequence of A
gen-
eration (as reviewed by Zotova et al., 2010), evidence
continues to accumulate (for review, see Clark et al.,
2010) for excess cerebral TNF, and therefore the cascade
of cytokines it initiates, to be viewed as an essential
preillness step in its pathogenesis. Some time ago,
higher cerebrospinal fluid (CSF) levels of TNF from 56
subjects with mild cognitive impairment, but not 25
age-matched controls subjects, were reported to predict
which patients would develop frank AD (Tarkowski et
al., 2003). Other researchers, taking advantage of the
increased sensitivity of assaying for soluble TNF recep-
tors rather than TNF itself, found that their levels in
serum and CSF predicted, over a 4- to 6-year period,
conversion to clinical AD (Buchhave et al., 2010). Some
groups studied plasma levels of C-reactive protein (CRP)
and
1-antichymotrypsin (ACT) (Engelhart et al., 2004)
(two acute-phase proteins up-regulated by TNF or IL-1)
or of CRP alone (Laurin et al., 2009; Schuitemaker et al.,
2009) and found that these markers of inflammation
were present in serum and CSF before any indications of
increased A
.
These studies on the primary role of inflammatory
mediators have now been extended by a report that
plasma levels of another acute-phase protein, clusterin
(apolipoprotein J), are intimately associated with onset,
progression, and severity of AD (Thambisetty et al.,
2010). A novel proteomic neuroimaging paradigm was
employed. Unfortunately, the authors offered only an
A
-based rationale for these findings and did not note
that clusterin is an acute-phase protein (Hardardo´ttir et
al., 1994). Therefore, it is a marker, as surely as are CRP
and ACT, of increased pro-inflammatory cytokines such
as TNF and IL-1. One of their more telling findings was
that clusterin is raised 10 years earlier in the course of
the disease than is fibrillar A
deposition. Moreover, a
metastudy has determined that CLU, the clusterin gene,
is the second highest of a list of the 15 top-rated genes
linked to AD on the Alzgene web-based collection (Ol-
giati et al., 2011). Taken together, these arguments are
consistent with the key nature of inflammation in AD
onset. The induction of insulin resistance, accepted for
years by the wider literature to be mediated by TNF (see
section V.D), is, like TNF, also a very early event in AD,
even preceding the onset of minimal cognitive impair-
ment (Baker et al., 2011). Indeed, insulin resistance has
been reported to be associated with reduced executive
function in older people lacking any evidence of T2DM or
dementia. The concept of age-related cytokine increase
driving this insulin resistance was one of the possibili-
ties the authors considered (Abbatecola et al., 2004).
IV. Tumor Necrosis Factor, Amyloid
, and
With more than 30 years of dominance of the AD
literature by amyloid
precursor protein (A
PP) and its
cleavage product, A
, it is not surprising that most
pathologic conditions associated wth AD, including in-
sulin resistance, have been seen as consequences of A
deposition (Balaraman et al., 2006; Perry et al., 2007;
Townsend et al., 2007; Li et al., 2010; Lei et al., 2011).
However, current clinical trial outcomes are consistent
with A
being little more than a marker for more rele-
vant events (Holmes et al., 2008; Green et al., 2009;
Salloway et al., 2009; Extance, 2010).
Unfortunately, the direction of AD research has a
momentum that has not yet, on the whole, taken into
account that A
is a highly TNF-dependent protein. For
instance, A
PP, the centerpiece of the amyloid theory of
AD pathogenesis, is induced by inflammatory cytokines,
including TNF and IL-1. This is a widespread phenom-
enon. In addition to the fact that the promotor region of
the A
PP gene is controlled by these cytokines (Ge and
Lahiri, 2002), its induction by these inflammatory cyto-
kines is reported in endothelial cells (Goldgaber et al.,
1989), skeletal muscle (Schmidt et al., 2008), and 3T3 L1
adipocytes (Sommer et al., 2009) as well as brain (Brugg
et al., 1995; Buxbaum et al., 1998). Its presence in brain
is not confined to noninfectious diseases, being described
in AIDS dementia (Stanley et al., 1994) and cerebral
malaria (Medana et al., 2002). Regarding A
PP cleav-
age, in 2004 it was reported that IFN-
, IL-1
, and TNF
specifically stimulate
-secretase activity, with an ac-
companying increased production of A
(Liao et al.,
2004). IFN-
and TNF were subsequently shown to en-
hance A
production from A
PP-expressing astrocytes
and cortical neurons, and the numbers of astrocytes
expressing IFN-
were shown to have increased
(Yamamoto et al., 2007). This group also showed that 1)
TNF directly stimulates
-site A
PP-cleaving enzyme
(or
-secretase) expression and thus enhances
-site pro-
cessing of A
PP in astrocytes and 2) that TNFR1 deple-
tion reduced
-site A
PP-cleaving enzyme activity, as
well as learning and memory deficits (Yamamoto et al.,
2007). Taken together, these data imply that anti-TNF
agents should be effective A
PP cleavage inhibitors.
DCLARK ET AL.
Data from a mouse AD model after long-term inhibition
of TNF are functionally consistent with this (McAlpine
et al., 2009).
In contrast, it now seems reasonably appreciated that
inflammatory cytokines such as TNF mediate events
downstream of A
. Nearly a decade ago TNF was re-
ported to alter synaptic transmission in hippocampal
slices (Tancredi et al., 1992). Several years later (Wang
et al., 2005b; Rowan et al., 2007), it was shown that this
earlier observation explained the ability of A
, through
TNF, to do the same. Other researchers expanded the
roles of TNF in this context (Pickering et al., 2005;
Stellwagen et al., 2005). The capacity of A
to act as a
ligand for CD14 and toll-like receptor-2 (Fassbender et
al., 2004; Jana et al., 2008; Tu¨kel et al., 2009) indicates
that these findings with A
(Wang et al., 2005b; Rowan
et al., 2007) are consistent with basic immunology, be-
cause occupancy of CD14 and toll-like receptors is how
the usual bacterial- and protozoal-origin inducers of
TNF operate (Beutler and Poltorak, 2001). Key support
for this concept has been provided by the recent demon-
stration that the release of proinflammatory cytokines
from astrocytes is necessary for either A
to be neuro-
toxic or
phosphorylation to be initiated (Garwood et al.,
2011). Much research on A
’s causing the pathological
features of AD (Hardy and Selkoe, 2002; Games et al.,
2006; Marwarha et al., 2010) appears yet to take this
body of literature into consideration. In short, it should
by now be clear that many experimental observations
attributed to added A
might well actually be caused by
the inflammatory cytokines, including TNF. This addi-
tional TNF may have added to the total load (Fig. 1), but
if it were a significant contributor to the clinical out-
come, we would expect the human trials of antiamy-
loid therapies discussed earlier to have given positive
results.
Likewise, hyperphosphorylated
, another histological
sign of high cytokine activity (Medana et al., 2005, 2007;
Gorlovoy et al., 2009) can be regarded as one of the
obvious markers of GSK-3 (see section VI) activation
subsequent to cytokine-induced insulin resistance,
rather than as an essential early step in the pathogen-
esis of the disease. Hyperphosphorylated
has been
advocated for many years as a primary mechanism of
loss of cerebral function and cell loss (Goedert, 2004;
Go¨tz et al., 2012), and mice expressing mutant human
are reported to exhibit many of the features of AD
(Takeuchi et al., 2011). However, claims for a direct
harmful effect on neurons need to be reconciled with
evidence of a large reversible increase in hyperphospho-
rylated
, leaving function and structure able to return
unscathed once experimentally induced mammalian hi-
bernation is reversed (Ha¨rtig et al., 2007). Conceivably
this particular phosphorylation, although spectacular
down a microscope, may well be the least important of
the myriad of other, unseen, phosphorylations caused by
GSK-3 activation. In summary, we argue that the now-
known complexities of the current literature on the cy-
tokines, insulin resistance, and GSK-3 reduce the need
for incorporating the traditional AD hallmark proteins,
however histologically intriguing, into our model to un-
derstand the origins and mechanism of this disease.
V. Insulin
A. Insulin in Basic Biology and the Brain
Over the decades, the literature of soluble mediators
referred to as cytokines or hormones (conceivably inter-
changeable terms) have taken unexpected turns, typi-
cally through the discovery of functions quite unrelated
to those for which they first came to notice. For instance,
given its original function of tumor killing (Carswell et
al., 1975), it was difficult to get acceptance of any role for
TNF in innate immunity or disease pathogenesis (Clark
et al., 1981). Insulin receptors had already been noted to
be widely distributed in the central nervous system of
the rat (Havrankova et al., 1978). This unexpected in-
formation was soon followed by reports of conventional
insulin and insulin receptors in flies, earthworms, and
bacteria (LeRoith et al., 1981a,b). The central relevance
of insulin to brain physiology was a ground-breaking
revelation (for review, see Adamo et al., 1989). Clearly,
these and similar developments indicate that insulin
has a central importance in biological signaling.
As has recently been reviewed (Correia et al., 2011),
once bound to the extracellular domain of a specific
tyrosine kinase receptor, insulin causes autophosphory-
lation of its intracellular component, triggering a chain
of tyrosine kinase activity. As these authors discuss,
subsequent phosphorylation activates cascades that in-
clude phosphoinositide 3-kinase/protein kinase B (Akt).
This pathway (one of those inhibited by excess TNF) in
turn phosphorylates and thereby inhibits (Cross et al.,
1995) the
and
cytosolic forms of GSK-, which is a
serine/threonine protein kinase with profound impor-
tance in many biological systems, including neurotrans-
mission at the synaptic level (Smillie and Cousin, 2011).
Other pathways, such as c-Jun N-terminal kinase/mito-
gen activated protein kinase, omitted here for brevity,
are also involved. The phosphoinositide 3-kinase/Akt
cascade also triggers translocation of the insulin-sensi-
tive glucose transporter 4 to the cell surface, enhancing
glucose uptake (Bryant et al., 2002). This is clearly cen-
tral to mitochondrial function and therefore ATP pro-
duction in AD. Nevertheless, there is ample evidence
that insulin has the capacity to control memory indepen-
dently of its effects on glucose uptake (Craft et al., 1996,
1999).
B. Insulin Resistance and Alzheimer’s Disease
Insulin resistance can be regarded as 1) a decreased
response in the presence of normal insulin levels or as
2) the need for more insulin for a normal response (i.e.,
the uptake of glucose, amino acids and fatty acid by
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD E
peripheral tissues). As far as we are aware, the first
suggestions that insulin function was suppressed in AD
were made more than a decade ago in the context of
energy metabolism (Hoyer et al., 1994, 2000). Given the
many essential roles of insulin documented in neuro-
physiology, the consequences of alterations in cerebral
insulin resistance are inevitably widespread. This re-
view focuses on more recent studies on the control of
insulin resistance, and its implications, in a number of
diseases, including AD, where it is attracting much cur-
rent attention from prominent groups coming from quite
different directions (Correia et al., 2011; Liu et al., 2011;
McNay and Recknagel, 2011).
C. Tumor Necrosis Factor and Insulin Resistance
The basic literature that spans both immunity and
metabolism accepts that pro-inflammatory cytokines
cause insulin resistance, and anti-inflammatory cyto-
kines promote insulin sensitivity (Chawla et al., 2011).
Nevertheless, the implications of this link have not yet
reached the AD literature, even though it dwells consid-
erably on both inflammation and insulin resistance, two
of the most recognized processes associated with the
disease. Likewise, an awareness of this connection adds
an important additional dimension to the literature on
the pathogenesis of fetal alcohol syndrome disorder. As
with AD, research on this disorder contains two fields,
presently discrete, that it would be useful to conceptu-
ally merge: 1) ethanol induction of TNF in vivo (Qin et
al., 2008) and in vitro (Boyadjieva and Sarkar, 2010),
thus harming neurons (Boyadjieva and Sarkar, 2010;
Hicks and Miller, 2011), and 2) ethanol-induced insulin
resistance (de la Monte et al., 2005, 2011; de la Monte
and Wands, 2010). As Fig. 1 illustrates, this would open
up a wider awareness of treatment possibilities for both
conditions.
The causative link between TNF and insulin resis-
tance has a long history. In 1967, insulin resistance was
observed in a patient with tularemia (Shambaugh and
Beisel, 1967), a condition caused by Francisella tularen-
sis, a Gram-negative tick-borne coccobacillus that much
later proved to be a strong inducer of TNF and its down-
stream cytokines (Golovliov et al., 1996). By 1974, insu-
lin resistance had been reported in septic and trauma-
tized patients (Gump et al., 1974) and was generated in
vivo by injecting bacterial endotoxin (Chaudry et al.,
1974). This same agent was shown, in 1975, to be the
prototype inducer of TNF (Carswell et al., 1975). That
year also saw burn injury, recognized decades later to
increase TNF to a functionally important degree (Gi-
roir et al., 1994; Boehm et al., 2010), being reported to
cause insulin resistance in rats (Frayn, 1975). The
endotoxin concept of insulin resistance in sepsis was
extended to skeletal (Raymond, 1984) and cardiac
(Raymond et al., 1988) muscle, although with no men-
tion of TNF or other cytokines as intermediaries. Im-
portantly, these authors proposed that a post-insulin
receptor site was responsible.
In the early 1980s, after acceptance that harmful ef-
fects of bacterial endotoxin and other functionally simi-
lar agents were caused through host-origin soluble pro-
teins (eventually termed cytokines) that were elicited
from patients’ cells, these proteins were linked with
induction of insulin resistance. Initially, a semipurified
protein that bacterial endotoxin released from macro-
phages was demonstrated to cause insulin resistance in
adipocytes (Pekala et al., 1983). This undefined protein,
in a class then termed monokines, did not affect insulin
binding or stimulation of glucose uptake. Two years
later it was sequenced (Beutler et al., 1985) and found,
unexpectedly, to be identical to a previously sequenced
molecule, TNF (Aggarwal et al., 1985). In 1989, a group
who explored this area by infecting rats with Esche-
richia coli (Lang and Dobrescu, 1989) also predicted a
defect in insulin signaling distal to receptor binding but
again did not mention the link, by then well established,
between endotoxin from this bacterium and TNF induc-
tion (Carswell et al., 1975).
A milestone article, also in 1989 (Fraker et al., 1989),
demonstrated that injecting insulin and recombinant
TNF concurrently into rats prevented or significantly
reduced a range of metabolic and pathological changes
seen in acute TNF toxicity. Various interpretations were
proposed, but none proved satisfactory. With hindsight,
it seems plausible that sufficient insulin had been in-
jected to overcome much of the insulin resistance caused
by the coadministered TNF. If so, the breadth of meta-
bolic and histological observation in this text gives an
intriguing insight into the wide influence of signal mod-
ification driven by cytokine-induced insulin resistance,
much still unexplored. Weiner et al. (1991) found insulin
and recombinant TNF to produce potent and opposing
physiological signals in adipocytes. This paved the way
for groups interested in various non-AD diseases, includ-
ing examples caused by infectious agents known to in-
duce TNF, to demonstrate that this cytokine was a po-
tent cause of insulin resistance (Lang et al., 1992;
McCall et al., 1992; Davis et al., 1993; Feinstein et al.,
1993; Hotamisligil et al., 1993, 1996; Li et al., 2007; Qin
et al., 2007; Lorenzo et al., 2008). Feinstein et al. (1993)
seem to have been the first to argue that TNF exerts a
major part of its antiinsulin effect by interrupting insu-
lin-stimulated tyrosine phosphorylation, a key observa-
tion that was confirmed in cells from knockout mice by
Nieto-Vazquez et al. (2007). Much of this work was done
in the context of T2DM. Newer reports from within the
T2DM and AD interface discuss, as one entity, the cere-
bral and peripheral TNF and insulin relationship (Liu et
al., 2011; Bomfim et al., 2012).
Inhibiting TNF can prevent or reverse insulin resis-
tance. Uysal et al. (1997) showed that insulin resistance
did not develop in obese mice lacking TNF function.
Seven years later a series of patient studies began to
FCLARK ET AL.
appear in which commercial anti-TNF biological agents
reduced insulin resistance in T2DM (Yazdani-Biuki et
al., 2004), rheumatoid arthritis [in some (Kiortsis et al.,
2005; Gonzalez-Gay et al., 2006, 2010, 2012) but not all
(Ferraz-Amaro et al., 2011) reports], and ankylosing
spondylitis (Kiortsis et al., 2005). In addition, inflix-
imab, one of these commercial anti-TNF biological
agents, was tested in obese diabetic mice, and it im-
proved insulin signal transduction in muscle, liver, and
hypothalamus. In doing so, it completely restored the
activity of insulin-induced insulin receptor, insulin re-
ceptor substrate-1, and receptor substrate-2 tyrosine
phosphorylation and Akt and forkhead box protein O1
serine phosphorylation (Arau´jo et al., 2007). In the same
vein, others have reported that insulin signaling in en-
dothelial progenitor cells, measured by the phosphory-
lated to total Akt ratio, was reduced by 56% on exposure
to TNF (Desouza et al., 2011). Even more recently, inf-
liximab has been employed to demonstrate that the in
vitro insulin resistance induced by A
is inhibited by
neutralizing TNF (Bomfim et al., 2012).
Because treatment of AD with large anti-TNF biolog-
ical agents is focused on delivering them into the CSF
(section XI.B), the most AD-relevant in vivo demonstra-
tion to date of altering insulin resistance in this way has
been done by Arruda et al. (2011), who showed that
intracerebroventricular infliximab improved insulin sig-
nal transduction through insulin receptor substrate 1.
This was accompanied by a whole-body reduction in
insulin resistance.
D. Functional Links of Glucagon-Like Peptide-1 to
Insulin Resistance
Certain gut peptides, the most prominent being
GLP-1, have emerged as central to understanding
both brain function (During et al., 2003) and insulin
physiology. As has been reviewed in the context of
neurodegenerative disease (Greig et al., 2004b;
Ho¨lscher and Li, 2010; Holst et al., 2011), GLP-1, an
endogenous insulinotropic peptide, reduces insulin re-
sistance (Cabou et al., 2008; Knauf et al., 2008). It was
originally believed to arise only from L cells in the
distal ileum and colon but is now intensively studied
as a peptide of brain origin, with key brain functions.
GLP-1 and its mimetics provide a set of signals that
are the reverse of those (e.g., regarding Akt and c-Jun
N-terminal kinase) generated by excess TNF (Li et al.,
2005; Ferdaoussi et al., 2008; Natalicchio et al., 2010).
Consequently, GLP-1 mimetics have the capacity to
reduce insulin resistance in ways shared by exogenous
anti-TNF agents. Because GLP-1 is rapidly degraded
in vivo, degradation-resistant analogs have been de-
veloped and are in therapeutic use for T2DM. As dis-
cussed in section XI.F, these agents also show promise
in AD models.
VI. Tumor Necrosis Factor and Glycogen
Synthase Kinase-3
In the literature, GSK-3
predominates over the very
similar GSK-3
form and, for simplicity, is referred to
exclusively in this text. As with a number of other mol-
ecules with very high profiles, the fame of GSK-3
not
rests not on its first description [arising from the capac-
ity to phosphorylate and thence inactivate glycogen syn-
thase (Embi et al., 1980)] but on the gradual realization
of its very great number of substrates, more than 50 of
them documented by 2003 (Doble and Woodgett, 2003).
Fifteen years earlier, TNF had caused a similar stir
when, as a cytokine still traditionally linked in most
minds only to tumor necrosis, it was noted to possess a
remarkably high number of physiological as well as
pathological functions (Nathan, 1989). We have previ-
ously discussed this in the CNS context (Clark et al.,
2010). In hindsight, this information contained the po-
tential to have opened minds to the possibility of a
functional link between these two strikingly pleiotropic
molecules associated with normal physiology, innate im-
munity, and inflammation.
As with TNF, many groups are interested in the roles
of GSK-3
in brain function. Over the years, those ar-
guing for a primary role for hyperphosphorylated
in
AD pathogenesis have, as expected, focused on GSK-3
as the phosphorylating kinase that generates this form
of
(Mandelkow et al., 1992; Lovestone et al., 1994;
Lovestone and Reynolds, 1997). Other have examined
its effects on the brain itself and produced much com-
pelling data suggesting that GSK-3
, in its inhibited
state, is essential for normal brain function, and its
activated state leads to the array of functional loss seen
in AD (Jope and Johnson, 2004; Balaraman et al., 2006;
Engel et al., 2006; Hooper et al., 2007; Kimura et al.,
2008; Salcedo-Tello et al., 2011; Smillie and Cousin,
2011). As noted in section VI, the argument that when-
ever GSK-3
activation is high, the hyperphosphory-
lated
generated initiates disease (Goedert, 2004) has
yet to explain the reported harmlessness of this protein
in induced mammalian hibernation (Ha¨rtig et al., 2007).
Certainly, the position of hyperphosphorylated
in Fig.
1 is reinforced by the evidence that it is reduced when
the tap is turned off at the top of the cascade by LH
ablation (Lin et al., 2010), anti-TNF (Shi et al., 2011),
IL-1 signaling blockade (Kitazawa et al., 2011), minocy-
cline (Garwood et al., 2010), sex steroids (Carroll et al.,
2007), or additional insulin (Hong and Lee, 1997).
TNF and GSK-3
have proved to be functionally
linked, both in physiology and disease pathogenesis.
Insulin resistance, a phenomenon readily caused by
TNF (see section V.C), has long been known to influence
GSK-3
activity (for review, see Jope 2004; Jope and
Johnson, 2004). As noted above (Fraker et al., 1989),
insulin also reduces the harmful effects of excess TNF
production, as do sex steroids (e.g., (Jiang et al., 2009).
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD G
In addition, the reduction of endotoxin and peptidogly-
can-induced pathologic conditions in rats by insulin, in-
dependent of blood glucose changes, has been demon-
strated to involve GSK-3 inhibition (Dugo et al., 2006).
Others reported that the insulin resistance and associ-
ated increase in GSK-3
activity in brains of a mouse
model of T2DM, as well as what the authors noted were
learning difficulties parallel to those seen in AD, were
corrected by administering insulin (Jolivalt et al., 2008).
Moreover, the specific GSK-3
inhibitor 3-(2,4-dichloro-
phenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione
(SB216763) altered the cytokines released from endotoxin-
stimulated human monocytes to a strongly anti-inflamma-
tory profile (Martin et al., 2005). Nevertheless, the separa-
tion of the two in the minds of most researchers has led to
the existence of both inflammatory and GSK-3 models of
AD. Rarely are they discussed together, and then only in
terms of post-A
secondary inflammation, not as two sub-
units of an essential component of disease initiation
(Hooper et al., 2008).
VII. Tumor Necrosis Factor and
Mitochondrial Dysfunction
As we have reviewed previously (Clark et al., 2010),
the concept of dysfunctional mitochondria, and thus poor
oxygen utilization and energy production, is important
in understanding disease pathogenesis. This seems to
have been first suggested, on the basis of organelle elec-
tron microscopy, in malaria (Maegraith, 1954). Nearly
20 years later, it appeared in the sepsis literature (Mela
et al., 1972). A further 2 decades later, TNF, by then well
established as a key mediator in infectious disease, was
first demonstrated to suppress mitochondrial respira-
tion (Stadler et al., 1992). By the end of that decade,
extensive functional studies in this context were begun
(Fink, 1997, 2000, 2001), and the general reasoning was
extended to HIV dementia (Kruman et al., 1999) and
influenza encephalopathy (Yokota, 2003). More recent
research on TNF’s ability to induce mitochondrial dys-
function (Chen et al., 2010) again noted that direct treat-
ment with TNF led to reduced intracellular ATP and
more generation of reactive oxygen species. More signif-
icantly, the anti-TNF biological agent etanercept has
been shown to ameliorate cardiac mitochondrial dys-
function in vivo (Moe et al., 2004). Systemic mitochon-
drial dysfunction, as part of cytokine-induced inflamma-
tion, is still very topical in the pathophysiology of sepsis
(Garrabou et al., 2012).
Mitochondrial dysfunction has an early onset in AD
(Hauptmann et al., 2009) and is widely regarded as
important in its pathogenesis (Castellani et al., 2002).
Although the first group to propose mitochondrial func-
tional defects as a mechanism for AD was primarily
interested in oxidative stress as a mechanism (Blass and
Gibson, 1991), it did not take long for researchers of A
,
by then dominating AD disease pathogenesis, to incor-
porate the mechanism of mitochondrial dysfunction into
their reasoning (Kaneko et al., 1995). This view persists
to the present day (Borger et al., 2011) and has remained
largely unquestioned despite the implications of the
prevalence of TNF in AD brains (section III) and the
widely published capacity of this cytokine to induce both
A
PP and A
(section IV) and to directly cause mito-
chondrial dysfunction (see above). Young-Collier et al.
(2012) found that the reduced expression of mRNA of
genes responsible for mitochondrial function in human
AD neurons could not be duplicated with 3 days of cul-
ture with A
1–42
(instead, expression rose); this might
now encourage investigation into other possible mecha-
nisms for the finding.
Does mitochondrial dysfunction precede or follow in-
sulin resistance? Both viewpoints appear in the litera-
ture, with discussion of excessive fat intake being com-
mon to several articles arguing that mitochondrial
dysfunction occurs first (Anderson et al., 2009; Rector et
al., 2010). In contrast, a study designed to investigate
the mitochondrial dysfunction during fasting concluded
it was a consequence rather than a cause of insulin
resistance (Hoeks et al., 2010). Indeed, when associated
with cell death or apoptosis, mitochondrial dysfunction
is reported to occur after GSK-3
activation (Petit-Paitel
et al., 2009; Wang et al., 2011), placing it after insulin
resistance (Fig. 1). Its position here is consistent with
experiments in which the mitochondrial dysfunction in-
duced by SZT, an agent that mimics T2DM or AD (sec-
tion IX.A), was corrected by administering insulin
(Chowdhury et al., 2010). Clearly, this implies that im-
proved cerebral mitochondrial function is a plausible
consequence of treating AD with intranasal insulin (sec-
tion XI.E; Fig. 1).
VIII. Tumor Necrosis Factor and
Progenitor Cells
A. Progenitor Cells, Tumor Necrosis Factor, and
Insulin Resistance
Adult neurogenesis, which is low or absent in the
shrunken brains of patients with AD, is essential for
normal memory formation (Clark et al., 2010). Such
progenitor activity, requiring activin A (Abdipranoto-
Cowley et al., 2009), is part of a normal organism-wide
pattern in which such cells are controlled by a sex hor-
mone-TNF pathway to maintain cellular homeostasis.
The dynamic regulation of neurogenesis by hypothalamic-
pituitary-gonadal axis hormones, including activins, go-
nadotropins, and sex hormones, particularly progesto-
gens, is well established (Vadakkadath Meethal and
Atwood, 2005). However, the sequence of events through
which sex hormones seem to modulate TNF during con-
trol of progenitor cells is not yet known. Despite this
incomplete picture, the literature to date is consistent
with the idea that increased TNF induces insulin resis-
tance, and thus GSK-3
activation (Verhees et al.,
HCLARK ET AL.
2011), and constitutes the major pathway in progenitor
cell homeostasis.
T2DM is a good example of a disease combining
chronic systemic inflammation, insulin resistance, and
widespread defects in progenitor cell function. It is tell-
ing that it should both predispose to AD (Arvanitakis et
al., 2004) and share cerebral insulin resistance with this
condition (Liu et al., 2011). The details of the signaling
deficits are also the same (Liu et al., 2011). In neurogen-
esis (and, as far as has been examined, a general rule in
other progenitors), physiological levels of TNF and its
downstream cytokines enhance proliferation, whereas
supraphysiological levels inhibit proliferation (Ber-
nardino et al., 2008). This phenomenon has been dem-
onstrated in thymocytes (Ranges et al., 1988; Herna´n-
dez-Caselles and Stutman, 1993), hepatocytes (Bour et
al., 1996; Diehl and Rai, 1996), hematopoiesis (Clark
and Chaudhri, 1988; Rebel et al., 1999) and, of plausible
relevance to data from AD brains (Sheng et al., 2012),
impaired mitochondria biogenesis (Valerio et al., 2006).
The relevance of TNF-induced insulin resistance to the
pathogenesis of the widespread degenerative change
that characterizes chronic inflammatory diseases can be
gleaned from the literature on endothelial cell progeni-
tors (Cubbon et al., 2009; Abbas et al., 2011; Desouza et
al., 2011) and thus nephropathy; muscle progenitors
(Pajak et al., 2008) and thus cachexia; fibroblast progen-
itors (Frankel et al., 2006; Goren et al., 2006; Siqueira et
al., 2010) and thus poor wound healing; cartilage pro-
genitors (Alblowi et al., 2009; Kayal et al., 2009) and
thus poor fracture repair; and erythroblasts (Tsinka-
lovsky et al., 2007) and thus the anemia of chronic dis-
ease. Whether the recorded protective effect of sex hor-
mones in some of these circumstances is an independent
property parallel to their ability to reduce production of
TNF (He et al., 2004; Kipp et al., 2007) is a yet to be
tested. We note, however, that estrogen has been re-
ported to promote cutaneous wound healing (Campbell
2010) by means other than its anti-inflammatory mech-
anism. Nevertheless, the poor wound healing in rheu-
matoid arthritis, a condition exhibiting insulin resis-
tance reversible by anti-TNF agents (Kiortsis et al.,
2005; Gonzalez-Gay et al., 2006), has been reported to be
countered by anti-TNF treatment (Shanmugam et al.,
2011). Although on a small scale, this study is intrigu-
ing, because conventional wisdom, predicated on the
idea that such anti-TNF treatment has the potential to
suppress immunity against certain pathogens that could
infect wounds, would have us expect the opposite out-
come. As might be expected, elevated levels of sex ste-
roids, such as during pregnancy or after hormonal re-
placement therapy, and known to suppress TNF, leads
to diminished disease activity in rheumatoid arthritis
(Ostensen et al., 1983; Kanik and Wilder, 2000; Islander
et al., 2011). In contrast, the disease is often aggravated
after parturition (Ostensen et al., 1983).
B. Clock Genes, Controlled by Tumor Necrosis Factor,
Govern Progenitor Activity
The control of progenitor cells homeostasis by TNF is
but a small part of the broader control that tissue clocks
exert in all tissues through circadian, or clock, genes.
Indeed, cell division in general is under their control
(Matsuo et al., 2003). Likewise, the normal diurnal cy-
cles in food intake, sleep, insulin requirements, and
mitochondrial function, kept in their normal circadian
patterns when these genes remain under physiological
diurnal fluctuations of sex hormones, TNF and down-
stream cytokines (Kohsaka and Bass, 2007), run amok
in well documented ways during illness (Hart, 1988;
Bluthe et al., 1994; Dantzer and Kelley, 2007). For as
long as TNF and IL-1
are in pathological disease-in-
duced excess, clock genes undergo a longer-term sup-
pression (Cavadini et al., 2007). Hence mechanisms gov-
erned by clock genes, including cell cycling (Matsuo et
al., 2003), can be expected to undergo pathological
change. Several years ago we proposed that such sup-
pression can explain the pattern of pathology that char-
acterizes severe bacterial, protozoal, viral and post-
trauma disease (Clark et al., 2008).
Among the clock genes suppressed by excess TNF and
IL-1
are the period genes, Per1,Per2 and Per3 and the
central, interconnecting, response element clock gene,
rev-erb
(Cavadini et al., 2007). The existence of an
essentially parallel literature on reproductive hormones
and clock genes (Nakamura et al., 2008; Nakamura et
al., 2010; Karatsoreos et al., 2011) again demonstrate
the present minimal awareness of the functionally im-
portant adjacent positions of sex hormones and TNF in
the same regulatory pathway. Certain clock genes have
been demonstrated to undergo insulin-dependent regu-
lation (Tahara et al., 2011), and to control adult neuro-
genesis, including in the hippocampus (Moriya et al.,
2007; Borgs et al., 2009; Kimiwada et al., 2009), as well
as endothelial cell (Wang et al., 2008) and cartilage
(Mengatto et al., 2011) progenitors. Of particular rele-
vance here are the data from experiments published in
2004 (Kuriyama et al., 2004) in which the normal circa-
dian clock oscillation, present in all tissues, was exam-
ined in heart and liver of mice in which diabetes was
generated with SZT. Per2 was diurnally inhibited, but
this could be corrected by injecting insulin, i.e., by over-
coming insulin resistance. This is consistent with gluca-
gon-like peptide-1 (GLP-1) mimetics, clinically useful
against T2DM because of their ability to correct insulin
resistance (see next Section), promoting neurogenesis in
AD models (Hamilton et al., 2011; Holst et al., 2011).
Predictably (Jope and Johnson, 2004), the degree of ac-
tivation of GSK-3
proves to be what ultimately controls
the clock genes, and thus proliferation (Hirota et al.,
2008; Ko et al., 2010; Kozikowski et al., 2011). Again as
expected, phosphorylation of GSK-3
itself normally un-
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD I
dergoes robust circadian oscillation, and it readily phos-
phorylates Per 2 (Iitaka et al., 2005).
Taken together, the wider progenitor cell literature is
therefore consistent with inhibition of neurogenesis in
AD by excess brain TNF through a pathway that in-
volves inhibition of clock genes by insulin resistance,
thus damping down or switching off progenitor cells.
Evidence also incriminates chronic inflammation in re-
duced recruitment of new neurons into the hippocampal
networks that underlie memory consolidation (Belarbi
et al., 2012a). Inhibited neurogenesis and distorted cell
cycling (Yang and Herrup, 2007) in AD are but two
consequences of this widely applicable principle in dis-
ease pathogenesis. Logically speaking, AD is therefore
susceptible to treatment with any of the approaches
discussed herein that rectify insulin resistance. More-
over, the main stages of this pathway are recognized
general principles as much at home in physiology [e.g.,
in the metabolic shutdown of hibernation (Stieler et al.,
2011) and dauer, or suspended animation, forms of the
nematode Caenorhabditis elegans (Tissenbaum and Ru-
vkun, 1998; Forsythe et al., 2006)] as in other human
diseases beyond AD and T2DM, including stroke (Val-
erio et al., 2011) and depression (Li and Jope, 2010)
(Fig. 2). These last two conditions have an emerging
literature on therapy with anti-TNF biological agents
(Uguz et al., 2009; Tobinick, 2011).
IX. Streptozotocin
A. Streptozotocin Model for Diabetes and
Alzheimer’s Disease
SZT, originally isolated from Streptomyces achromo-
genes for use as an antibiotic (Vavra et al., 1959), was
later realized to be a potent diabetogenic agent (Junod et
al., 1967); since then, it has had an important role in
diabetes research. In 1983, awareness developed that
SZT, besides leading to pancreatic
-cell destruction,
also inhibits metabolic responsiveness to insulin rather
than its binding to its receptor (Hansen et al., 1983). A
decade later it was appreciated that intracerebroventric-
ular injection of SZT produces changes in glucose me-
tabolism that parallel those seen in AD (Plaschke and
Hoyer, 1993); subsequently, its ability to bring about a
wide range of the rest of the changes seen in AS began to
be uncovered. For example, rats receiving intracerebro-
ventricular injections of SZT, which does not alter sys-
temic glucose metabolism, develop insulin receptor de-
fects and thus insulin resistance (Hoyer et al., 2000).
They also exhibit brain atrophy, neurodegeneration, gli-
osis, and increased immunoreactivity for activated
GSK-3
and hyperphosphorylated
, as observed in AD
(Lester-Coll et al., 2006). This model is seen as increas-
ingly important because, in contrast to genetically gen-
erated mouse strains that dominate the experimental
literature, it closely resembles the most common human
condition, termed sporadic AD. There is now a consider-
able literature on the use of SZT to establish models of
insulin resistance (Blondel and Portha, 1989; Koopmans
et al., 2006; Cheng et al., 2010; Thackeray et al., 2011).
As reviewed previously (de la Monte and Wands, 2008),
these SZT-induced changes could be reduced or pre-
vented by early treatment with peroxisome proliferator-
activated receptor agonists in doses smaller than rou-
tinely used to treat diabetes type 2. TNF down-regulates
certain peroxisome proliferator-activated receptor re-
ceptors (Beier et al., 1997).
B. Streptozotocin Induces Tumor Necrosis Factor
The capacity for SZT to induce TNF has been docu-
mented since the mid-1990s (Sagara et al., 1994; Herold
et al., 1996). Cai et al. (2011) reported that it increases
TNF and IL-1
in rat hippocampus. This activity of SZT
to induce TNF has been exploited to help understand the
consequent pathologic features of diabetes (Sagara et
al., 1994; Holstad and Sandler, 2001; Zauli et al., 2010;
Devaraj et al., 2011). Specific examples include diabetic
cardiomyopathy (Westermann et al., 2007) and diabetic
nephropathy (Mensah-Brown et al., 2005; Navarro et al.,
2005). In the latter, successful experimental treatments
include combined insulin (overcoming insulin resis-
tance) and curcumin [reducing the inflammatory re-
sponse (Sharma et al., 2007)] as well as curcumin alone
(Soetikno et al., 2011). A commercial anti-TNF biological
agent has also been used, to good effect, for this purpose
(Yamakawa et al., 2011). As far as we are aware, Isik et
al. (2009) are the only researchers to use an anti-inflam-
matory approach (curcumin) to rationalize post-SZT in-
sulin resistance. As might be expected from the inter-
play between reproductive hormones and TNF, sex
steroids are well recognized to reverse post-SZT insulin
resistance and protect from insulin resistance in rats
exhibiting SZT-induced diabetes (Coleman et al., 1982;
Ordonez et al., 2008). Surprisingly, the consensus from
FIG. 2. The TNF-induced pathway leading to inhibition of progenitor
cells. The first two cell types listed are relevant to conditions such as
Alzheimer’s disease, where excess TNF production is largely restricted to
the brain, and all the cell types listed can be expected to be relevant to
T2DM and severe systemic disease. As discussed and shown, these prin-
ciples are manifest in the physiology of mammalian hibernation and the
dauer forms of C. elegans.
JCLARK ET AL.
this literature (i.e., that neutralizing excess TNF is a
logical step in alleviating pathologic features of diabe-
tes) has yet to translate across to research that uses SZT
to duplicate AD. This should prove to be an excellent
model in which to develop a close laboratory-based un-
derstanding of the effects of anti-TNF agents and sex
hormones in this disease.
X. The Broader Picture—Stroke, Traumatic
Brain Injury, and Infectious Disease
This review focuses on the pathogenesis of AD, with
some reference to T2DM, but the gist of the pathway we
have constructed (Fig. 1) evidently extends to under-
standing other encephalopathies in which cerebral TNF
is increased by routes with current explanations other
than LH/FSH (Section II). Stroke, traumatic brain in-
jury (TBI), and brain involvement in malaria, a systemic
infectious disease, are examples (Fig. 3). As summarized
by Simpkins et al. (2009), AD, stroke and TBI tend to
become one syndrome with the passage of time since
onset. Moreover, TBI is often seen and postcerebral ma-
laria syndrome is usually seen (Boivin et al., 2007; Ki-
hara et al., 2009; Idro et al., 2010) in the young, before
reproduction or menopause.
Induction of TNF in the penumbra of brain ischemia,
the area surrounding the region worst affected by the
vascular obstruction, involves glutamate and nuclear
factor-
B (Kaushal and Schlichter, 2008) and is inhib-
ited by regulatory T cells (Liesz et al., 2009). Cerebral
ischemia has also been reported (Wen et al., 2004a) to
induce aberrant neuronal cell cycle re-entry that can be
reduced by 17
-estradiol, an inhibitor of TNF (Hsu et
al., 2000), a cytokine with a long history of interfering
with mitosis (Darzynkiewicz et al., 1984) and more re-
cently demonstrated to cause aneuploidy (Wu et al.,
2011), the phenomenon that sets the scene for aberrant
cell cycling and thus apoptosis. As recently reviewed by
Clark et al. (2010), trauma triggers release of inflamma-
tory cytokines through the action of mitochondrial DNA
set free from disrupted cells (Zhang et al., 2010). In
infectious diseases, much evidence exists for the direct
induction of TNF by products of the pathogen, beginning
with the example of bacterial lipopolysaccharide in the
original TNF article (Carswell et al., 1975). For instance,
ample evidence exists for the malaria toxin as a TNF
inducer (Bate et al., 1989; Tachado and Schofield, 1994).
As with AD and T2DM, insulin resistance is documented
in stroke (Calleja et al., 2011), TBI (Mowery et al., 2009;
Ley et al., 2011), and cerebral malaria (Eltahir et al.,
2010b). Likewise, A
and hyperphosphorylated
, the
proteins widely regarded as AD hallmarks and appreci-
ated to be indicators of chronically high TNF (section IV)
and GSK-3 hyperactivation induced by insulin resis-
tance (section VI), respectively, are also present in
stroke (Irving et al., 1996; Nihashi et al., 2001; Wen et
al., 2004b), TBI (Irving et al., 1996; Smith et al., 2003;
Tran et al., 2011), and cerebral malaria (Medana et al.,
2002, 2005).
Before expanding on the encephalopathies of system
infectious diseases, we recall the proposal that the A
induced in AD (Bowen et al., 2004) and deposited in the
cerebrovasculature is a response, albeit sometimes an
insufficient one, to seal these vessels to minimize blood-
brain barrier (BBB) breakdown (Atwood et al., 2002;
Atwood, 2010). These arguments, developed in part to
explain the neuroinflammatory reaction frequently ob-
served during normal aging (Wilson et al., 2008), provide
a plausible novel amyloid-based degree of complexity to
the development of the BBB changes commonly seen in
the encephalopathies of infectious disease. For instance,
this reasoning plausibly applies to the A
PP present in
cerebral malaria brains (Medana et al., 2002). In addi-
tion, the antimicrobial properties of its cleavage product,
A
(Soscia et al., 2010), can be expected to minimize
secondary bacterial invasion (a common problem in ma-
laria because of immunosuppression) at this critical lo-
cation. The association of BBB lesions with TNF gener-
ated by infectious agents is already in place in the
encephalopathies associated with sepsis (Alexander et
al., 2008), trypanosomiasis (Quan et al., 1999; Kristens-
son et al., 2010), malaria (Adams et al., 2002) influenza
(Ichiyama et al., 1996), and AIDS (Mastroianni et al.,
1990; Nolting et al., 2009).
Research on cerebral insulin resistance and GSK-3
activation is sparse regarding the encephalopathies of
infectious disease, although some publications link sys-
temic insulin resistance with poor cognitive performance
in women infected with HIV (Valcour et al., 2012) and
fatal malaria with cerebral symptoms (Eltahir et al.,
2010a).
XI. Therapeutic Implications
A. Specific Inhibition of Tumor Necrosis Factor
The obvious way to capitalize on the relationship be-
tween inflammation and insulin resistance is to specifi-
FIG. 3. Examples of the range of different inducers, in different dis-
eases, that can lead to increased cerebral TNF and thence clinically
similar outcomes.
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD K
cally neutralize excessive TNF, as is widely recognized
to be useful in a number of systemic, but not cerebral,
inflammatory diseases. Clearly, this limitation is im-
posed by the large molecular size of current therapeuti-
cally successful specific anti-TNF biological agents, such
as infliximab and etanercept, which precludes their pas-
sage through the blood-brain barrier when administered
subcutaneously or intravenously. Indeed, a negative re-
sult (a small 24-week double-blind trial) with subcuta-
neous etanercept against AD has been reported (Bohac
et al., 2002), as has a positive mouse intracerebroven-
tricular injection trial, albeit measuring only the indi-
rect indicator A
(Shi et al., 2011). Because the intrace-
rebroventricular route is a precarious one, unsuited to
regular administration to the same patient, a number of
ways to circumvent this problem are being developed to
widen the use of these highly successful biological
agents to a new patient group. The earliest of these is a
novel approach termed the perispinal route (Tobinick et
al., 2006, 2010, 2012). Its logic depends on 1) a short
period of head-down tilting to gain a gravitational ad-
vantage, 2) an awareness of anatomy of Batson’s plexus
[a valveless venous system that surrounds the spinal
column in continuum with the choroid plexus (Nathoo et
al., 2011)], and 3) knowledge of the effect of acute hy-
pertension on choroid plexus permeability [a 30-fold in-
crease in albumin in CSF within 10 min of pharmaco-
logically induced acute local hypertension (Murphy and
Johanson, 1985)]. Not surprisingly, therefore, the grav-
itational effect on this valveless blood column of a 5-min
head-down tilt of head and trunk has been reported, in
anesthetized rabbits, to increase dramatically the pas-
sage of albumin and globulin, molecules of etanercept
size, from plasma to the cerebrospinal fluid (Wen et al.,
1994). The authors noted that this would be a useful way
to get large molecules into the CSF for therapeutic
purposes.
The apparent indifference of the makers of etanercept
to the claims of the perispinal anti-brain TNF approach
to treating AD (Tobinick et al., 2006; Tobinick and
Gross, 2008) has not deterred other investigators from
aspiring to the same outcome by several approaches.
One group is developing what it refers to as a molecular
Trojan horse decoy receptor system to get a similar
anti-TNF fusion molecule into the brain (Pardridge,
2010; Zhou et al., 2011). Others (http://www.neurokine.
com/index-3.html) employ encapsulation of etanercept in
liposomes, a well recognized technology (Paolino et al.,
2011), to get the same result. It is encouraging that this
web site notes Dr. Patrick McGeer, a long-time exponent
of earlier approaches to minimizing brain inflammation
(McGeer and McGeer, 1995), as a consultant. Another
approach under way is to devise anti-TNF nanoantibod-
ies small enough to pass the BBB (Harmsen and De
Haard, 2007; Vandenbroucke et al., 2010).
It warrants noting here that the dual activity of TNF
as a component of innate immunity and disease patho-
genesis has made it inevitable that certain infections,
particularly tuberculosis and those caused by certain
protozoa, have a tendency to be exacerbated during long-
term anti-TNF therapy. This has been comprehensively
reviewed (Clark et al., 2010). The very extensive use of
this treatment in a number of inflammatory diseases,
particularly rheumatoid arthritis, demonstrates that
this challenge can be managed successfully.
B. Nonspecific Inhibition of Tumor Necrosis Factor
1. Thalidomide and Curcumin. Brain TNF levels
can also be diminished therapeutically by thalidomide
(Alkam et al., 2008; Ryu and McLarnon, 2008) or its
derivatives (Greig et al., 2004a; Tweedie et al., 2007;
Belarbi et al., 2012b), and current research programs
are examining this in an AD context. Likewise, cur-
cumin, a long-appreciated inhibitor of TNF (Chan,
1995), is used for this purpose in its original form (Cole
et al., 2007) as well as more effective (i.e., in terms of
brain entry) derivative forms (Chiu et al., 2011; Tsai et
al., 2011). All of the authors whose work is cited in this
section might have unwittingly been improving insulin
signaling as well as achieving their stated aims, but this
remains unexplored. The exception appears to be IIsik et
al. (2009), who employed the anti-inflammatory activity
of curcumin to examine its effects on both insulin resis-
tance and memory in a rat model of SZT-induced AD.
Curcumin is also reported to protect testosterone-pro-
ducing Leydig cells and pancreatic cells from toxicity
(Giannessi et al., 2008).
2. Minocycline. Minocycline is a particularly broad-
spectrum oral tetracycline that was synthesized from a
naturally occurring antibiotic decades ago (Church et
al., 1971). Being the most lipid-soluble of this class of
drug, it enters the brain more readily than the rest.
Although not without side effects, it has been known for
15 years to be anti-inflammatory in vivo (Tilley et al.,
1995), and its avid brain penetration is responsible for
the attention it has received in the neuroinflammation
literature (Peng et al., 2006). It is often termed an in-
hibitor of microglial activation, and the list of inflamma-
tory cytokines it down-regulates, in brain and else-
where, includes TNF and IL-1
(Ce´le´rier et al., 1996;
Lee et al., 2004; Suk, 2004; Wang et al., 2005a). Consis-
tent with the overarching pathway central to this re-
view, minocycline shows experimental promise as a
treatment, complementary to the others we discuss, for
the various manifestations of excess production of these
cytokines in the brain (Familian et al., 2006; Seabrook et
al., 2006; Choi et al., 2007; Fan et al., 2007; Noble et al.,
2009). A human AD trial with minocycline is under way
(http://clinicaltrials.gov/ct2/show/NCT01463384).
3. Erythropoietin. Another endogenous humoral fac-
tor, these days referred to as a cytokine but described
decades before this term was in use, is erythropoietin
(EPO). It was discovered as a hormone that drives eryth-
rogenesis and thus provides the means to deliver more
LCLARK ET AL.
oxygen to tissues. Apart from its large-scale clinical use
in treating chronic anemias, it gained notoriety as a
performance-enhancing drug, in due course an illegal
one, in sports. EPO warrants mention in this section
because it has proved to be extremely pleiotropic, in
retrospect probably because of its capacity to inhibit
nuclear factor
B-inducible pathways (Nairz et al.,
2011). It has been reported for many years to have
protective roles in stroke and traumatic brain injury (for
review, see Sargin et al., 2010; Chateauvieux et al.,
2011; Nairz et al., 2012; Sølling, 2012), and it is referred
to as a multifunctional tissue-protective cytokine. EPO
is also on record as enhancing neurogenesis (Osredkar et
al., 2010), oligodendroglial progenitors (Kim and Jung,
2010), endothelial progenitors (Xu et al., 2011), and mi-
tochondrial biogenesis (Carraway et al., 2010). It has the
potential to inhibit cell-mediated immunity as well as
disease (Nairz et al., 2011), as can anti-TNF treatment
(Mayordomo et al., 2002). Hippocampal memory is also
reported to be enhanced in mice treated with EPO for 3
weeks (Adamcio et al., 2008).
All of these phenomena have TNF mirror images in
the literature, sometimes discussed in terms of the in-
sulin resistance TNF induces (Meistrell et al., 1997;
Valerio et al., 2006; Alkam et al., 2008; Bernardino et al.,
2008; Cubbon et al., 2009; Chio et al., 2010; Chen et al.,
2011). Thus, the concept of endogenous anti-TNF activ-
ity being one of the biological roles of EPO is very plau-
sible, as is harnessing this attribute for disease therapy.
Unfortunately, the long history of indifferent recombi-
nant EPO trials in disease has been clouded by a pro-
pensity for its erythropoietic properties to dominate,
with a sometimes fatal thrombosis a feature of its
chronic use (Patel et al., 2011a). Thus nonerythropoietic
variants of this molecule are being developed. They fall
into two main categories: carbamylated EPO (Ramirez
et al., 2009; Leconte et al., 2011) and nonerythropoietic
tissue-protective proteins that mimic the three-dimen-
sional structure of EPO, such as pyroglutamate helix
B-surface peptide (Patel et al., 2011b). Hand and Brines
(2011) and Sølling (2012) have reviewed this area. In-
formation such as toxicity and efficacy within the wide
range of activities of native EPO is still being gathered
for these variants.
The retarded neurogenesis seen in infection with
Japanese encephalitis virus has been reported to be
reversible by abrogating the inflammatory response of
microglia, including TNF production, with exposure to
minocycline (Das et al., 2011). Protection against sim-
ian cerebral pathologic conditions related to HIV by
minocycline has also recently been recorded (Ratai et
al., 2010; Campbell et al., 2011). Therefore, it war-
rants testing whether all of the above reasoning ap-
plies to possible treatments for the encephalopathies
of systemic infectious disease.
As noted above, malaria comes into the category of
systemic inflammatory diseases that may develop an
associated encephalopathy. This condition in children in
tropical Africa is also noteworthy for a well documented
AD-like syndrome that can follow acute cerebral symp-
toms, despite recovery from systemic disease. This syn-
drome correlates with CSF levels of TNF and exhibits
long-term cognitive impairment, including deficits in
memory, attention, visuospatial skills, language, and
executive function (Carter et al., 2005; Boivin et al.,
2007; John et al., 2008a,b; Kihara et al., 2009). This
condition is also noted for aggressive behavior (Idro et
al., 2010), as is AD (Ballard and Walker, 1999). We have
reviewed the literature linking TNF with aggression
(Clark et al., 2010). Researchers were alerted to the
possible implications of the tissue-protective aspects of
EPO in malaria through evidence that its recombinant
form lowered TNF levels and prevented cerebral compli-
cations and death in a mouse model of the disease (Kai-
ser et al., 2006). Raised serum levels of EPO have been
reported to be associated with a lower incidence of neu-
rological sequelae in Kenyan children infected with ma-
laria (Casals-Pascual et al., 2008), leading to the sugges-
tion of using this cytokine therapeutically to protect
against brain damage (Casals-Pascual et al., 2009). Al-
though a short-term open trial in Mali showed no in-
creased mortality (Picot et al., 2009), others (John et al.,
2010) have pointed out the limited opportunity to detect
thrombotic side effects, the chief concern in the wider
literature (Patel et al., 2011a), on using unaltered eryth-
ropoietin. Meanwhile, there appears to be a consensus
(Casals-Pascual et al., 2009; John et al., 2010) to await
the outcome of basic studies of the EPO variants dis-
cussed earlier (Hand and Brines, 2011; Leconte et al.,
2011; Patel et al., 2011b). Some of the potentially less
damaging approaches depicted in Fig. 1, such as oral
minocycline or inhaled insulin, could be considered in
the meantime. Nevertheless, we regard EPO variants as
exciting future prospects for therapeutically addressing
the overarching pathway developed in this review.
C. Administering Leptin As a Counter to
Insulin Resistance
Clark et al. (2011) summarized the literature on leptin
and TNF having mirror image effects on AD. As noted,
administering additional leptin and lowering TNF levels
are both on record as improving memory and learning,
reducing anxiety, lowering A
and hyperphosphorylated
, reducing
-secretase activity, increasing dendritic
spine growth, activating GSK-3, and activating AMP
kinase-activated, pentylenetetrazole-induced seizures.
As we noted (Clark et al., 2011), this pattern had previ-
ously escaped recognition. A case for the centrality of
insulin resistance in AD is further strengthened by the
opposite effects on insulin resistance of leptin (German
et al., 2010; Koch et al., 2010) and TNF, leptin reducing
and TNF increasing it.
Leptin may also have additional direct effects on neu-
rons as an antiapoptotic, proneurogenic adipokine (Paz-
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD M
Filho et al., 2010b), and its administration to leptin-
deficient humans has altered brain function and
increased gray matter (London et al., 2011). Leptin is
currently administered in other diseases, such as lipo-
dystrophy syndromes, hypothalamic amenorrhea, and
nonalcoholic steatohepatitis, but its CNS effects have
not yet been thoroughly evaluated. Its endogenous levels
have been negatively correlated with the risk of devel-
oping AD in lean (leptin sensitive) but not obese (leptin
insensitive) older people (Lieb et al., 2009), implying
that only lean people would be susceptible to treatment
with leptin (Paz-Filho et al., 2010a).
Neither leptin nor anti-TNF agents yet appear to have
been tested against the SZT model of AD, although
leptin reduces insulin resistance (Lin et al., 2002) and
leptin deficiency increases it (German et al., 2011), in
the SZT model of T2DM. Because normal insulin sensi-
tivity keeps GSK-3
activity low, administering leptin
should also decrease its activation and
hyperphospho-
rylation. This, too, has also been reported in neurons
(Greco et al., 2009). The ability of leptin to increase
insulin sensitivity has yet to be explored as an explana-
tion for the novel observation that intracerebroventric-
ular injection of leptin dramatically, albeit briefly, im-
proves a wide range of pathologic features in a mouse
model of type 1 diabetes (Fujikawa et al., 2010). The
systemic improvements recollect those achieved, as dis-
cussed earlier, with intracerebroventricular injections of
infliximab, a commercial anti-TNF biological agent (Ar-
ruda et al., 2011).
D. Administering Insulin As a Counter to
Insulin Resistance
Interest in this approach to treating AD appears to
have arisen when it was realized that the temporary
memory improvement in patient brain function after
systemic administration of insulin was independent of
the attendant serum glucose concentration (Craft et al.,
1996, 1999). Evidently, more subtle pathways are at
work. Others found that the effects of intranasal insulin
could alter basic central nervous system function in eu-
glycemic healthy volunteers (Kern et al., 1999) and a few
years later documented the changes in CSF levels of
insulin so caused, as well as the absence of systemic
changes in insulin or glucose (Born et al., 2002). By 2004
(Benedict et al., 2004), they had reported improvements,
in a similar group of healthy volunteers, in memory and
mood after 8 weeks of treatment. Soon after, another
group reported a pilot trial that exhibited memory im-
provement after this treatment in patients with AD
(Reger et al., 2006). They also reported, then and subse-
quently (Reger et al., 2008), that responses were absent,
under the conditions tested, in apolipoprotein E (apoE)4-
positive patients. Given that a major controller of insu-
lin resistance is the inflammatory cytokine TNF (see
section V.C), it is important, when considering possible
reasons for this difference, to take into account the in-
teractions between apoE4
and TNF. One study of pos-
sible relevance, as yet unexplored, concerns the inflam-
matory status of microglia (and thus effects on insulin
resistance, although this was not in their protocol) from
mice expressing different numbers and types of human
apoE genes (Vitek et al., 2009). This is discussed further
in section XI.H.
Ott et al. (2012) discussed the pitfalls and potential of
intranasal insulin administration on cognitive function
in general, and Ketterer et al. (2011) focused on possible
ways, through new insulin analogs, such as aspart and
detemir, to optimize reduction of cerebral insulin resis-
tance. Subsequently, Benedict et al. (2012) and Schio¨th
et al. (2012) documented an association between im-
paired insulin resistance with deficits in verbal fluency
and temporal lobe gray matter volume in the elderly and
evaluated the therapeutic potential of reversing this
resistance with intranasal insulin. The former of these
publications is reminiscent of the association observed
between insulin resistance and executive function (Ab-
batecola et al., 2004).
E. Glucagon-Like Peptide-1 Mimetics and Dipeptidyl
Peptidase-4 Inhibitors As Counters to
Insulin Resistance
Being rapidly degraded (minutes) in vivo by DPP-4
(see section V.E), native GLP-1 is impractical as a ther-
apy. Hence, degradation-resistant GLP-1 receptor ago-
nists, often termed GLP-1 mimetics, have been devel-
oped (Ahre´n, 2011a,b), and a number of these agents are
in therapeutic use subcutaneously in T2DM. Exenatide
is an example from a group of drugs based on exendin-4,
a GLP-1-like molecule isolated from a reptile. Another
approach, based on synthesizing GLP-1 analogs, has led
to other subcutaneous agents, such as liraglutide (Buse
et al., 2009). Both of these types of GLP-1 mimetics pass
through the blood-brain barrier and have proved to be
strikingly active against AD models (Perry et al., 2003;
Liu et al., 2009; Porter et al., 2010; Li et al., 2011;
McClean et al., 2011) as well as against a model of the
cognitive defects in T2DM (Gault et al., 2010). Recent
basic studies give impressively detailed reinforcement to
this approach (Bomfim et al., 2012; Talbot et al., 2012).
In addition, a number of agents have been developed,
such as sitagliptin, which inhibits the catalytic site of
DPP-4 (Ahre´n and Foley, 2008; Ahre´n, 2009) to extend
the life of endogenous GLP-1. They are marketed for
treating T2DM and have the practical advantage of be-
ing administered orally, although their testing in AD
models remains in its infancy (D’Amico et al., 2010).
F. Glycogen Synthase Kinase-3 Antagonists
SB216763, presumably tested for its effects on endotoxin-
treated human monocytes (Martin et al., 2005), showed a
degree of promise in a model of AD generated by injecting
an A
oligomer into aged rats but made outcomes worse in
control rats (Hu et al., 2009). As this group suggested, less
NCLARK ET AL.
potent inhibitors that do not inhibit constitutional GSK-3
may be necessary. The study by Engel et al. (2006) is
consistent with this. In terms of lowering phosphorylated
levels and decreasing spatial memory loss, others success-
fully tested a GSK-3 inhibitor, a thiadiazolidinone termed
4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (NP12),
in a transgenic mouse model of AD (Sereno´ et al., 2009).
However, the mice did not live longer. 2-Methyl-5-(3-{4-
[(S)-methylsulfinyl]phenyl}-1-benzofuran-5-yl)-1,3,4-
oxadiazole, another novel GSK-3 inhibitor, has recently
been reported to produce a similar positive outcome in
vitro and in a mouse model (Onishi et al., 2011). Success-
fully targeting GSK-3, now a goal in many disease fields, is
evidently a complex undertaking. Engagingly, cardiac re-
searchers have referred to the challenge it presents as a
very sharp double-edged sword (Cheng et al., 2011). This
approach, as well as those discussed in the rest of this
section, is shown in Fig. 1. The major treatment concepts
and their intended or predictable consequences are col-
lected in Table 1.
H. Apolipoprotein E Mimetics and Bexarotene
As reviewed by Laskowitz et al. (2001), the protein
apoE was identified by its role in the transport and
metabolism of cholesterol and triglycerides. It is the
major apolipoprotein generated in the brain, where it
originates from glial cells. Human genetic variation ac-
commodates three isoforms: apoE2, apoE3 (the most
common), and apoE4. In brief, the more apoE4 gener-
ated, the less functional apoE protein is present (Riddell
et al., 2008). Almost as soon as it was appreciated that
the presence of the apoE4 allele was robustly associated
with an increased risk of developing most forms of AD
(Corder et al., 1993), two apparently unrelated threads
of research on apoE function developed. One was based
on connecting apoE, through its lipid-binding domain, to
the formation of the A
and hyperphosphorylated
, the
histologically discernible proteins historically associated
with AD (Strittmatter et al., 1993, 1994). The other
thread, not lipid-related, focusing on innate immunity
and inflammatory mediators rather than lipids, sought
to explain why the link between apoE4 and disease risk
was far wider than AD, encompassing traumatic brain
injury and stroke, and bacterial infections (Roselaar and
Daugherty, 1998; de Bont et al., 1999), which have all
been argued to be inflammatory conditions since the
early 1990s. Indeed, the apoE4 connection with disease
susceptibility goes as far as HIV dementia (Corder et al.,
1998) and cerebral malaria (Aucan et al., 2004).
This second line of enquiry led to seminal outcomes
such as suppression of glial cell secretion of TNF by
apoE (Laskowitz et al., 1997) and inhibition of glial cell
activation and the endogenous CNS inflammatory re-
sponse (Lynch et al., 2001) and the general type 1 in-
flammatory response (Ali et al., 2005), which is medi-
ated by cytokines such as TNF and IL-1
.A
comprehensive review of these concepts appeared 3
years ago (Vitek et al., 2009). This approach has led to
the attainment of a clinically useful anti-inflammatory
milieu in many mouse models of inflammatory disease
by subcutaneous injection of segments of the apoE mol-
ecule (apoE mimetics) that are small enough to enter the
brain. This duplicates the anti-inflammatory action of
complete apoE (Laskowitz et al., 2001). Examples in-
clude traumatic brain injury (Lynch et al., 2005; Las-
kowitz et al., 2007; Hoane et al., 2009; Kaufman et al.,
2010), stroke (Tukhovskaya et al., 2009), and AD (Vitek
et al., 2012).
Cramer et al. (2012) describe removal of A
plaque
and correction of functional deficits in a strain of mice
prone to AD-like changes after oral administration of
bexarotene, an anti-tumor drug in clinical use. This
agent is small enough to enter the brain, where it in-
creases endogenous apoE levels through its activity as a
retinoid X receptor agonist. In a functional sense, bex-
arotene therefore promises to be the equivalent of the
apoE mimetics. Surprisingly, A
plaque removal was
the only mechanism considered by these authors, de-
spite the doubt cast on the utility of this endpoint by the
AD patient trial of AN1792 (A
42; Elan Pharmaceuti-
cals, South San Francisco, CA) several years ago (Hol-
mes et al., 2008). It is useful to recall, when interpreting
TABLE 1
A reference guide for the treatment concepts embodied in this review, and their consequences, either intended by the authors or predictable from
the literature
Treatment Concepts Predictable Consequences in
the Brain Predictable Downstream Consequences in
the Brain Reference
apoE mimetics 2TNF 2Insulin resistance Laskowitz et al., 2001
Thalidomide derivatives 2TNF 2Insulin resistance Greig et al., 2004a
Anti-TNF biologicals 2TNF 2Insulin resistance Tobinick et al., 2006
Minocycline 2TNF 2Insulin resistance Seabrook et al.. 2006
Leuprolide 2TNF 2Insulin resistance Clark and Atwood, 2011
Curcumin derivatives 2TNF 2Insulin resistance Tsai et al., 2011
Erythropoietin 2TNF 2Insulin resistance Nairz et al., 2011a
Bexarotene 2TNF 2Insulin resistance Cramer et al. 2012
GLP-1 mimetics 2Insulin resistance 2GSK3 activation Perry et al., 2003
Intranasal insulin 2Insulin resistance 2GSK3 activation Benedict et al., 2004
Leptin 2Insulin resistance 2GSK3 activation Koch et al., 2010
DPP-4 inhibitors 2Insulin resistance 2GSK3 activation D’Amico et al., 2010
GSK-3 antagonists 2GSK3 activation 2Harmful enzyme phosphorylation Onishi et al., 2011
2, reduction.
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD O
this bexarotene data, that anti-TNF treatment produces
a very similar outcome in the same mouse model (Shi et
al., 2011), and an apoE mimetic has since done so in a
related mouse strain (Vitek et al., 2012). Literature ex-
ists on the side effects of bexarotene, but whether these
arise from high apoE or other consequences of retinoid X
receptor activation has yet to be determined.
XII. Conclusions
It seems not yet to have been taken into account in AD
research that TNF and related inflammatory cytokines
induce insulin resistance or that SZT also induces TNF.
The introduction of these concepts into this field consol-
idates numerous non-A
models of AD. Accordingly, nu-
merous proposed treatments for AD, presently under-
taken with different rationales, seem to be functionally
linked by events that lower TNF levels (and thus insulin
resistance), lower insulin resistance directly, or deal
with its consequences. These presently independent sets
of arguments for therapy reinforce the logic of chronic
cerebral insulin resistance and, therefore, the degree to
which GSK-3 activation, and thus mitochondrial dys-
function, is central to understanding this disease. We
have reasoned that these superficially unrelated ap-
proaches to treatment are all aimed at chronic inflam-
mation and its consequences. Thus, they are, in a sense,
one tool, which invites collaborative searches for thera-
peutic synergy.
When considering how rapidly the therapies discussed
in this review might have the opportunity to demon-
strate whether they can help patients, we note that
anti-TNF biological agents, minocycline, and GLP-1 mi-
metics already have a history of clinical use for other
conditions, the first two for much longer, and on a much
larger scale, than the third. Intranasal insulin has been
used in human trials, produced no side effects, and
seems innately harmless at such doses. Leptin, like in-
sulin an endogenous molecule, has been used long-term
in a small number of patients without apparent harm,
whereas EPO variants still require basic toxicity and
efficacy studies. ApoE mimetics and GSK-3 inhibitors
warrant extending beyond rodent models.
Authorship Contributions
Wrote or contributed to the writing of the manuscript: Clark, At-
wood, Bowen, Paz-Filho, and Vissel.
References
Abbas A, Imrie H, Viswambharan H, Sukumar P, Rajwani A, Cubbon RM, Gage M,
Smith J, Galloway S, Yuldeshava N, et al. (2011) The insulin-like growth factor-1
receptor is a negative regulator of nitric oxide bioavailability and insulin sensitiv-
ity in the endothelium. Diabetes 60:2169–2178.
Abbatecola AM, Paolisso G, Lamponi M, Bandinelli S, Lauretani F, Launer L, and
Ferrucci L (2004) Insulin resistance and executive dysfunction in older persons.
J Am Geriatr Soc 52:1713–1718.
Abdipranoto-Cowley A, Park JS, Croucher D, Daniel J, Henshall S, Galbraith S,
Mervin K, and Vissel B (2009) Activin A is essential for neurogenesis following
neurodegeneration. Stem Cells 27:1330–1346.
Adamcio B, Sargin D, Stradomska A, Medrihan L, Gertler C, Theis F, Zhang M,
Mu¨ ller M, Hassouna I, Hannke K, et al. (2008) Erythropoietin enhances hippocam-
pal long-term potentiation and memory. BMC Biol 6:37.
Adamo M, Raizada MK, and LeRoith D (1989) Insulin and insulin-like growth factor
receptors in the nervous system. Mol Neurobiol 3:71–100.
Adams S, Brown H, and Turner G (2002) Breaking down the blood-brain barrier:
signaling a path to cerebral malaria? Trends Parasitol 18:360–366.
Aggarwal BB, Kohr WJ, Hass PE, Moffat B, Spencer SA, Henzel WJ, Bringman TS,
Nedwin GE, Goeddel DV, and Harkins RN (1985) Human tumor necrosis factor.
Production, purification, and characterization. J Biol Chem 260:2345–2354.
Ahre´ n B (2009) Clinical results of treating type 2 diabetic patients with sitagliptin,
vildagliptin or saxagliptin–diabetes control and potential adverse events. Best
Pract Res Clin Endocrinol Metab 23:487–498.
Ahre´ n B (2011a) The future of incretin-based therapy: novel avenues–novel targets.
Diabetes Obes Metab 13 (Suppl 1):158–166.
Ahre´ n B (2011b) GLP-1 for type 2 diabetes. Exp Cell Res 317:1239–1245.
Ahre´ n B and Foley JE (2008) The islet enhancer vildagliptin: mechanisms of im-
proved glucose metabolism. Int J Clin Pract Suppl (159):8–14.
Alblowi J, Kayal RA, Siqueira M, Siqueria M, McKenzie E, Krothapalli N, McLean
J, Conn J, Nikolajczyk B, Einhorn TA, et al. (2009) High levels of tumor necrosis
factor-alpha contribute to accelerated loss of cartilage in diabetic fracture healing.
Am J Pathol 175:1574–1585.
Alexander JJ, Jacob A, Cunningham P, Hensley L, and Quigg RJ (2008) TNF is a key
mediator of septic encephalopathy acting through its receptor, TNF receptor-1.
Neurochem Int 52:447–456.
Ali K, Middleton M, Pure´ E, and Rader DJ (2005) Apolipoprotein E suppresses the
type I inflammatory response in vivo. Circ Res 97:922–927.
Alkam T, Nitta A, Mizoguchi H, Saito K, Seshima M, Itoh A, Yamada K, and
Nabeshima T (2008) Restraining tumor necrosis factor-alpha by thalidomide pre-
vents the amyloid beta-induced impairment of recognition memory in mice. Behav
Brain Res 189:100–106.
Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, Price JW, 3rd,
Kang L, Rabinovitch PS, et al. (2009) Mitochondrial H2O2 emission and cellular
redox state link excess fat intake to insulin resistance in both rodents and humans.
J Clin Invest 119:573–581.
Arau´ jo EP, De Souza CT, Ueno M, Cintra DE, Bertolo MB, Carvalheira JB, Saad MJ,
and Velloso LA (2007) Infliximab restores glucose homeostasis in an animal model
of diet-induced obesity and diabetes. Endocrinology 148:5991–5997.
Arruda AP, Milanski M, Coope A, Torsoni AS, Ropelle E, Carvalho DP, Carvalheira
JB, and Velloso LA (2011) Low-grade hypothalamic inflammation leads to defec-
tive thermogenesis, insulin resistance, and impaired insulin secretion. Endocri-
nology 152:1314–1326.
Arvanitakis Z, Wilson RS, Bienias JL, Evans DA, and Bennett DA (2004) Diabetes
mellitus and risk of Alzheimer disease and decline in cognitive function. Arch
Neurol 61:661–666.
Asthana S, Baker LD, Craft S, Stanczyk FZ, Veith RC, Raskind MA, and Plymate SR
(2001) High-dose estradiol improves cognition for women with AD: results of a
randomized study. Neurology 57:605–612.
Atwood CS (2010) Amyloid-beta aggregation as a protective acute-phase response to
injury/neurodegeneration: a barrier function for amyloid-beta deposits, in Func-
tional Amyloid Aggregation (Rigacci S and Bucciantini M eds) pp 115–134, Re-
search Signpost, Kerala, India.
Atwood CS, Bishop GM, Perry G, and Smith MA (2002) Amyloid-beta: a vascular
sealant that protects against hemorrhage? J Neurosci Res 70:356.
Aucan C, Walley AJ, and Hill AV (2004) Common apolipoprotein E polymorphisms
and risk of clinical malaria in the Gambia. Journal of Medical Genetics 41:21–24.
Baker LD, Cross DJ, Minoshima S, Belongia D, Watson GS, and Craft S (2011)
Insulin resistance and Alzheimer-like reductions in regional cerebral glucose
metabolism for cognitively normal adults with prediabetes or early type 2 diabetes.
Arch Neurol 68:51–57.
Balaraman Y, Limaye AR, Levey AI, and Srinivasan S (2006) Glycogen synthase
kinase 3beta and Alzheimer’s disease: pathophysiological and therapeutic signif-
icance. Cell Mol Life Sci 63:1226–1235.
Ballard C and Walker M (1999) Neuropsychiatric aspects of Alzheimer’s disease.
Curr Psychiatry Rep 1:49–60.
Bate CA, Taverne J, and Playfair JH (1989) Soluble malarial antigens are toxic and
induce the production of tumour necrosis factor in vivo. Immunology 66:600–605.
Beier K, Vo¨ lkl A, and Fahimi HD (1997) TNF-alpha downregulates the peroxisome
proliferator activated receptor-alpha and the mRNAs encoding peroxisomal pro-
teins in rat liver. FEBS Lett 412:385–387.
Belarbi K, Arellano C, Ferguson R, Jopson T, and Rosi S (2012a) Chronic neuroin-
flammation impacts the recruitment of adult-born neurons into behaviorally rel-
evant hippocampal networks. Brain Behav Immun 26:18–23.
Belarbi K, Jopson T, Tweedie D, Arellano C, Luo W, Greig NH, and Rosi S (2012b)
TNF-alpha protein synthesis inhibitor restores neuronal function and reverses
cognitive deficits induced by chronic neuroinflammation. J Neuroinflammation
9:23.
Benedict C, Brooks SJ, Kullberg J, Burgos J, Kempton MJ, Nordenskjo¨ldR,Ny-
lander R, Kilander L, Craft S, Larsson EM, et al. (2012) Impaired insulin sensi-
tivity as indexed by the HOMA score is associated with deficits in verbal fluency
and temporal lobe gray matter volume in elderly men and women. Diabetes Care
35:488–494.
Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, and Kern W
(2004) Intranasal insulin improves memory in humans. Psychoneuroendocrinology
29:1326–1334.
Bernardino L, Agasse F, Silva B, Ferreira R, Grade S, and Malva JO (2008) Tumor
necrosis factor-alpha modulates survival, proliferation, and neuronal differentia-
tion in neonatal subventricular zone cell cultures. Stem Cells 26:2361–2371.
Berry A, Tomidokoro Y, Ghiso J, and Thornton J (2008) Human chorionic gonado-
tropin (a luteinizing hormone homologue) decreases spatial memory and increases
brain amyloid-beta levels in female rats. Horm Behav 54:143–152.
Beutler B, Greenwald D, Hulmes JD, Chang M, Pan YC, Mathison J, Ulevitch R, and
Cerami A (1985) Identity of tumour necrosis factor and the macrophage-secreted
factor cachectin. Nature 316:552–554.
Beutler B and Poltorak A (2001) Sepsis and evolution of the innate immune re-
sponse. Crit Care Med 29:S2–S6.
PCLARK ET AL.
Blass JP and Gibson GE (1991) The role of oxidative abnormalities in the patho-
physiology of Alzheimer’s disease. Rev Neurol 147:513–525.
Blondel O and Portha B (1989) Early appearance of in vivo insulin resistance in adult
streptozotocin-injected rats. Diabetes Metab 15:382–387.
Bluthe´ RM, Pawlowski M, Suarez S, Parnet P, Pittman Q, Kelley KW, and Dantzer
R (1994) Synergy between tumor necrosis factor alpha and interleukin-1 in the
induction of sickness behavior in mice. Psychoneuroendocrinology 19:197–207.
Boehm J, Fischer K, and Bohnert M (2010) Putative role of TNF-alpha, interleukin-8
and ICAM-1 as indicators of an early inflammatory reaction after burn: a morpho-
logical and immunohistochemical study of lung tissue of fire victims. J Clin Pathol
63:967–971.
Bohac D, Burke W, Cotter R, Zheng J and Potter J (2002) A 24-week randomized,
double-blind, placebo-controlled study of the efficacy and tolerability of TNFR: Fc
(etanercept) in the treatment of dementia of the Alzheimer type (Abstract 315).
Neurobiol Aging 23 (Suppl 1):S1–S606.
Boivin MJ, Bangirana P, Byarugaba J, Opoka RO, Idro R, Jurek AM, and John CC
(2007) Cognitive impairment after cerebral malaria in children: a prospective
study. Pediatrics 119:e360–e366.
Bomfim TR, Forny-Germano L, Sathler LB, Brito-Moreira J, Houzel JC, Decker H,
Silverman MA, Kazi H, Melo HM, McClean PL, et al. (2012) An anti-diabetes agent
protects the mouse brain from defective insulin signaling caused by Alzheimer’s
disease-associated Abeta oligomers. J Clin Invest 122:1339–1353.
Borger E, Aitken L, Muirhead KE, Allen ZE, Ainge JA, Conway SJ, and Gunn-Moore
FJ (2011) Mitochondrial beta-amyloid in Alzheimer’s disease. Biochem Soc Trans
39:868–873.
Borgs L, Beukelaers P, Vandenbosch R, Nguyen L, Moonen G, Maquet P, Albrecht U,
Belachew S, and Malgrange B (2009) Period 2 regulates neural stem/progenitor
cell proliferation in the adult hippocampus. BMC Neurosci 10:30.
Born J, Lange T, Kern W, McGregor GP, Bickel U, and Fehm HL (2002) Sniffing
neuropeptides: a transnasal approach to the human brain. Nat Neurosci 5:514
516.
Bour ES, Ward LK, Cornman GA, and Isom HC (1996) Tumor necrosis factor-alpha-
induced apoptosis in hepatocytes in long-term culture. Am J Pathol 148:485–495.
Bowen RL and Atwood CS (2004) Living and dying for sex. A theory of aging based
on the modulation of cell cycle signaling by reproductive hormones. Gerontology
50:265–290.
Bowen RL, Verdile G, Liu T, Parlow AF, Perry G, Smith MA, Martins RN, and
Atwood CS (2004) Luteinizing hormone, a reproductive regulator that modulates
the processing of amyloid-beta precursor protein and amyloid-beta deposition.
J Biol Chem 279:20539–20545.
Boyadjieva NI and Sarkar DK (2010) Role of microglia in ethanol’s apoptotic action
on hypothalamic neuronal cells in primary cultures. Alcohol Clin Exp Res 34:
1835–1842.
Brennan FM, Chantry D, Jackson A, Maini R, and Feldmann M (1989) Inhibitory
effect of TNF alpha antibodies on synovial cell interleukin-1 production in rheu-
matoid arthritis. Lancet 2:244–247.
Brugg B, Dubreuil YL, Huber G, Wollman EE, Delhaye-Bouchaud N, and Mariani J
(1995) Inflammatory processes induce beta-amyloid precursor protein changes in
mouse brain. Proc Natl Acad Sci USA 92:3032–3035.
Bryan KJ, Mudd JC, Richardson SL, Chang J, Lee HG, Zhu X, Smith MA, and
Casadesus G (2010) Down-regulation of serum gonadotropins is as effective as
estrogen replacement at improving menopause-associated cognitive deficits.
J Neurochem 112:870–881.
Bryant NJ, Govers R, and James DE (2002) Regulated transport of the glucose
transporter GLUT4. Nat Rev Mol Cell Biol 3:267–277.
Buchhave P, Zetterberg H, Blennow K, Minthon L, Janciauskiene S, and Hansson O
(2010) Soluble TNF receptors are associated with Abeta metabolism and conver-
sion to dementia in subjects with mild cognitive impairment. Neurobiol Aging
31:1877–1884.
Buse JB, Rosenstock J, Sesti G, Schmidt WE, Montanya E, Brett JH, Zychma M,
Blonde L, and LEAD-6 Study Group (2009) Liraglutide once a day versus ex-
enatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group,
multinational, open-label trial (LEAD-6). Lancet 374:39–47.
Buxbaum JD, Liu KN, Luo Y, Slack JL, Stocking KL, Peschon JJ, Johnson RS,
Castner BJ, Cerretti DP, and Black RA (1998) Evidence that tumor necrosis factor
alpha converting enzyme is involved in regulated alpha-secretase cleavage of the
Alzheimer amyloid protein precursor. J Biol Chem 273:27765–27767.
Cabou C, Campistron G, Marsollier N, Leloup C, Cruciani-Guglielmacci C, Pe´ nicaud
L, Drucker DJ, Magnan C, and Burcelin R (2008) Brain glucagon-like peptide-1
regulates arterial blood flow, heart rate, and insulin sensitivity. Diabetes 57:2577–
2587.
Cai Z, Zhao Y, Yao S, and Bin Zhao B (2011) Increases in beta-amyloid protein in the
hippocampus caused by diabetic metabolic disorder are blocked by minocycline
through inhibition of NF-kappaB pathway activation. Pharmacol Rep 63:381–391.
Calleja AI, García-Bermejo P, Cortijo E, Bustamante R, Rojo Martínez E, Gonza´ lez
Sarmiento E, Ferna´ ndez-Herranz R, and Arenillas JF (2011) Insulin resistance is
associated with a poor response to intravenous thrombolysis in acute ischemic
stroke. Diabetes Care 34:2413–2417.
Campbell JH, Burdo TH, Autissier P, Bombardier JP, Westmoreland SV, Soulas C,
Gonza´ lez RG, Ratai EM, and Williams KC (2011) Minocycline inhibition of mono-
cyte activation correlates with neuronal protection in SIV neuroAIDS. PLoS One
6:e18688.
Carraway MS, Suliman HB, Jones WS, Chen CW, Babiker A, and Piantadosi CA
(2010) Erythropoietin activates mitochondrial biogenesis and couples red cell mass
to mitochondrial mass in the heart. Circ Res 106:1722–1730.
Carroll JC, Rosario ER, Chang L, Stanczyk FZ, Oddo S, LaFerla FM, and Pike CJ
(2007) Progesterone and estrogen regulate Alzheimer-like neuropathology in fe-
male 3xTg-AD mice. J Neurosci 27:13357–13365.
Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, and Williamson B (1975) An
endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci
USA 72:3666–3670.
Carter JA, Mung’ala-Odera V, Neville BG, Murira G, Mturi N, Musumba C, and
Newton CR (2005) Persistent neurocognitive impairments associated with severe
falciparum malaria in Kenyan children. J Neurol Neurosurg Psychiatry 76:476
481.
Casadesus G, Milliken EL, Webber KM, Bowen RL, Lei Z, Rao CV, Perry G, Keri RA,
and Smith MA (2007) Increases in luteinizing hormone are associated with de-
clines in cognitive performance. Mol Cell Endocrinol 269:107–111.
Casadesus G, Webber KM, Atwood CS, Pappolla MA, Perry G, Bowen RL, and Smith
MA (2006) Luteinizing hormone modulates cognition and amyloid-beta deposition
in Alzheimer APP transgenic mice. Biochim Biophys Acta 1762:447–452.
Casals-Pascual C, Idro R, Gicheru N, Gwer S, Kitsao B, Gitau E, Mwakesi R, Roberts
DJ, and Newton CR (2008) High levels of erythropoietin are associated with
protection against neurological sequelae in African children with cerebral malaria.
Proc Natl Acad Sci USA 105:2634–2639.
Casals-Pascual C, Idro R, Picot S, Roberts DJ, and Newton CR (2009) Can erythro-
poietin be used to prevent brain damage in cerebral malaria? Trends Parasitol
25:30–36.
Castellani R, Hirai K, Aliev G, Drew KL, Nunomura A, Takeda A, Cash AD,
Obrenovich ME, Perry G, and Smith MA (2002) Role of mitochondrial dysfunction
in Alzheimer’s disease. J Neurosci Res 70:357–360.
Cavadini G, Petrzilka S, Kohler P, Jud C, Tobler I, Birchler T, and Fontana A (2007)
TNF-alpha suppresses the expression of clock genes by interfering with E-box-
mediated transcription. Proc Natl Acad Sci USA 104:12843–12848.
Ce´le´ rier P, Litoux P, and Dre´no B (1996) In vitro modulation of epidermal inflam-
matory cytokines (IL-1 alpha, IL-6, TNF alpha) by minocycline. Arch Dermatol Res
288:411–414.
Chan MM (1995) Inhibition of tumor necrosis factor by curcumin, a phytochemical.
Biochem Pharmacol 49:1551–1556.
Charles P, Elliott MJ, Davis D, Potter A, Kalden JR, Antoni C, Breedveld FC, Smolen
JS, Eberl G, deWoody K, et al. (1999) Regulation of cytokines, cytokine inhibitors,
and acute-phase proteins following anti-TNF-alpha therapy in rheumatoid arthri-
tis. J Immunol 163:1521–1528.
Chateauvieux S, Grigorakaki C, Morceau F, Dicato M, and Diederich M (2011)
Erythropoietin, erythropoiesis and beyond. Biochem Pharmacol 82:1291–1303.
Chaudry IH, Sayeed MM, and Baue AE (1974) Insulin resistance in experimental
shock. Arch Surg 109:412–415.
Chawla A, Nguyen KD, and Goh YP (2011) Macrophage-mediated inflammation in
metabolic disease. Nat Rev Immunol 11:738–749.
Chen KB, Uchida K, Nakajima H, Yayama T, Hirai T, Watanabe S, Guerrero AR,
Kobayashi S, Ma WY, Liu SY, et al. (2011) Tumor necrosis factor-alpha antagonist
reduces apoptosis of neurons and oligodendroglia in rat spinal cord injury. Spine
36:1350–1358.
Chen XH, Zhao YP, Xue M, Ji CB, Gao CL, Zhu JG, Qin DN, Kou CZ, Qin XH, Tong
ML, et al. (2010) TNF-alpha induces mitochondrial dysfunction in 3T3–L1 adi-
pocytes. Mol Cell Endocrinol 328:63–69.
Cheng H, Woodgett J, Maamari M, and Force T (2011) Targeting GSK-3 family
members in the heart: a very sharp double-edged sword. J Mol Cell Cardiol
51:607–613.
Cheng HH, Ma CY, Chou TW, Chen YY, and Lai MH (2010) Gamma-oryzanol
ameliorates insulin resistance and hyperlipidemia in rats with streptozotocin/
nicotinamide-induced type 2 diabetes. Int J Vitam Nutr Res 80:45–53.
Chio CC, Lin JW, Chang MW, Wang CC, Kuo JR, Yang CZ, and Chang CP (2010)
Therapeutic evaluation of etanercept in a model of traumatic brain injury. J Neu-
rochem 115:921–929.
Chiu SS, Lui E, Majeed M, Vishwanatha JK, Ranjan AP, Maitra A, Pramanik D,
Smith JA, and Helson L (2011) Differential distribution of intravenous curcumin
formulations in the rat brain. Anticancer Res 31:907–911.
Choi Y, Kim HS, Shin KY, Kim EM, Kim M, Kim HS, Park CH, Jeong YH, Yoo J, Lee
JP, et al. (2007) Minocycline attenuates neuronal cell death and improves cogni-
tive impairment in Alzheimer’s disease models. Neuropsychopharmacology 32:
2393–2404.
Chowdhury SK, Zherebitskaya E, Smith DR, Akude E, Chattopadhyay S, Jolivalt
CG, Calcutt NA, and Fernyhough P (2010) Mitochondrial respiratory chain dys-
function in dorsal root ganglia of streptozotocin-induced diabetic rats and its
correction by insulin treatment. Diabetes 59:1082–1091.
Church RF, Schaub RE, and Weiss MJ (1971) Synthesis of 7-dimethylamino-6-
demethyl-6-deoxytetracycline (minocycline) via 9-nitro-6-demethyl-6-deoxytetra-
cycline. J Org Chem 36:723–725.
Clark IA, Alleva LM, and Vissel B (2010) The roles of TNF in brain dysfunction and
disease. Pharmacol Ther 128:519–548.
Clark IA, Alleva LM, and Vissel B (2011) TNF and leptin tell essentially the same
story in Alzheimer’s disease. J Alzheimers Dis 26:201–205.
Clark IA and Atwood CS (2011) Is TNF a link between aging-related reproductive
endocrine dyscrasia and Alzheimer’s disease? J Alzheimers Dis 27:691–699.
Clark IA, Budd AC, and Alleva LM (2008) Sickness behaviour pushed too far–the
basis of the syndrome seen in severe protozoal, bacterial and viral diseases and
post-trauma. Malar J 7:208.
Clark IA and Chaudhri G (1988) Tumour necrosis factor may contribute to the
anaemia of malaria by causing dyserythropoiesis and erythrophagocytosis. Br J
Haematol 70:99–103.
Clark IA, Virelizier JL, Carswell EA, and Wood PR (1981) Possible importance of
macrophage-derived mediators in acute malaria. Infect Immun 32:1058–1066.
Cole GM, Teter B, and Frautschy SA (2007) Neuroprotective effects of curcumin. Adv
Exp Med Biol 595:197–212.
Coleman DL, Leiter EH, and Schwizer RW (1982) Therapeutic effects of dehydro-
epiandrosterone (DHEA) in diabetic mice. Diabetes 31:830–833.
Corder EH, Robertson K, Lannfelt L, Bogdanovic N, Eggertsen G, Wilkins J, and
Hall C (1998) HIV-infected subjects with the E4 allele for APOE have excess
dementia and peripheral neuropathy. Nat Med 4:1182–1184.
Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW,
Roses AD, Haines JL, and Pericak-Vance MA (1993) Gene dose of apolipoprotein E
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD Q
type 4 allele and the risk of Alzheimer’s disease in late onset families. Science
261:921–923.
Correia SC, Santos RX, Perry G, Zhu X, Moreira PI, and Smith MA (2011) Insulin-
resistant brain state: the culprit in sporadic Alzheimer’s disease? Ageing Res Rev
10:264–273.
Craft S, Asthana S, Newcomer JW, Wilkinson CW, Matos IT, Baker LD, Cherrier M,
Lofgreen C, Latendresse S, Petrova A, et al. (1999) Enhancement of memory in
Alzheimer disease with insulin and somatostatin, but not glucose. Arch Gen
Psychiatry 56:1135–1140.
Craft S, Newcomer J, Kanne S, Dagogo-Jack S, Cryer P, Sheline Y, Luby J, Dagogo-
Jack A, and Alderson A (1996) Memory improvement following induced hyperin-
sulinemia in Alzheimer’s disease. Neurobiol Aging 17:123–130.
Cramer PE, Cirrito JR, Wesson DW, Lee CY, Karlo JC, Zinn AE, Casali BT, Restivo
JL, Goebel WD, James MJ, et al. (2012) ApoE-directed therapeutics rapidly clear
beta-amyloid and reverse deficits in AD mouse models. Science 335:1503–1506.
Cross DA, Alessi DR, Cohen P, Andjelkovich M, and Hemmings BA (1995) Inhibition
of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature
378:785–789.
Cubbon RM, Kahn MB, and Wheatcroft SB (2009) Effects of insulin resistance on
endothelial progenitor cells and vascular repair. Clin Sci (Lond)117:173–190.
D’Amico M, Di Filippo C, Marfella R, Abbatecola AM, Ferraraccio F, Rossi F, and
Paolisso G (2010) Long-term inhibition of dipeptidyl peptidase-4 in Alzheimer’s
prone mice. Exp Gerontol 45:202–207.
Dantzer R and Kelley KW (2007) Twenty years of research on cytokine-induced
sickness behavior. Brain Behav Immun 21:153–160.
Darzynkiewicz Z, Williamson B, Carswell EA, and Old LJ (1984) Cell cycle-specific
effects of tumor necrosis factor. Cancer Res 44:83–90.
Das S, Dutta K, Kumawat KL, Ghoshal A, Adhya D, and Basu (2011)A Abrogated
inflammatory response promotes neurogenesis in a murine model of Japanese
encephalitis. PLoS One 6:e17225.
Davis TM, Brown AE, and Smith CD (1993) Metabolic disturbances in Plasmodium
coatneyi-infected rhesus monkeys. Int J Parasitol 23:557–563.
de Bont N, Netea MG, Demacker PN, Verschueren I, Kullberg BJ, van Dijk KW, van
der Meer JW, and Stalenhoef AF (1999) Apolipoprotein E knock-out mice are
highly susceptible to endotoxemia and Klebsiella pneumoniae infection. J Lipid
Res 40:680–685.
de Castro J, Sevillano J, Marciniak J, Rodriguez R, Gonza´ lez-Martín C, Viana M,
Eun-suk OH, de Mouzon SH, Herrera E, and Ramos MP (2011) Implication of low
level inflammation in the insulin resistance of adipose tissue at late pregnancy.
Endocrinology 152:4094–4105.
de la Monte SM, Tong M, Bowling N, and Moskal P (2011) si-RNA inhibition of brain
insulin or insulin-like growth factor receptors causes developmental cerebellar
abnormalities: relevance to fetal alcohol spectrum disorder. Mol Brain 4:13.
de la Monte SM and Wands JR (2008) Alzheimer’s disease is type 3 diabetes-evidence
reviewed. J Diabetes Sci Technol 2:1101–1113.
de la Monte SM and Wands JR (2010) Role of central nervous system insulin
resistance in fetal alcohol spectrum disorders. J Popul Ther Clin Pharmacol
17:e390–404.
de la Monte SM, Xu XJ, and Wands JR (2005) Ethanol inhibits insulin expression
and actions in the developing brain. Cell Mol Life Sci 62:1131–1145.
Desouza CV, Hamel FG, Bidasee K, and O’Connell K (2011) Role of inflammation
and insulin resistance in endothelial progenitor cell dysfunction. Diabetes 60:
1286–1294.
Devaraj S, Tobias P, and Jialal I (2011) Knockout of toll-like receptor-4 attenuates
the pro-inflammatory state of diabetes. Cytokine 55:441–445.
Diehl AM and Rai R (1996) Review: regulation of liver regeneration by pro-
inflammatory cytokines. J Gastroenterol Hepatol 11:466–470.
Doble BW and Woodgett JR (2003) GSK-3: tricks of the trade for a multi-tasking
kinase. J Cell Sci 116:1175–1186.
Dugo L, Collin M, Allen DA, Murch O, Foster SJ, Yaqoob MM, and Thiemermann C
(2006) Insulin reduces the multiple organ injury and dysfunction caused by coad-
ministration of lipopolysaccharide and peptidoglycan independently of blood glu-
cose: role of glycogen synthase kinase-3beta inhibition. Crit Care Med 34:1489
1496.
During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, Bland RJ, Klug-
mann M, Banks WA, Drucker DJ, et al. (2003) Glucagon-like peptide-1 receptor is
involved in learning and neuroprotection. Nat Med 9:1173–1179.
Eltahir EM, El Ghazali G, A-Elgadir TM, A-Elbasit IE, Elbashir MI, and Giha HA
(2010a) Raised plasma insulin level and homeostasis model assessment (HOMA)
score in cerebral malaria: evidence for insulin resistance and marker of virulence.
Acta Biochim Pol 57:513–520.
Eltahir EM, El Ghazali G, A-Elgadir TM, A-Elbasit IE, Elbashir MI, and Giha HA
(2010b) Raised plasma insulin level and homeostasis model assessment (HOMA)
score in cerebral malaria: evidence for insulin resistance and marker of virulence.
Acta Biochim Pol 57:513–520.
Embi N, Rylatt DB, and Cohen P (1980) Glycogen synthase kinase-3 from rabbit
skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phos-
phorylase kinase. Eur J Biochem 107:519–527.
Engel T, Herna´ ndez F, Avila J, and Lucas JJ (2006) Full reversal of Alzheimer’s
disease-like phenotype in a mouse model with conditional overexpression of gly-
cogen synthase kinase-3. J Neurosci 26:5083–5090.
Engelhart MJ, Geerlings MI, Meijer J, Kiliaan A, Ruitenberg A, van Swieten JC,
Stijnen T, Hofman A, Witteman JC, and Breteler MM (2004) Inflammatory pro-
teins in plasma and the risk of dementia: the Rotterdam study. Arch Neurol
61:668–672.
Extance A (2010) Alzheimer’s failure raises questions about disease-modifying strat-
egies. Nat Rev Drug Discov 9:749–751.
Familian A, Boshuizen RS, Eikelenboom P, and Veerhuis R (2006) Inhibitory effect
of minocycline on amyloid beta fibril formation and human microglial activation.
Glia 53:233–240.
Fan R, Xu F, Previti ML, Davis J, Grande AM, Robinson JK, and Van Nostrand WE
(2007) Minocycline reduces microglial activation and improves behavioral deficits
in a transgenic model of cerebral microvascular amyloid. J Neurosci 27:3057–3063.
Fassbender K, Walter S, Ku¨ hl S, Landmann R, Ishii K, Bertsch T, Stalder AK,
Muehlhauser F, Liu Y, Ulmer AJ, et al. (2004) The LPS receptor (CD14) links
innate immunity with Alzheimer’s disease. FASEB J 18:203–205.
Feinstein R, Kanety H, Papa MZ, Lunenfeld B, and Karasik A (1993) Tumor necrosis
factor-alpha suppresses insulin-induced tyrosine phosphorylation of insulin recep-
tor and its substrates. J Biol Chem 268:26055–26058.
Ferdaoussi M, Abdelli S, Yang JY, Cornu M, Niederhauser G, Favre D, Widmann C,
Regazzi R, Thorens B, Waeber G, et al. (2008) Exendin-4 protects beta-cells from
interleukin-1 beta-induced apoptosis by interfering with the c-Jun NH2-terminal
kinase pathway. Diabetes 57:1205–1215.
Ferraz-Amaro I, Arce-Franco M, Mun˜izJ,Lo´pez-Ferna´ ndez J, Herna´ ndez-
Herna´ ndez V, Franco A, Quevedo J, Martínez-Martín J, and Díaz-Gonza´ lez F
(2011) Systemic blockade of TNF-alpha does not improve insulin resistance in
humans. Horm Metab Res 43:801–808.
Ferreira RA, Vieira CS, Rosa-E-Silva JC, Rosa-e-Silva AC, Nogueira AA, and Fer-
riani RA (2010) Effects of the levonorgestrel-releasing intrauterine system on
cardiovascular risk markers in patients with endometriosis: a comparative study
with the GnRH analogue. Contraception 81:117–122.
Ficicioglu C, Kumbak B, Akcin O, Attar R, Yildirim G, and Yesildaglar N (2010)
Comparison of follicular fluid and serum cytokine concentrations in women un-
dergoing assisted reproductive treatment with GnRH agonist long and antagonist
protocols. Gynecol Endocrinol 26:181–186.
Fink M (1997) Cytopathic hypoxia in sepsis. Acta Anaesthesiol Scand Suppl 110:87–95.
Fink MP (2000) Cytopathic hypoxia. A concept to explain organ dysfunction in sepsis.
Minerva Anestesiol 66:337–342.
Fink MP (2001) Cytopathic hypoxia. Mitochondrial dysfunction as mechanism con-
tributing to organ dysfunction in sepsis. Crit Care Clin 17:219–237.
Forsythe ME, Love DC, Lazarus BD, Kim EJ, Prinz WA, Ashwell G, Krause MW, and
Hanover JA (2006) Caenorhabditis elegans ortholog of a diabetes susceptibility
locus: oga-1 (O-GlcNAcase) knockout impacts O-GlcNAc cycling, metabolism, and
dauer. Proc Natl Acad Sci USA 103:11952–11957.
Fraker DL, Merino MJ, and Norton JA (1989) Reversal of the toxic effects of
cachectin by concurrent insulin administration. Am J Physiol 256:E725–E731.
Frankel SK, Cosgrove GP, Cha SI, Cool CD, Wynes MW, Edelman BL, Brown KK,
and Riches DW (2006) TNF-alpha sensitizes normal and fibrotic human lung
fibroblasts to Fas-induced apoptosis. Am J Respir Cell Mol Biol 34:293–304.
Frayn KN (1975) Effects of burn injury on insulin secretion and on sensitivity to
insulin in the rat in vivo. Eur J Clin Invest 5:331–337.
Frye CA, Duffy CK, and Walf AA (2007) Estrogens and progestins enhance spatial
learning of intact and ovariectomized rats in the object placement task. Neurobiol
Learn Mem 88:208–216.
Fujikawa T, Chuang JC, Sakata I, Ramadori G, and Coppari R (2010) Leptin therapy
improves insulin-deficient type 1 diabetes by CNS-dependent mechanisms in mice.
Proc Natl Acad Sci USA 107:17391–17396.
Games D, Buttini M, Kobayashi D, Schenk D, and Seubert P (2006) Mice as models:
transgenic approaches and Alzheimer’s disease. J Alzheimers Dis 9:133–149.
Garrabou G, More´ n C, Lo´ pez S, Tobías E, Cardellach F, Miro´ O, and Casademont J
(2012) The effects of sepsis on mitochondria. J Infect Dis 205:392–400.
Garwood CJ, Cooper JD, Hanger DP, and Noble W (2010) Anti-inflammatory impact
of minocycline in a mouse model of tauopathy. Front Psychiatry 1:136.
Garwood CJ, Pooler AM, Atherton J, Hanger DP, and Noble W (2011) Astrocytes are
important mediators of Abeta-induced neurotoxicity and tau phosphorylation in
primary culture. Cell Death Dis 2:e167.
Gault VA, Porter WD, Flatt PR, and Holscher C (2010) Actions of exendin-4 therapy
on cognitive function and hippocampal synaptic plasticity in mice fed a high-fat
diet. Int J Obes (Lond)34:1341–1344.
Ge YW and Lahiri DK (2002) Regulation of promoter activity of the APP gene by
cytokines and growth factors: implications in Alzheimer’s disease. Ann NY Acad
Sci 973:463–467.
German JP, Wisse BE, Thaler JP, Oh-I S, Sarruf DA, Ogimoto K, Kaiyala KJ,
Fischer JD, Matsen ME, Taborsky GJ Jr, et al. (2010) Leptin deficiency causes
insulin resistance induced by uncontrolled diabetes. Diabetes 59:1626–1634.
Giannessi F, Giambelluca MA, Grasso L, Scavuzzo MC, and Ruffoli R (2008) Cur-
cumin protects Leydig cells of mice from damage induced by chronic alcohol
administration. Med Sci Monit 14:BR237–BR242.
Giroir BP, Horton JW, White DJ, McIntyre KL, and Lin CQ (1994) Inhibition of
tumor necrosis factor prevents myocardial dysfunction during burn shock. Am J
Physiol 267:H118–H124.
Goedert M (2004) Tau protein and neurodegeneration. Semin Cell Dev Biol 15:45–49.
Goldgaber D, Harris HW, Hla T, Maciag T, Donnelly RJ, Jacobsen JS, Vitek MP, and
Gajdusek DC (1989) Interleukin 1 regulates synthesis of amyloid beta-protein
precursor mRNA in human endothelial cells. Proc Natl Acad Sci USA 86:7606
76010.
Golovliov I, Kuoppa K, Sjostedt A, Tarnvik A, and Sandstrom G (1996)Cytokine
expression in the liver of mice infected with a highly virulent strain of Francisella
tularensis. FEMS Immunol Med Microbiol 13:239–244.
Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua C, Sanchez-
Andrade A, Martin J, and Llorca J (2006) Anti-tumor necrosis factor-alpha block-
ade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp
Rheumatol 24:83–86.
Gonzalez-Gay MA, Gonzalez-Juanatey C, Vazquez-Rodriguez TR, Miranda-Filloy
JA, and Llorca J (2010) Insulin resistance in rheumatoid arthritis: the impact of
the anti-TNF-alpha therapy. Ann NY Acad Sci 1193:153–159.
Gonza´ lez-Gay MA, Gonza´ lez-Juanatey C, Miranda-Filloy JA, and Llorca J (2012)
The potential effect of TNF-alpha antagonist therapy in rheumatoid arthritis may
depend on the degree and severity of insulin resistance before the onset of this
therapy. Horm Metab Res 44:558–559.
Goren I, Mu¨ ller E, Pfeilschifter J, and Frank S (2006) Severely impaired insulin
RCLARK ET AL.
signaling in chronic wounds of diabetic ob/ob mice: a potential role of tumor
necrosis factor-alpha. Am J Pathol 168:765–777.
Gorlovoy P, Larionov S, Pham TT, and Neumann H (2009) Accumulation of tau
induced in neurites by microglial proinflammatory mediators. FASEB J 23:2502–
2513.
Go¨ tz J, Ittner A, and Ittner LM (2012) Tau-targeted treatment strategies in Alzhei-
mer’s disease. Br J Pharmacol 165:1246–1259.
Greco SJ, Sarkar S, Casadesus G, Zhu X, Smith MA, Ashford JW, Johnston JM, and
Tezapsidis N (2009) Leptin inhibits glycogen synthase kinase-3beta to prevent tau
phosphorylation in neuronal cells. Neurosci Lett 455:191–194.
Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, Zavitz KH,
and Tarenflurbil Phase 3 Study Group (2009) Effect of tarenflurbil on cognitive
decline and activities of daily living in patients with mild Alzheimer disease: a
randomized controlled trial. JAMA 302:2557–2564.
Greig NH, Giordano T, Zhu X, Yu QS, Perry TA, Holloway HW, Brossi A, Rogers JT,
Sambamurti K, and Lahiri DK (2004a) Thalidomide-based TNF-alpha inhibitors
for neurodegenerative diseases. Acta Neurobiol Exp (Wars)64:1–9.
Greig NH, Mattson MP, Perry T, Chan SL, Giordano T, Sambamurti K, Rogers JT,
Ovadia H, and Lahiri DK (2004b) New therapeutic strategies and drug candidates
for neurodegenerative diseases: p53 and TNF-alpha inhibitors, and GLP-1 recep-
tor agonists. Ann NY Acad Sci 1035:290–315.
Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ, White CL 3rd, and
Araoz C (1989) Brain interleukin 1 and S-100 immunoreactivity are elevated in
Down syndrome and Alzheimer disease. Proc Natl Acad Sci USA 86:7611–7615.
Gump FE, Long C, Killian P, and Kinney JM (1974) Studies of glucose intolerance in
septic injured patients. J Trauma 14:378–388.
Hamilton A, Patterson S, Porter D, Gault VA, and Holscher C (2011) Novel GLP-1
mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in
the brain. J Neurosci Res 89:481–489.
Hand CC and Brines M (2011) Promises and pitfalls in erythopoietin-mediated
tissue protection: are nonerythropoietic derivatives a way forward? J Investig Med
59:1073–1082.
Hansen FM, Nilsson P, Sonne O, Hustvedt BE, Nilsson-Ehle P, Nielsen JH, and Løvø
A (1983) Variations in insulin responsiveness in rat fat cells are due to metabolic
differences rather than insulin binding. Diabetologia 24:131–135.
Hardardo´ ttir I, Kunitake ST, Moser AH, Doerrler WT, Rapp JH, Gru¨ nfeld C, and
Feingold KR (1994) Endotoxin and cytokines increase hepatic messenger RNA
levels and serum concentrations of apolipoprotein J (clusterin) in Syrian hamsters.
J Clin Invest 94:1304–1309.
Hardy J and Selkoe DJ (2002) The amyloid hypothesis of Alzheimer’s disease:
progress and problems on the road to therapeutics. Science 297:353–356.
Harmsen MM and De Haard HJ (2007) Properties, production, and applications of
camelid single-domain antibody fragments. Appl Microbiol Biotechnol 77:13–22.
Hart BL (1988) Biological basis of the behavior of sick animals. Neurosci Biobehav
Rev 12:123–137.
Ha¨ rtig W, Stieler J, Boerema AS, Wolf J, Schmidt U, Weissfuss J, Bullmann T,
Strijkstra AM, and Arendt T (2007) Hibernation model of tau phosphorylation in
hamsters: selective vulnerability of cholinergic basal forebrain neurons - implica-
tions for Alzheimer’s disease. Eur J Neurosci 25:69–80.
Hauptmann S, Scherping I, Dro¨ se S, Brandt U, Schulz KL, Jendrach M, Leuner K,
Eckert A, and Mu¨ ller WE (2009) Mitochondrial dysfunction: an early event in
Alzheimer pathology accumulates with age in AD transgenic mice. Neurobiol
Aging 30:1574–1586.
Havrankova J, Roth J, and Brownstein M (1978) Insulin receptors are widely
distributed in the central nervous system of the rat. Nature 272:827–829.
He J, Evans CO, Hoffman SW, Oyesiku NM, and Stein DG (2004) Progesterone and
allopregnanolone reduce inflammatory cytokines after traumatic brain injury. Exp
Neurol 189:404–412.
Herna´ ndez-Caselles T and Stutman O (1993) Immune functions of tumor necrosis
factor. I. Tumor necrosis factor induces apoptosis of mouse thymocytes and can
also stimulate or inhibit IL-6-induced proliferation depending on the concentration
of mitogenic costimulation. J Immunol 151:3999–4012.
Herold KC, Vezys V, Sun Q, Viktora D, Seung E, Reiner S, and Brown DR (1996)
Regulation of cytokine production during development of autoimmune diabetes
induced with multiple low doses of streptozotocin. J Immunol 156:3521–3527.
Hicks SD and Miller MW (2011) Effects of ethanol on transforming growth factor
B1-dependent and -independent mechanisms of neural stem cell apoptosis. Exp
Neurol 229:372–380.
Hirota T, Lewis WG, Liu AC, Lee JW, Schultz PG, and Kay SA (2008) A chemical
biology approach reveals period shortening of the mammalian circadian clock by
specific inhibition of GSK-3beta. Proc Natl Acad Sci USA 105:20746–20751.
Hoane MR, Kaufman N, Vitek MP, and McKenna SE (2009) COG1410 improves
cognitive performance and reduces cortical neuronal loss in the traumatically
injured brain. J Neurotrauma 26:121–129.
Hoeks J, van Herpen NA, Mensink M, Moonen-Kornips E, van Beurden D, Hesselink
MK, and Schrauwen P (2010) Prolonged fasting identifies skeletal muscle mito-
chondrial dysfunction as consequence rather than cause of human insulin resis-
tance. Diabetes 59:2117–2125.
Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A, Jones RW,
Bullock R, Love S, Neal JW, et al. (2008) Long-term effects of Abeta42 immuni-
sation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase
I trial. Lancet 372:216–223.
Ho¨ lscher C and Li L (2010) New roles for insulin-like hormones in neuronal signal-
ling and protection: new hopes for novel treatments of Alzheimer’s disease? Neu-
robiol Aging 31:1495–1502.
Holst JJ, Burcelin R, and Nathanson E (2011) Neuroprotective properties of GLP-1:
theoretical and practical applications. Curr Med Res Opin 27:547–558.
Holstad M and Sandler S (2001) A transcriptional inhibitor of TNF-alpha prevents
diabetes induced by multiple low-dose streptozotocin injections in mice. J Auto-
immun 16:441–447.
Hong M and Lee VM (1997) Insulin and insulin-like growth factor-1 regulate tau
phosphorylation in cultured human neurons. J Biol Chem 272:19547–19553.
Honjo H, Ogino Y, Naitoh K, Urabe M, Kitawaki J, Yasuda J, Yamamoto T, Ishihara
S, Okada H, and Yonezawa T (1989) In vivo effects by estrone sulfate on the central
nervous system-senile dementia (Alzheimer’s type). J Steroid Biochem 34:521–
525.
Hooper C, Killick R, and Lovestone S (2008) The GSK3 hypothesis of Alzheimer’s
disease. J Neurochem 104:1433–1439.
Hooper C, Markevich V, Plattner F, Killick R, Schofield E, Engel T, Hernandez F,
Anderton B, Rosenblum K, Bliss T, et al. (2007) Glycogen synthase kinase-3
inhibition is integral to long-term potentiation. Eur J Neurosci 25:81–86.
Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, and Spiegelman BM
(1996) IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in
TNF-alpha- and obesity-induced insulin resistance. Science 271:665–668.
Hotamisligil GS, Shargill NS, and Spiegelman BM (1993) Adipose expression of
tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Sci-
ence 259:87–91.
Hoyer S, Lee SK, Lo¨ ffler T, and Schliebs R (2000) Inhibition of the neuronal insulin
receptor. An in vivo model for sporadic Alzheimer disease? Ann NY Acad Sci
920:256–258.
Hoyer S, Mu¨ ller D, and Plaschke K (1994) Desensitization of brain insulin receptor.
Effect on glucose/energy and related metabolism. J Neural Transm Suppl 44:259
268.
Hsu SM, Chen YC, and Jiang MC (2000) 17 beta-estradiol inhibits tumor necrosis
factor-alpha-induced nuclear factor-kappa B activation by increasing nuclear fac-
tor-kappa B p105 level in MCF-7 breast cancer cells. Biochem Biophys Res Com-
mun 279:47–52.
Hu S, Begum AN, Jones MR, Oh MS, Beech WK, Beech BH, Yang F, Chen P, Ubeda
OJ, Kim PC, et al. (2009) GSK3 inhibitors show benefits in an Alzheimer’s disease
(AD) model of neurodegeneration but adverse effects in control animals. Neurobiol
Dis 33:193–206.
Huang X, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JD, Hanson GR,
Stokes KC, Leopold M, Multhaup G, Goldstein LE, et al. (1999) Cu(II) potentiation
of Alzheimer Abeta neurotoxicity. Correlation with cell-free hydrogen peroxide
production and metal reduction. J Biol Chem 274:37111–37116.
Hung LW, Ciccotosto GD, Giannakis E, Tew DJ, Perez K, Masters CL, Cappai R,
Wade JD, and Barnham KJ (2008) Amyloid-beta peptide (Abeta) neurotoxicity is
modulated by the rate of peptide aggregation: Abeta dimers and trimers correlate
with neurotoxicity. J Neurosci 28:11950–11958.
Ichiyama T, Hayashi T, and Furukawa S (1996) Cerebrospinal fluid concentrations
of soluble tumor necrosis factor receptor in bacterial and aseptic meningitis.
Neurology 46:837–838.
Idro R, Kakooza-Mwesige A, Balyejjussa S, Mirembe G, Mugasha C, Tugumisirize J,
and Byarugaba J (2010) Severe neurological sequelae and behaviour problems
after cerebral malaria in Ugandan children. BMC Res Notes 3:104.
Iitaka C, Miyazaki K, Akaike T, and Ishida N (2005) A role for glycogen synthase
kinase-3beta in the mammalian circadian clock. J Biol Chem 280:29397–29402.
Iqbal J, Sun L, Kumar TR, Blair HC, and Zaidi M (2006) Follicle-stimulating
hormone stimulates TNF production from immune cells to enhance osteoblast and
osteoclast formation. Proc Natl Acad Sci USA 103:14925–14930.
Irving EA, Nicoll J, Graham DI, and Dewar D (1996) Increased tau immunoreactiv-
ity in oligodendrocytes following human stroke and head injury. Neurosci Lett
213:189–192.
Isik AT, Celik T, Ulusoy G, Ongoru O, Elibol B, Doruk H, Bozoglu E, Kayir H, Mas
MR, and Akman S (2009) Curcumin ameliorates impaired insulin/IGF signalling
and memory deficit in a streptozotocin-treated rat model. Age 31:39–49.
Islander U, Jochems C, Lagerquist MK, Forsblad-d’Elia H, and Carlsten H (2011)
Estrogens in rheumatoid arthritis; the immune system and bone. Mol Cell Endo-
crinol 335:14–29.
Jana M, Palencia CA, and Pahan K (2008) Fibrillar amyloid-beta peptides activate
microglia via TLR2: implications for Alzheimer’s disease. J Immunol 181:7254
7262.
Jiang C, Wang J, Li X, Liu C, Chen N, and Hao Y (2009) Progesterone exerts
neuroprotective effects by inhibiting inflammatory response after stroke. Inflamm
Res 58:619–624.
John CC, Bangirana P, Byarugaba J, Opoka RO, Idro R, Jurek AM, Wu B, and Boivin
MJ (2008a) Cerebral malaria in children is associated with long-term cognitive
impairment. Pediatrics 122:e92–e99.
John CC, Kutamba E, Mugarura K, and Opoka RO (2010) Adjunctive therapy for
cerebral malaria and other severe forms of Plasmodium falciparum malaria.
Expert Rev Anti Infect Ther 8:997–1008.
John CC, Panoskaltsis-Mortari A, Opoka RO, Park GS, Orchard PJ, Jurek AM, Idro
R, Byarugaba J, and Boivin MJ (2008b) Cerebrospinal fluid cytokine levels and
cognitive impairment in cerebral malaria. Am J Trop Med Hyg 78:198–205.
Jolivalt CG, Lee CA, Beiswenger KK, Smith JL, Orlov M, Torrance MA, and Masliah
E (2008) Defective insulin signaling pathway and increased glycogen synthase
kinase-3 activity in the brain of diabetic mice: parallels with Alzheimer’s disease
and correction by insulin. J Neurosci Res 86:3265–3274.
Jope RS and Johnson GV (2004) The glamour and gloom of glycogen synthase
kinase-3. Trends Biochem Sci 29:95–102.
Junod A, Lambert AE, Orci L, Pictet R, Gonet AE, and Renold AE (1967) Studies of
the diabetogenic action of streptozotocin. Proc Soc Exp Biol Med 126:201–205.
Kaiser K, Texier A, Ferrandiz J, Buguet A, Meiller A, Latour C, Peyron F, Cespuglio
R, and Picot S (2006) Recombinant human erythropoietin prevents the death of
mice during cerebral malaria. J Infect Dis 193:987–995.
Kaneko I, Yamada N, Sakuraba Y, Kamenosono M, and Tutumi S (1995) Suppres-
sion of mitochondrial succinate dehydrogenase, a primary target of beta-amyloid,
and its derivative racemized at Ser residue. J Neurochem 65:2585–2593.
Kanik KS and Wilder RL (2000) Hormonal alterations in rheumatoid arthritis,
including the effects of pregnancy. Rheum Dis Clin North Am 26:805–823.
Karatsoreos IN, Butler MP, Lesauter J, and Silver R (2011) Androgens modulate
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD S
structure and function of the suprachiasmatic nucleus brain clock. Endocrinology
152:1970–1978.
Karran E, Mercken M, and De Strooper B (2011) The amyloid cascade hypothesis for
Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev
Drug Discov 10:698–712.
Kåss AS, Lea TE, Torjesen PA, Gulseth HC, and Førre ØT (2010) The association of
luteinizing hormone and follicle-stimulating hormone with cytokines and markers
of disease activity in rheumatoid arthritis: a case-control study. Scand J Rheuma-
tol 39:109–117.
Kaufman NA, Beare JE, Tan AA, Vitek MP, McKenna SE, and Hoane MR (2010)
COG1410, an apolipoprotein E-based peptide, improves cognitive performance and
reduces cortical loss following moderate fluid percussion injury in the rat. Behav
Brain Res 214:395–401.
Kaushal V and Schlichter LC (2008) Mechanisms of microglia-mediated neurotoxic-
ity in a new model of the stroke penumbra. J Neurosci 28:2221–2230.
Kayal RA, Alblowi J, McKenzie E, Krothapalli N, Silkman L, Gerstenfeld L, Einhorn
TA, and Graves DT (2009) Diabetes causes the accelerated loss of cartilage during
fracture repair which is reversed by insulin treatment. Bone 44:357–363.
Kern W, Born J, Schreiber H, and Fehm HL (1999) Central nervous system effects
of intranasally administered insulin during euglycemia in men. Diabetes 48:557–
563.
Ketterer C, Tschritter O, Preissl H, Heni M, Ha¨ ring HU, and Fritsche A (2011)
Insulin sensitivity of the human brain. Diabetes Res Clin Pract 93 (Suppl 1):S47–
S51.
Khan KN, Kitajima M, Hiraki K, Fujishita A, Sekine I, Ishimaru T, and Masuzaki H
(2010) Changes in tissue inflammation, angiogenesis and apoptosis in endometri-
osis, adenomyosis and uterine myoma after GnRH agonist therapy. Hum Reprod
25:642–653.
Kihara M, Carter JA, Holding PA, Vargha-Khadem F, Scott RC, Idro R, Fegan GW,
de Haan M, Neville BG, and Newton CR (2009) Impaired everyday memory
associated with encephalopathy of severe malaria: the role of seizures and hip-
pocampal damage. Malar J 8:273.
Kim YJ and Jung YW (2010) Systemic injection of recombinant human erythropoi-
etin after focal cerebral ischemia enhances oligodendroglial and endothelial pro-
genitor cells in rat brain. Anat Cell Biol 43:140–149.
Kimiwada T, Sakurai M, Ohashi H, Aoki S, Tominaga T, and Wada K (2009) Clock
genes regulate neurogenic transcription factors, including NeuroD1, and the neu-
ronal differentiation of adult neural stem/progenitor cells. Neurochem Int 54:277–
285.
Kimura T, Yamashita S, Nakao S, Park JM, Murayama M, Mizoroki T, Yoshiike Y,
Sahara N, and Takashima A (2008) GSK-3beta is required for memory reconsoli-
dation in adult brain. PLoS One 3:e3540.
Kiortsis DN, Mavridis AK, Vasakos S, Nikas SN, and Drosos AA (2005) Effects of
infliximab treatment on insulin resistance in patients with rheumatoid arthritis
and ankylosing spondylitis. Ann Rheum Dis 64:765–766.
Kipp M, Karakaya S, Johann S, Kampmann E, Mey J, and Beyer C (2007) Oestrogen
and progesterone reduce lipopolysaccharide-induced expression of tumour necrosis
factor-alpha and interleukin-18 in midbrain astrocytes. J Neuroendocrinol 19:
819–822.
Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, Cribbs DH,
and LaFerla FM (2011) Blocking IL-1 signaling rescues cognition, attenuates tau
pathology, and restores neuronal beta-catenin pathway function in an Alzheimer’s
disease model. J Immunol 187:6539–6549.
Knauf C, Cani PD, Ait-Belgnaoui A, Benani A, Dray C, Cabou C, Colom A, Uldry M,
Rastrelli S, Sabatier E, et al. (2008) Brain glucagon-like peptide 1 signaling
controls the onset of high-fat diet-induced insulin resistance and reduces energy
expenditure. Endocrinology 149:4768–4777.
Ko HW, Kim EY, Chiu J, Vanselow JT, Kramer A, and Edery I (2010) A hierarchical
phosphorylation cascade that regulates the timing of PERIOD nuclear entry
reveals novel roles for proline-directed kinases and GSK-3beta/SGG in circadian
clocks. J Neurosci 30:12664–12675.
Koch C, Augustine RA, Steger J, Ganjam GK, Benzler J, Pracht C, Lowe C, Schwartz
MW, Shepherd PR, Anderson GM, et al. (2010) Leptin rapidly improves glucose
homeostasis in obese mice by increasing hypothalamic insulin sensitivity. J Neu-
rosci 30:16180–16187.
Kohsaka A and Bass J (2007) A sense of time: how molecular clocks organize
metabolism. Trends Endocrinol Metab 18:4–11.
Koopmans SJ, Mroz Z, Dekker R, Corbijn H, Ackermans M, and Sauerwein H (2006)
Association of insulin resistance with hyperglycemia in streptozotocin-diabetic
pigs: effects of metformin at isoenergetic feeding in a type 2-like diabetic pig model.
Metabolism 55:960–971.
Kozikowski AP, Gunosewoyo H, Guo S, Gaisina IN, Walter RL, Ketcherside A,
McClung CA, Mesecar AD, and Caldarone B (2011) Identification of a glycogen
synthase kinase-3beta inhibitor that attenuates hyperactivity in CLOCK mutant
mice. ChemMedChem 6:1593–1602.
Kristensson K, Nygård M, Bertini G, and Bentivoglio M (2010) African trypanosome
infections of the nervous system: parasite entry and effects on sleep and synaptic
functions. Prog Neurobiol 91:152–171.
Kruman II, Nath A, Maragos WF, Chan SL, Jones M, Rangnekar VM, Jakel RJ, and
Mattson MP (1999) Evidence that Par-4 participates in the pathogenesis of HIV
encephalitis. Am J Pathol 155:39–46.
Kuriyama K, Sasahara K, Kudo T, and Shibata S (2004) Daily injection of insulin
attenuated impairment of liver circadian clock oscillation in the streptozotocin-
treated diabetic mouse. FEBS Lett 572:206–210.
Lang CH and Dobrescu C (1989) In vivo insulin resistance during nonlethal hyper-
metabolic sepsis. Circ Shock 28:165–178.
Lang CH, Dobrescu C, and Bagby GJ (1992) Tumor necrosis factor impairs insulin
action on peripheral glucose disposal and hepatic glucose output. Endocrinology
130:43–52.
Laskowitz DT, Goel S, Bennett ER, and Matthew WD (1997) Apolipoprotein E
suppresses glial cell secretion of TNF-alpha. J Neuroimmunol 76:70–74.
Laskowitz DT, McKenna SE, Song P, Wang H, Durham L, Yeung N, Christensen D,
and Vitek MP (2007) COG1410, a novel apolipoprotein E-based peptide, improves
functional recovery in a murine model of traumatic brain injury. J Neurotrauma
24:1093–1107.
Laskowitz DT, Thekdi AD, Thekdi SD, Han SK, Myers JK, Pizzo SV, and Bennett ER
(2001) Downregulation of microglial activation by apolipoprotein E and apoE-
mimetic peptides. Exp Neurol 167:74–85.
Laurin D, David Curb J, Masaki KH, White LR, and Launer LJ (2009) Midlife
C-reactive protein and risk of cognitive decline: a 31-year follow-up. Neurobiol
Aging 30:1724–1727.
Leconte C, Bihel E, Lepelletier FX, Boue¨ t V, Saulnier R, Petit E, Boulouard M,
Bernaudin M, and Schumann-Bard P (2011) Comparison of the effects of erythro-
poietin and its carbamylated derivative on behaviour and hippocampal neurogen-
esis in mice. Neuropharmacology 60:354–364.
Lee SM, Yune TY, Kim SJ, Kim YC, Oh YJ, Markelonis GJ, and Oh TH (2004)
Minocycline inhibits apoptotic cell death via attenuation of TNF-alpha expression
following iNOS/NO induction by lipopolysaccharide in neuron/glia co-cultures.
J Neurochem 91:568–578.
Lei P, Ayton S, Bush AI, and Adlard PA (2011) GSK-3 in neurodegenerative diseases.
Int J Alzheimers Dis 2011:189246.
LeRoith D, Lesniak MA, and Roth J (1981a) Insulin in insects and annelids. Diabetes
30:70–76.
LeRoith D, Shiloach J, Roth J, and Lesniak MA (1981b) Insulin or a closely related
molecule is native to Escherichia coli. J Biol Chem 256:6533–6536.
Lester-Coll N, Rivera EJ, Soscia SJ, Doiron K, Wands JR, and de la Monte SM (2006)
Intracerebral streptozotocin model of type 3 diabetes: relevance to sporadic Alz-
heimer’s disease. J Alzheimers Dis 9:13–33.
Ley EJ, Srour MK, Clond MA, Barnajian M, Tillou A, Mirocha J, and Salim A (2011)
Diabetic patients with traumatic brain injury: insulin deficiency is associated with
increased mortality. J Trauma 70:1141–1144.
Li G, Barrett EJ, Barrett MO, Cao W, and Liu Z (2007) Tumor necrosis factor-alpha
induces insulin resistance in endothelial cells via a p38 mitogen-activated protein
kinase-dependent pathway. Endocrinology 148:3356–3363.
Li L, El-Kholy W, Rhodes CJ, and Brubaker PL (2005) Glucagon-like peptide-1
protects beta cells from cytokine-induced apoptosis and necrosis: role of protein
kinase B. Diabetologia 48:1339–1349.
Li X and Jope RS (2010) Is glycogen synthase kinase-3 a central modulator in mood
regulation? Neuropsychopharmacology 35:2143–2154.
Li Y, Duffy KB, Ottinger MA, Ray B, Bailey JA, Holloway HW, Tweedie D, Perry T,
Mattson MP, Kapogiannis D, et al. (2010) GLP-1 receptor stimulation reduces
amyloid-beta peptide accumulation and cytotoxicity in cellular and animal models
of Alzheimer’s disease. J Alzheimers Dis 19:1205–1219.
Liao YF, Wang BJ, Cheng HT, Kuo LH, and Wolfe MS (2004) Tumor necrosis
factor-alpha, interleukin-1beta, and interferon-gamma stimulate gamma-
secretase-mediated cleavage of amyloid precursor protein through a JNK-
dependent MAPK pathway. J Biol Chem 279:49523–49532.
Lieb W, Beiser AS, Vasan RS, Tan ZS, Au R, Harris TB, Roubenoff R, Auerbach S,
DeCarli C, Wolf PA, et al. (2009) Association of plasma leptin levels with incident
Alzheimer disease and MRI measures of brain aging. JAMA 302:2565–2572.
Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, Giese T, and
Veltkamp R (2009) Regulatory T cells are key cerebroprotective immunomodula-
tors in acute experimental stroke. Nat Med 15:192–199.
Lin CY, Higginbotham DA, Judd RL, and White BD (2002) Central leptin increases
insulin sensitivity in streptozotocin-induced diabetic rats. Am J Physiol Endocri-
nol Metab 282:E1084–E1091.
Lin J, Li X, Yuan F, Lin L, Cook CL, Rao ChV, and Lei Z (2010) Genetic ablation of
luteinizing hormone receptor improves the amyloid pathology in a mouse model of
Alzheimer disease. J Neuropathol Exp Neurol 69:253–261.
Liu H, Dear AE, Knudsen LB, and Simpson RW (2009) A long-acting glucagon-like
peptide-1 analogue attenuates induction of plasminogen activator inhibitor type-1
and vascular adhesion molecules. J Endocrinol 201:59–66.
Liu Y, Liu F, Grundke-Iqbal I, Iqbal K, and Gong CX (2011) Deficient brain insulin
signalling pathway in Alzheimer’s disease and diabetes. J Pathol 225:54–62.
London ED, Berman SM, Chakrapani S, Delibasi T, Monterosso J, Erol HK, Paz-
Filho G, Wong ML, and Licinio J (2011) Short-term plasticity of gray matter
associated with leptin deficiency and replacement. J Clin Endocrinol Metab 96:
E1212–E1220.
Lorenzo M, Ferna´ ndez-Veledo S, Vila-Bedmar R, Garcia-Guerra L, De Alvaro C, and
Nieto-Vazquez I (2008) Insulin resistance induced by tumor necrosis factor-alpha
in myocytes and brown adipocytes. J Anim Sci 86:E94–E104.
Lovestone S and Reynolds CH (1997) The phosphorylation of tau: a critical stage in
neurodevelopment and neurodegenerative processes. Neuroscience 78:309–324.
Lovestone S, Reynolds CH, Latimer D, Davis DR, Anderton BH, Gallo JM, Hanger D,
Mulot S, Marquardt B, and Stabel S (1994) Alzheimer’s disease-like phosphoryla-
tion of the microtubule-associated protein tau by glycogen synthase kinase-3 in
transfected mammalian cells. Curr Biol 4:1077–1086.
Lynch JR, Morgan D, Mance J, Matthew WD, and Laskowitz DT (2001) Apolipopro-
tein E modulates glial activation and the endogenous central nervous system
inflammatory response. J Neuroimmunol 114:107–113.
Lynch JR, Wang H, Mace B, Leinenweber S, Warner DS, Bennett ER, Vitek MP,
McKenna S, and Laskowitz DT (2005) A novel therapeutic derived from apolipo-
protein E reduces brain inflammation and improves outcome after closed head
injury. Exp Neurol 192:109–116.
Maegraith BG (1954) Physiological aspects of protozoan infection. Annu Rev Micro-
biol 8:273–288.
Mandelkow EM, Drewes G, Biernat J, Gustke N, Van Lint J, Vandenheede JR, and
Mandelkow E (1992) Glycogen synthase kinase-3 and the Alzheimer-like state of
microtubule-associated protein tau. FEBS Lett 314:315–321.
Martin M, Rehani K, Jope RS, and Michalek SM (2005) Toll-like receptor-mediated
cytokine production is differentially regulated by glycogen synthase kinase 3. Nat
Immunol 6:777–784.
TCLARK ET AL.
Marwarha G, Dasari B, Prabhakara JP, Schommer J, and Ghribi O (2010)
-Amyloid
regulates leptin expression and tau phosphorylation through the mTORC1 signal-
ing pathway. J Neurochem 115:373–384.
Mastroianni CM, Paoletti F, Massetti AP, Falciano M, and Vullo V (1990) Elevated
levels of tumor necrosis factor (TNF) in the cerebrospinal fluid from patients with
HIV-associated neurological disorders. Acta Neurol Napoli 12:66–67.
Matsuo T, Yamaguchi S, Mitsui S, Emi A, Shimoda F, and Okamura H (2003)
Control mechanism of the circadian clock for timing of cell division in vivo. Science
302:255–259.
Mayordomo L, Marenco JL, Gomez-Mateos J, and Rejon E (2002) Pulmonary miliary
tuberculosis in a patient with anti-TNF-alpha treatment. Scand J Rheumatol
31:44–45.
McAlpine FE, Lee JK, Harms AS, Ruhn KA, Blurton-Jones M, Hong J, Das P, Golde
TE, LaFerla FM, Oddo S, et al. (2009) Inhibition of soluble TNF signaling in a
mouse model of Alzheimer’s disease prevents pre-plaque amyloid-associated neu-
ropathology. Neurobiol Dis 34:163–177.
McCall JL, Tuckey JA, and Parry BR (1992) Serum tumour necrosis factor alpha and
insulin resistance in gastrointestinal cancer. Br J Surg 79:1361–1363.
McClean PL, Parthsarathy V, Faivre E, and Ho¨ lscher C (2011) The diabetes drug
liraglutide prevents degenerative processes in a mouse model of Alzheimer’s
disease. J Neurosci 31:6587–6594.
McConnell SE, Alla J, Wheat E, Romeo RD, McEwen B, and Thornton JE (2012) The
role of testicular hormones and luteinizing hormone in spatial memory in adult
male rats. Horm Behav 61:479–486.
McGeer PL and McGeer EG (1995) The inflammatory response system of brain:
implications for therapy of Alzheimer and other neurodegenerative diseases. Brain
Res Rev 21:195–218.
McNay EC and Recknagel AK (2011) Brain insulin signaling: a key component of
cognitive processes and a potential basis for cognitive impairment in type 2
diabetes. Neurobiol Learn Mem 96:432–442.
Medana IM, Day NP, Hien TT, Mai NT, Bethell D, Phu NH, Farrar J, Esiri MM,
White NJ, and Turner GD (2002) Axonal injury in cerebral malaria. Am J Pathol
160:655–666.
Medana IM, Idro R, and Newton CR (2007) Axonal and astrocyte injury markers in
the cerebrospinal fluid of Kenyan children with severe malaria. J Neurol Sci
258:93–98.
Medana IM, Lindert RB, Wurster U, Hien TT, Day NP, Phu NH, Mai NT, Chuong
LV, Chau TT, Turner GD, et al. (2005) Cerebrospinal fluid levels of markers of
brain parenchymal damage in Vietnamese adults with severe malaria. Trans R
Soc Trop Med Hyg 99:610–617.
Meistrell ME 3rd, Botchkina GI, Wang H, Di Santo E, Cockroft KM, Bloom O,
Vishnubhakat JM, Ghezzi P, and Tracey KJ (1997) Tumor necrosis factor is a brain
damaging cytokine in cerebral ischemia. Shock 8:341–348.
Mela LM, Miller LD, Bacalzo LV Jr, Olofsson K, and White RR 4th (1972) Alterations
of mitochondrial structure and energy-linked functions in hemorrhagic shock and
endotoxemia. Adv Exp Med Biol 33:231–242.
Mengatto CM, Mussano F, Honda Y, Colwell CS, and Nishimura I (2011) Circadian
rhythm and cartilage extracellular matrix genes in osseointegration: a genome-
wide screening of implant failure by vitamin D deficiency. PLoS One 6:e15848.
Mensah-Brown EP, Obineche EN, Galadari S, Chandranath E, Shahin A, Ahmed I,
Patel SM, and Adem A (2005) Streptozotocin-induced diabetic nephropathy in rats:
the role of inflammatory cytokines. Cytokine 31:180–190.
Meresman GF, Bilotas MA, Lombardi E, Tesone M, Sueldo C, and Baran˜ ao RI (2003)
Effect of GnRH analogues on apoptosis and release of interleukin-1beta and
vascular endothelial growth factor in endometrial cell cultures from patients with
endometriosis. Hum Reprod 18:1767–1771.
Moe GW, Marin-Garcia J, Konig A, Goldenthal M, Lu X, and Feng Q (2004) In vivo
TNF-alpha inhibition ameliorates cardiac mitochondrial dysfunction, oxidative
stress, and apoptosis in experimental heart failure. Am J Physiol Heart Circ
Physiol 287:H1813–H1820.
Moriya T, Hiraishi K, Horie N, Mitome M, and Shinohara K (2007) Correlative
association between circadian expression of mousePer2 gene and the proliferation
of the neural stem cells. Neuroscience 146:494–498.
Mowery NT, Gunter OL, Guillamondegui O, Dossett LA, Dortch MJ, Morris JA Jr,
and May AK (2009) Stress insulin resistance is a marker for mortality in traumatic
brain injury. J Trauma 66:145–151.
Mueller L, von Seggern L, Schumacher J, Goumas F, Wilms C, Braun F, and
Broering DC (2010) TNF-alpha similarly induces IL-6 and MCP-1 in fibroblasts
from colorectal liver metastases and normal liver fibroblasts. Biochem Biophys Res
Commun 397:586–591.
Murphy VA and Johanson CE (1985) Adrenergic-induced enhancement of brain
barrier system permeability to small nonelectrolytes: choroid plexus versus cere-
bral capillaries. J Cereb Blood Flow Metab 5:401–412.
Nairz M, Schroll A, Moschen AR, Sonnweber T, Theurl M, Theurl I, Taub N, Jamnig
C, Neurauter D, Huber LA, et al. (2011) Erythropoietin contrastingly affects
bacterial infection and experimental colitis by inhibiting nuclear factor-kappaB-
inducible immune pathways. Immunity 34:61–74.
Nairz M, Sonnweber T, Schroll A, Theurl I, and Weiss G (2012) The pleiotropic
effects of erythropoietin in infection and inflammation. Microbes Infect 14:238
246.
Nakamura TJ, Sellix MT, Kudo T, Nakao N, Yoshimura T, Ebihara S, Colwell CS,
and Block GD (2010) Influence of the estrous cycle on clock gene expression in
reproductive tissues: effects of fluctuating ovarian steroid hormone levels. Steroids
75:203–212.
Nakamura TJ, Sellix MT, Menaker M, and Block GD (2008) Estrogen directly
modulates circadian rhythms of PER2 expression in the uterus. Am J Physiol
Endocrinol Metab 295:E1025–E1031.
Natalicchio A, De Stefano F, Orlando MR, Melchiorre M, Leonardini A, Cignarelli A,
Labarbuta R, Marchetti P, Perrini S, Laviola L, et al. (2010) Exendin-4 prevents
c-Jun N-terminal protein kinase activation by tumor necrosis factor-alpha (TNFal-
pha) and inhibits TNFalpha-induced apoptosis in insulin-secreting cells. Endocri-
nology 151:2019–2029.
Nathan C (1989) Secretory products of macrophages. J Clin Immunol 79:319–326.
Nathoo N, Caris EC, Wiener JA, and Mendel E (2011) History of the vertebral venous
plexus and the significant contributions of Breschet and Batson. Neurosurgery
69:1007–1014.
Navarro JF, Milena FJ, Mora C, Leon C, Claverie F, Flores C, and Garcia J (2005)
Tumor necrosis factor-alpha gene expression in diabetic nephropathy: relationship
with urinary albumin excretion and effect of angiotensin-converting enzyme inhi-
bition. Kidney Int Suppl (99):S98–S102.
Nieto-Vazquez I, Ferna´ ndez-Veledo S, de Alvaro C, Rondinone CM, Valverde AM,
and Lorenzo M (2007) Protein-tyrosine phosphatase 1B-deficient myocytes show
increased insulin sensitivity and protection against tumor necrosis factor-alpha-
induced insulin resistance. Diabetes 56:404–413.
Nihashi T, Inao S, Kajita Y, Kawai T, Sugimoto T, Niwa M, Kabeya R, Hata N,
Hayashi S, and Yoshida J (2001) Expression and distribution of beta amyloid
precursor protein and beta amyloid peptide in reactive astrocytes after transient
middle cerebral artery occlusion. Acta Neurochir (Wien)143:287–295.
Noble W, Garwood C, Stephenson J, Kinsey AM, Hanger DP, and Anderton BH
(2009) Minocycline reduces the development of abnormal tau species in models of
Alzheimer’s disease. FASEB J 23:739–750.
Nolting T, Lindecke A, Koutsilieri E, Maschke M, Husstedt IW, Sopper S, Stu¨ve O,
Hartung HP, Arendt G, and Competence Network HIV/AIDS (2009) Measurement
of soluble inflammatory mediators in cerebrospinal fluid of human immunodefi-
ciency virus-positive patients at distinct stages of infection by solid-phase protein
array. J Neurovirol 15:390–400.
Olgiati P, Politis AM, Papadimitriou GN, De Ronchi D, and Serretti A (2011)
Genetics of late-onset Alzheimer’s disease: update from the alzgene database and
analysis of shared pathways. Int J Alzheimers Dis 2011:832379.
Onishi T, Iwashita H, Uno Y, Kunitomo J, Saitoh M, Kimura E, Fujita H, Uchiyama
N, Kori M, and Takizawa M (2011) A novel glycogen synthase kinase-3 inhibitor
2-methyl-5-(3-{4-[(S)-methylsulfinyl]phenyl}-1-benzofuran-5-yl)-1,3,4-oxadiazole
decreases tau phosphorylation and ameliorates cognitive deficits in a transgenic
model of Alzheimer’s disease. J Neurochem 119:1330–1340.
Ordo´n˜ez P, Moreno M, Alonso A, Llaneza P, Díaz F, and Gonza´ lez C (2008) 17beta-
Estradiol and/or progesterone protect from insulin resistance in STZ-induced
diabetic rats. J Steroid Biochem Mol Biol 111:287–294.
Osredkar D, Sall JW, Bickler PE, and Ferriero DM (2010) Erythropoietin promotes
hippocampal neurogenesis in in vitro models of neonatal stroke. Neurobiol Dis
38:259–265.
Ostensen M, Aune B, and Husby G (1983) Effect of pregnancy and hormonal changes
on the activity of rheumatoid arthritis. Scand J Rheumatol 12:69–72.
Ott V, Benedict C, Schultes B, Born J, and Hallschmid M (2011) Intranasal admin-
istration of insulin to the brain impacts cognitive function and peripheral metab-
olism. Diabetes Obes Metab 14:214–221.
Pajak B, Orzechowska S, Pijet B, Pijet M, Pogorzelska A, Gajkowska B, and Or-
zechowski A (2008) Crossroads of cytokine signaling–the chase to stop muscle
cachexia. J Physiol Pharmacol 59 (Suppl 9):251–264.
Paolino D, Cosco D, Molinaro R, Celia C, and Fresta M (2011) Supramolecular
devices to improve the treatment of brain diseases. Drug Discov Today 16:311–
324.
Pardridge WM (2010) Biologic TNFalpha-inhibitors that cross the human blood-
brain barrier. Bioeng Bugs 1:231–234.
Patel NS, Collino M, Yaqoob MM, and Thiemermann C (2011a) Erythropoietin in the
intensive care unit: beyond treatment of anemia. Ann Intensive Care 1:40.
Patel NS, Nandra KK, Brines M, Collino M, Wong WF, Kapoor A, Benetti E, Goh FY,
Fantozzi R, Cerami A, et al. (2011b) A nonerythropoietic peptide that mimics the
3D structure of erythropoietin reduces organ injury/dysfunction and inflammation
in experimental hemorrhagic shock. Mol Med 17:883–892.
Paz-Filho G, Wong ML, and Licinio J (2010a) Leptin levels and Alzheimer disease.
JAMA 303:1478.
Paz-Filho G, Wong ML, and Licinio J (2010b) The procognitive effects of leptin in the
brain and their clinical implications. Int J Clin Pract 64:1808–1812.
Pekala P, Kawakami M, Vine W, Lane MD, and Cerami A (1983) Studies of insulin
resistance in adipocytes induced by macrophage mediator. J Exp Med 157:1360
1365.
Peng J, Xie L, Stevenson FF, Melov S, Di Monte DA, and Andersen JK (2006)
Nigrostriatal dopaminergic neurodegeneration in the weaver mouse is mediated
via neuroinflammation and alleviated by minocycline administration. J Neurosci
26:11644–11651.
Perry T, Lahiri DK, Sambamurti K, Chen D, Mattson MP, Egan JM, and Greig NH
(2003) Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide
(Abeta) levels and protects hippocampal neurons from death induced by Abeta and
iron. J Neurosci Res 72:603–612.
Perry VH, Cunningham C, and Holmes C (2007) Systemic infections and inflamma-
tion affect chronic neurodegeneration. Nat Rev Immunol 7:161–167.
Petit-Paitel A, Brau F, Cazareth J, and Chabry J (2009) Involvment of cytosolic and
mitochondrial GSK-3beta in mitochondrial dysfunction and neuronal cell death of
MPTP/MPP-treated neurons. PLoS One 4:e5491.
Pickering M, Cumiskey D, and O’Connor JJ (2005) Actions of TNF-alpha on gluta-
matergic synaptic transmission in the central nervous system. Exp Physiol 90:
663–670.
Picot S, Bienvenu AL, Konate S, Sissoko S, Barry A, Diarra E, Bamba K, Djimde´A,
and Doumbo OK (2009) Safety of epoietin beta-quinine drug combination in
children with cerebral malaria in Mali. Malar J 8:169.
Plaschke K and Hoyer S (1993) Action of the diabetogenic drug streptozotocin on
glycolytic and glycogenolytic metabolism in adult rat brain cortex and hippocam-
pus. Int J Dev Neurosci 11:477–483.
Porter DW, Kerr BD, Flatt PR, Holscher C, and Gault VA (2010) Four weeks
administration of Liraglutide improves memory and learning as well as glycaemic
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD U
control in mice with high fat dietary-induced obesity and insulin resistance.
Diabetes Obes Metab 12:891–899.
Qin B, Qiu W, Avramoglu RK, and Adeli K (2007) Tumor necrosis factor-alpha
induces intestinal insulin resistance and stimulates the overproduction of intes-
tinal apolipoprotein B48-containing lipoproteins. Diabetes 56:450–461.
Qin L, He J, Hanes RN, Pluzarev O, Hong JS, and Crews FT (2008) Increased
systemic and brain cytokine production and neuroinflammation by endotoxin
following ethanol treatment. J Neuroinflammation 5:10.
Quan N, Mhlanga JD, Whiteside MB, McCoy AN, Kristensson K, and Herkenham M
(1999) Chronic overexpression of proinflammatory cytokines and histopathology in
the brains of rats infected with Trypanosoma brucei. J Comp Neurol 414:114–130.
Ramirez R, Carracedo J, Nogueras S, Buendia P, Merino A, Can˜ adillas S, Rodríguez
M, Tetta C, Martin-Malo A, and Aljama P (2009) Carbamylated darbepoetin
derivative prevents endothelial progenitor cell damage with no effect on angiogen-
esis. J Mol Cell Cardiol 47:781–788.
Ranges GE, Zlotnik A, Espevik T, Dinarello CA, Cerami A, and Palladino MA Jr
(1988) Tumor necrosis factor alpha/cachectin is a growth factor for thymocytes.
Synergistic interactions with other cytokines. J Exp Med 167:1472–1478.
Rao SC, Li X, Rao ChV, and Magnuson DS (2003) Human chorionic gonadotropin/
luteinizing hormone receptor expression in the adult rat spinal cord. Neurosci Lett
336:135–138.
Ratai EM, Bombardier JP, Joo CG, Annamalai L, Burdo TH, Campbell J, Fell R,
Hakimelahi R, He J, Autissier P, et al. (2010) Proton magnetic resonance spec-
troscopy reveals neuroprotection by oral minocycline in a nonhuman primate
model of accelerated NeuroAIDS. PLoS One 5:e10523.
Raymond RM (1984) Skeletal muscle metabolism and insulin resistance during
endotoxin shock in the dog. Am J Emerg Med 2:45–59.
Raymond RM, McLane MP, Law WR, King NF, and Leutz DW (1988) Myocardial
insulin resistance during acute endotoxin shock in dogs. Diabetes 37:1684–1688.
Rebel VI, Hartnett S, Hill GR, Lazo-Kallanian SB, Ferrara JL, and Sieff CA (1999)
Essential role for the p55 tumor necrosis factor receptor in regulating hematopoi-
esis at a stem cell level. J Exp Med 190:1493–1504.
Rector RS, Thyfault JP, Uptergrove GM, Morris EM, Naples SP, Borengasser SJ,
Mikus CR, Laye MJ, Laughlin MH, Booth FW, et al. (2010) Mitochondrial dys-
function precedes insulin resistance and hepatic steatosis and contributes to the
natural history of non-alcoholic fatty liver disease in an obese rodent model.
J Hepatol 52:727–736.
Redl H, Schlag G, Paul E, Bahrami S, Buurman WA, Strieter RM, Kunkel SL, Davies
J, and Foulkes R (1996) Endogenous modulators of TNF and IL-1 response are
under partial control of TNF in baboon bacteremia. Am J Physiol 271:R1193–
R1198.
Reger MA, Watson GS, Frey WH, 2nd, Baker LD, Cholerton B, Keeling ML, Belongia
DA, Fishel MA, Plymate SR, et al. (2006) Effects of intranasal insulin on cognition
in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging
27:451–458.
Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, Plymate SR,
Cherrier MM, Schellenberg GD, Frey WH 2nd, et al. (2008) Intranasal insulin
administration dose-dependently modulates verbal memory and plasma amyloid-
beta in memory-impaired older adults. J Alzheimers Dis 13:323–331.
Riddell DR, Zhou H, Atchison K, Warwick HK, Atkinson PJ, Jefferson J, Xu L,
Aschmies S, Kirksey Y, Hu Y, et al. (2008) Impact of apolipoprotein E (ApoE)
polymorphism on brain ApoE levels. J Neurosci 28:11445–11453.
Roselaar SE and Daugherty A (1998) Apolipoprotein E-deficient mice have impaired
innate immune responses to Listeria monocytogenes in vivo. J Lipid Res 39:1740
1743.
Rowan MJ, Klyubin I, Wang Q, Hu NW, and Anwyl R (2007) Synaptic memory
mechanisms: Alzheimer’s disease amyloid beta-peptide-induced dysfunction.
Biochem Soc Trans 35:1219–1223.
Ryan EA, O’Sullivan MJ, and Skyler JS (1985) Insulin action during pregnancy.
Studies with the euglycemic clamp technique. Diabetes 34:380–389.
Ryu JK and McLarnon JG (2008) Thalidomide inhibition of perturbed vasculature
and glial-derived tumor necrosis factor-alpha in an animal model of inflamed
Alzheimer’s disease brain. Neurobiol Dis 29:254–266.
Sagara M, Satoh J, Zhu XP, Takahashi K, Fukuzawa M, Muto G, Muto Y, and Toyota
T (1994) Inhibition with N-acetylcysteine of enhanced production of tumor necrosis
factor in streptozotocin-induced diabetic rats. Clin Immunol Immunopathol 71:
333–337.
Salcedo-Tello P, Ortiz-Matamoros A, and Arias C (2011) GSK3 Function in the brain
during development, neuronal plasticity, and neurodegeneration. Int J Alzheimers
Dis 2011:189728.
Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, Raskind M, Sabbagh M,
Honig LS, Doody R, van Dyck CH, et al. (2009) A phase 2 multiple ascending dose
trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology 73:2061–
2070.
Sargin D, Friedrichs H, El-Kordi A, and Ehrenreich H (2010) Erythropoietin as
neuroprotective and neuroregenerative treatment strategy: comprehensive over-
view of 12 years of preclinical and clinical research. Best Pract Res Clin Anaesthe-
siol 24:573–594.
Schio¨ th HB, Frey WH, Brooks SJ, and Benedict C (2012) Insulin to treat Alzheimer’s
disease: just follow your nose? Expert Rev Clin Pharmacol 5:17–20.
Schmidt J, Barthel K, Wrede A, Salajegheh M, Ba¨ hr M, and Dalakas MC (2008)
Interrelation of inflammation and APP in sIBM: IL-1 beta induces accumulation of
beta-amyloid in skeletal muscle. Brain 131:1228–1240.
Schuitemaker A, Dik MG, Veerhuis R, Scheltens P, Schoonenboom NS, Hack CE,
Blankenstein MA, and Jonker C (2009) Inflammatory markers in AD and MCI
patients with different biomarker profiles. Neurobiol Aging 30:1885–1889.
Seabrook TJ, Jiang L, Maier M, and Lemere CA (2006) Minocycline affects microglia
activation, Abeta deposition, and behavior in APP-tg mice. Glia 53:776–782.
Sereno´ L, Coma M, Rodríguez M, Sa´ nchez-Ferrer P, Sa´ nchez MB, Gich I, Agullo´ JM,
Pe´ rez M, Avila J, Guardia-Laguarta C, et al. (2009) A novel GSK-3beta inhibitor
reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol Dis
35:359–367.
Shalaby MR, Waage A, Aarden L, and Espevik T (1989) Endotoxin tumor necrosis
factor-
and interleukin-1 induce interleukin-6 production in vivo. Clin Immunol
Immunopathol 53:488–498.
Shambaugh GE 3rd and Beisel WR (1967) Insulin response during tularemia in man.
Diabetes 16:369–376.
Shanmugam VK, DeMaria DM, and Attinger CE (2011) Lower extremity ulcers in
rheumatoid arthritis: features and response to immunosuppression. Clin Rheu-
matol 30:849–853.
Sharma S, Chopra K, and Kulkarni SK (2007) Effect of insulin and its combination
with resveratrol or curcumin in attenuation of diabetic neuropathic pain: partici-
pation of nitric oxide and TNF-alpha. Phytother Res 21:278–283.
Sheng B, Wang X, Su B, Lee HG, Casadesus G, Perry G, and Zhu X (2012) Impaired
mitochondrial biogenesis contributes to mitochondrial dysfunction in Alzheimer’s
disease. J Neurochem 120:419–429.
Shi JQ, Shen W, Chen J, Wang BR, Zhong LL, Zhu YW, Zhu HQ, Zhang QQ, Zhang
YD, and Xu J (2011) Anti-TNF-alpha reduces amyloid plaques and tau phosphor-
ylation and induces CD11c-positive dendritic-like cell in the APP/PS1 transgenic
mouse brains. Brain Res 1368:239–247.
Simpkins JW, Gatson JW, and Wigginton JG (2009) Commentary on “a roadmap for
the prevention of dementia II. Leon Thal Symposium 2008.” Rationale and recom-
mendations for first evaluating anti-Alzheimer’s disease medications in acute
brain injury patients. Alzheimers Dement 5:143–146.
Siqueira MF, Li J, Chehab L, Desta T, Chino T, Krothpali N, Behl Y, Alikhani M,
Yang J, Braasch C, et al. (2010) Impaired wound healing in mouse models of
diabetes is mediated by TNF-alpha dysregulation and associated with enhanced
activation of forkhead box O1 (FOXO1). Diabetologia 53:378–388.
Smillie KJ and Cousin MA (2011) The role of GSK3 in presynaptic function. Int J
Alzheimers Dis 2011:263673.
Smith DH, Chen XH, Iwata A, and Graham DI (2003) Amyloid beta accumulation in
axons after traumatic brain injury in humans. J Neurosurg 98:1072–1077.
Soetikno V, Sari FR, Veeraveedu PT, Thandavarayan RA, Harima M, Sukumaran V,
Lakshmanan AP, Suzuki K, Kawachi H, and Watanabe K (2011) Curcumin ame-
liorates macrophage infiltration by inhibiting NF-kappaB activation and proin-
flammatory cytokines in streptozotocin induced-diabetic nephropathy. Nutr Metab
(Lond)8:35.
Sølling C (2011) Organ-protective and immunomodulatory effects of erythropoietin -
an update on recent clinical trials. Basic Clin Pharmacol Toxicol 110:113–121.
Sommer G, Kralisch S, Lipfert J, Weise S, Krause K, Jessnitzer B, Lo¨ ssner U, Blu¨ her
M, Stumvoll M, and Fasshauer M (2009) Amyloid precursor protein expression is
induced by tumor necrosis factor alpha in 3T3–L1 adipocytes. J Cell Biochem
108:1418–1422.
Sonoda N, Katabuchi H, Tashiro H, Ohba T, Nishimura R, Minegishi T, and Oka-
mura H (2005) Expression of variant luteinizing hormone/chorionic gonadotropin
receptors and degradation of chorionic gonadotropin in human chorionic villous
macrophages. Placenta 26:298–307.
Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, Hyman B, Burton
MA, Goldstein LE, Duong S, Tanzi RE, et al. (2010) The Alzheimer’s disease-
associated amyloid beta protein is an antimicrobial peptide. PLoS One 5:e9505.
Sperling RA, Jack CR Jr, and Aisen PS (2011) Testing the right target and right drug
at the right stage. Sci Transl Med 3:111cm33.
Stadler J, Bentz BG, Harbrecht BG, Di Silvio M, Curran RD, Billiar TR, Hoffman
RA, and Simmons RL (1992) Tumor necrosis factor-alpha inhibits hepatocyte
mitochondrial respiration. Ann Surg 216:539–546.
Stanley LC, Mrak RE, Woody RC, Perrot LJ, Zhang S, Marshak DR, Nelson SJ, and
Griffin WS (1994) Glial cytokines as neuropathogenic factors in HIV infection:
pathogenic similarities to alzheimer’s disease. J Neuropathol Exp Neurol 53:231–
238.
Stellwagen D, Beattie EC, Seo JY, and Malenka RC (2005) Differential regulation of
AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha.
J Neurosci 25:3219–3228.
Stieler JT, Bullmann T, Kohl F, Tøien Ø, Bru¨ ckner MK, Ha¨ rtig W, Barnes BM, and
Arendt T (2010) The physiological link between metabolic rate depression and tau
phosphorylation in mammalian hibernation. PLoS One 6:e14530.
Strittmatter WJ, Saunders AM, Goedert M, Weisgraber KH, Dong LM, Jakes R,
Huang DY, Pericak-Vance M, Schmechel D, and Roses AD (1994) Isoform-specific
interactions of apolipoprotein E with microtubule-associated protein tau: implica-
tions for Alzheimer disease. Proc Natl Acad Sci USA 91:11183–11186.
Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen
GS, and Roses AD (1993) Apolipoprotein E: high-avidity binding to beta-amyloid
and increased frequency of type 4 allele in late-onset familial Alzheimer disease.
Proc Natl Acad Sci USA 90:1977–1981.
Suk K (2004) Minocycline suppresses hypoxic activation of rodent microglia in
culture. Neurosci Lett 366:167–171.
Tachado SD and Schofield L (1994) Glycosylphosphatidylinositol toxin of Trypano-
soma brucei regulates IL-1 alpha and TNF-alpha expression in macrophages by
protein tyrosine kinase mediated signal transduction. Biochem Biophys Res Com-
mun 205:984–991.
Tahara Y, Otsuka M, Fuse Y, Hirao A, and Shibata S (2011) Refeeding after fasting
elicits insulin-dependent regulation of Per2 and Rev-erbalpha with shifts in the
liver clock. J Biol Rhythms 26:230–240.
Takeuchi H, Iba M, Inoue H, Higuchi M, Takao K, Tsukita K, Karatsu Y, Iwamoto
Y, Miyakawa T, Suhara T, et al. (2011) P301S mutant human tau transgenic mice
manifest early symptoms of human tauopathies with dementia and altered sen-
sorimotor gating. PLoS One 6:e21050.
Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, Fuino RL, Kawaguchi
KR, Samoyedny AJ, Wilson RS, et al. (2012) Demonstrated brain insulin resis-
tance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1
dysregulation, and cognitive decline. J Clin Invest 122:1316–1338.
Tancredi V, D’Arcangelo G, Grassi F, Tarroni P, Palmieri G, Santoni A, and Eusebi
VCLARK ET AL.
F (1992) Tumor necrosis factor alters synaptic transmission in rat hippocampal
slices. Neurosci Lett 146:176–178.
Tarkowski E, Andreasen N, Tarkowski A, and Blennow K (2003) Intrathecal inflam-
mation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychi-
atry 74:1200–1205.
Terrando N, Monaco C, Ma D, Foxwell BM, Feldmann M, and Maze M (2010) Tumor
necrosis factor-alpha triggers a cytokine cascade yielding postoperative cognitive
decline. Proc Natl Acad Sci USA 107:20518–20522.
Thackeray JT, Radziuk J, Harper ME, Suuronen EJ, Ascah KJ, Beanlands RS, and
Dasilva JN (2011) Sympathetic nervous dysregulation in the absence of systolic
left ventricular dysfunction in a rat model of insulin resistance with hyperglyce-
mia. Cardiovasc Diabetol 10:75.
Thambisetty M, Simmons A, Velayudhan L, Hye A, Campbell J, Zhang Y, Wahlund
LO, Westman E, Kinsey A, Gu¨ ntert A, et al. (2010) Association of plasma clusterin
concentration with severity, pathology, and progression in Alzheimer’s disease.
Arch Gen Psychiatry 67:739–748.
Tilley BC, Alarco´ n GS, Heyse SP, Trentham DE, Neuner R, Kaplan DA, Clegg DO,
Leisen JC, Buckley L, Cooper SM, et al. (1995) Minocycline in rheumatoid arthri-
tis. A 48-week, double-blind, placebo-controlled trial. MIRA Trial Group. Ann
Intern Med 122:81–89.
Tissenbaum HA and Ruvkun G (1998) An insulin-like signaling pathway affects both
longevity and reproduction in Caenorhabditis elegans. Genetics 148:703–717.
Tobinick E (2010) Perispinal etanercept: a new therapeutic paradigm in neurology.
Expert Rev Neurother 10:985–1002.
Tobinick E (2011) Rapid improvement of chronic stroke deficits after perispinal
etanercept: three consecutive cases. CNS Drugs 25:145–155.
Tobinick E (2012) Deciphering the physiology underlying the rapid clinical effects of
perispinal etanercept in Alzheimer’s disease. Curr Alzheimer Res 9:99–109.
Tobinick EL and Gross H (2008) Rapid cognitive improvement in Alzheimer’s disease
following perispinal etanercept administration. J Neuroinflammation 5:2.
Tobinick EL, Gross H, Weinberger A, and Cohen H (2006) TNF-alpha modulation for
treatment of Alzheimer’s disease: a 6-month pilot study. MedGenMed 8:25.
Townsend M, Mehta T, and Selkoe DJ (2007) Soluble Abeta inhibits specific signal
transduction cascades common to the insulin receptor pathway. J Biol Chem
282:33305–33312.
Tran HT, Sanchez L, Esparza TJ, and Brody DL (2011) Distinct temporal and
anatomical distributions of amyloid-beta and tau abnormalities following con-
trolled cortical impact in transgenic mice. PLoS One 6:e25475.
Tsai YM, Chien CF, Lin LC, and Tsai TH (2011) Curcumin and its nano-formulation:
the kinetics of tissue distribution and blood-brain barrier penetration. Int J Pharm
416:331–338.
Tsampalas M, Gridelet V, Berndt S, Foidart JM, Geenen V, and Perrier d’Hauterive
S (2010) Human chorionic gonadotropin: a hormone with immunological and
angiogenic properties. J Reprod Immunol 85:93–98.
Tsinkalovsky O, Smaaland R, Rosenlund B, Sothern RB, Hirt A, Steine S, Badiee A,
Abrahamsen JF, Eiken HG, and Laerum OD (2007) Circadian variations in clock
gene expression of human bone marrow CD34cells. J Biol Rhythms 22:140–150.
Tu¨ kel C, Wilson RP, Nishimori JH, Pezeshki M, Chromy BA, and Ba¨ umler AJ (2009)
Responses to amyloids of microbial and host origin are mediated through toll-like
receptor 2. Cell Host Microbe 6:45–53.
Tukhovskaya EA, Yukin AY, Khokhlova ON, Murashev AN, and Vitek MP (2009)
COG1410, a novel apolipoprotein-E mimetic, improves functional and morpholog-
ical recovery in a rat model of focal brain ischemia. J Neurosci Res 87:677–682.
Tweedie D, Sambamurti K, and Greig NH (2007) TNF-alpha inhibition as a treat-
ment strategy for neurodegenerative disorders: new drug candidates and targets.
Curr Alzheimer Res 4:378–385.
Uguz F, Akman C, Kucuksarac S, and Tufekci O (2009) Anti-tumor necrosis factor-
alpha therapy is associated with less frequent mood and anxiety disorders in
patients with rheumatoid arthritis. Psychiatry Clin Neurosci 63:50–55.
Uysal KT, Wiesbrock SM, Marino MW, and Hotamisligil GS (1997) Protection from
obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature
389:610–614.
Vadakkadath Meethal S and Atwood CS (2005) The role of hypothalamic-pituitary-
gonadal hormones in the normal structure and functioning of the brain. Cell Mol
Life Sci 62:257–270.
Valcour V, Maki P, Bacchetti P, Anastos K, Crystal H, Young M, Mack WJ, Cohen M,
Golub ET, and Tien PC (2012) Insulin resistance and cognition among HIV-
infected and HIV-uninfected adult women: the women’s interagency HIV study.
AIDS Res Hum Retroviruses 28:447–453.
Valerio A, Bertolotti P, Delbarba A, Perego C, Dossena M, Ragni M, Spano P,
Carruba MO, De Simoni MG, and Nisoli E (2011) Glycogen synthase kinase-3
inhibition reduces ischemic cerebral damage, restores impaired mitochondrial
biogenesis and prevents ROS production. J Neurochem 116:1148–1159.
Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A, Palomba L, Cantoni
O, Clementi E, Moncada S, et al. (2006) TNF-alpha downregulates eNOS expres-
sion and mitochondrial biogenesis in fat and muscle of obese rodents. J Clin Invest
116:2791–2798.
Vandenbroucke K, de Haard H, Beirnaert E, Dreier T, Lauwereys M, Huyck L, Van
Huysse J, Demetter P, Steidler L, Remaut E, et al. (2010) Orally administered L.
lactis secreting an anti-TNF nanobody demonstrate efficacy in chronic colitis.
Mucosal Immunol 3:49–56.
Vavra JJ, Deboer C, Dietz A, Hanka LJ, and Sokolski WT (1959) Streptozotocin, a
new antibacterial antibiotic. Antibiot Annu 7:230–235.
Verhees KJ, Schols AM, Kelders MC, Op den Kamp CM, van der Velden JL, and
Langen RC (2011) Glycogen synthase kinase-3beta is required for the induction of
skeletal muscle atrophy. Am J Physiol Cell Phsyiol 301:C995–C1007.
Vitek MP, Brown CM, and Colton CA (2009) APOE genotype-specific differences in
the innate immune response. Neurobiol Aging 30:1350–1360.
Vitek MP, Christensen DJ, Wilcock D, Davis J, Van Nostrand WE, Li FQ, and Colton
CA (2012) APOE-mimetic peptides reduce behavioral deficits, plaques and tangles
in Alzheimer’s disease transgenics. Neurodegener Dis 10:122–126.
Wang AL, Yu AC, Lau LT, Lee C, Wu le M, Zhu X, and Tso MO (2005a) Minocycline
inhibits LPS-induced retinal microglia activation. Neurochem Int 47:152–158.
Wang CY, Wen MS, Wang HW, Hsieh IC, Li Y, Liu PY, Lin FC, and Liao JK (2008)
Increased vascular senescence and impaired endothelial progenitor cell function
mediated by mutation of circadian gene Per2. Circulation 118:2166–2173.
Wang Q, Wu J, Rowan MJ, and Anwyl R (2005b) Beta-amyloid inhibition of long-
term potentiation is mediated via tumor necrosis factor. Eur J Neurosci 22:2827–
2832.
Wang Y, Hao Y, and Alway SE (2011) Suppression of GSK-3beta activation by
M-cadherin protects myoblasts against mitochondria-associated apoptosis during
myogenic differentiation. J Cell Sci 124:3835–3847.
Weiner FR, Smith PJ, Wertheimer S, and Rubin CS (1991) Regulation of gene
expression by insulin and tumor necrosis factor alpha in 3T3–L1 cells. Modulation
of the transcription of genes encoding acyl-CoA synthetase and stearoyl-CoA
desaturase-1. J Biol Chem 266:23525–23528.
Wen TS, Randall DC, and Zolman JF (1994) Protein accumulation in cerebrospinal
fluid during -90 degrees head-down tilt in rabbit. J Appl Physiol 77:1081–1086.
Wen Y, Yang S, Liu R, Brun-Zinkernagel AM, Koulen P, and Simpkins JW (2004a)
Transient cerebral ischemia induces aberrant neuronal cell cycle re-entry and
Alzheimer’s disease-like tauopathy in female rats. J Biol Chem 279:22684–22692.
Wen Y, Yang S, Liu R, and Simpkins JW (2004b) Transient cerebral ischemia
induces site-specific hyperphosphorylation of tau protein. Brain Res 1022:30–38.
Westermann D, Van Linthout S, Dhayat S, Dhayat N, Schmidt A, Noutsias M, Song
XY, Spillmann F, Riad A, Schultheiss HP, et al. (2007) Tumor necrosis factor-alpha
antagonism protects from myocardial inflammation and fibrosis in experimental
diabetic cardiomyopathy. Basic Res Cardiol 102:500–507.
Wilson AC, Clemente L, Liu T, Bowen RL, Meethal SV, and Atwood CS (2008)
Reproductive hormones regulate the selective permeability of the blood-brain
barrier. Biochim Biophys Acta 1782:401–407.
Wu SR, Li CF, Hung LY, Huang AM, Tseng JT, Tsou JH, and Wang JM (2011)
CCAAT/enhancer-binding protein delta mediates tumor necrosis factor alpha-
induced Aurora kinase C transcription and promotes genomic instability. J Biol
Chem 286:28662–28670.
Xu Y, Tian Y, Wei HJ, Chen J, Dong JF, Zacharek A, and Zhang JN (2011)
Erythropoietin increases circulating endothelial progenitor cells and reduces the
formation and progression of cerebral aneurysm in rats. Neuroscience 181:292–
299.
Yamakawa I, Kojima H, Terashima T, Katagi M, Oi J, Urabe H, Sanada M, Kawai
H, Chan L, Yasuda H, et al. (2011) Inactivation of TNF alpha ameliorates diabetic
neuropathy in mice. Am J Physiol Endocrinol Metab 301:E844–E852.
Yamamoto M, Kiyota T, Horiba M, Buescher JL, Walsh SM, Gendelman HE, and
Ikezu T (2007) Interferon-gamma and tumor necrosis factor-alpha regulate amy-
loid-beta plaque deposition and beta-secretase expression in Swedish mutant APP
transgenic mice. Am J Pathol 170:680–692.
Yang Y and Herrup K (2007) Cell division in the CNS: protective response or lethal
event in post-mitotic neurons? Biochim Biophys Acta 1772:457–466.
Yazdani-Biuki B, Stelzl H, Brezinschek HP, Hermann J, Mueller T, Krippl P,
Graninger W, and Wascher TC (2004) Improvement of insulin sensitivity in
insulin resistant subjects during prolonged treatment with the anti-TNF-alpha
antibody infliximab. Eur J Clin Invest 34:641–642.
Yokota S (2003) Influenza-associated encephalopathy–pathophysiology and disease
mechanisms. Nippon Rinsho 61:1953–1958.
Young-Collier KJ, McArdle M, and Bennett JP (2012) The dying of the light: mito-
chondrial failure in Alzheimer’s disease. J Alzheimers Dis 28:771–781.
Zauli G, Toffoli B, di Iasio MG, Celeghini C, Fabris B, and Secchiero P (2010)
Treatment with recombinant tumor necrosis factor-related apoptosis-inducing li-
gand alleviates the severity of streptozotocin-induced diabetes. Diabetes 59:1261–
1265.
Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, and
Hauser CJ (2010) Circulating mitochondrial DAMPs cause inflammatory re-
sponses to injury. Nature 464:104–107.
Zhou QH, Boado RJ, Hui EK, Lu JZ, and Pardridge WM (2011) Brain-penetrating
tumor necrosis factor decoy receptor in the mouse. Drug Metab Dispos 39:71–76.
Ziegler SG and Thornton JE (2010) Low luteinizing hormone enhances spatial
memory and has protective effects on memory loss in rats. Horm Behav 58:705–
713.
Zotova E, Nicoll JA, Kalaria R, Holmes C, and Boche D (2010) Inflammation in Alzhei-
mer’s disease: relevance to pathogenesis and therapy. Alzheimers Res Ther 2:1.
TNF CAUSES INSULIN RESISTANCE:IMPLICATIONS FOR AD W
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Cramer et al. (Reports, 23 March 2012, p. 1503; published online 9 February 2012) demonstrates short-term bexarotene treatment clearing preexisting β-amyloid deposits from the brains of APP/PS1ΔE9 mice with low amyloid burden, providing a rationale for repurposing this anticancer agent as an Alzheimer's disease (AD) therapeutic. Using a nearly identical treatment regimen, we were unable to detect any evidence of drug efficacy despite demonstration of target engagement.
Article
Full-text available
HIV produces a chronic viral infection of the central nervous system that elicits chronic glial activation and overexpression of glial cytokines 1−5 that are also implicated in Alzheimer disease (AD) pathogenesis 6−11. A genetic risk factor for AD is the E4 isoform for apolipoprotein E (APOE)12,13. Here we compare the frequency of neurologic symptoms for subjects with and without the E4 isoform (E4(+)and E4(−), respectively) in an HIV cohort14−17. Compared with E4(−) subjects, twice as many E4(+) subjects were demented (30% compared with 15%) or had peripheral neuropathy (70% compared with 39%) at least once, and they had threefold more symptomatic examinations (13% compared with 3% and 42% compared with 14%, respectively)(P < 0.0001). Thus, neurologic symptoms for HIV-infection and AD are linked through an etiologic risk factor. Long-term survivors of HIV infection with E4 may be at high risk for AD; conversely, gene−viral interactions may speed AD pathogenesis.
Article
Full-text available
Obesity is associated with resistance to the actions of both leptin and insulin via mechanisms that remain incompletely understood. To investigate whether leptin resistance per se contributes to insulin resistance and impaired glucose homeostasis, we investigated the effect of acute leptin administration on glucose homeostasis in normal as well as leptin-or leptin receptor-deficient mice. In hyperglycemic, leptin-deficient Lep ob/ob mice, leptin acutely and potently improved glucose metabolism, before any change of body fat mass, via a mechanism involving the p110 and isoforms of phosphatidylinositol-3-kinase (PI3K). Unlike insulin, however, the anti-diabetic effect of leptin occurred independently of phospho-AKT, a major downstream target of PI3K, and instead involved enhanced sensitivity of the hypothalamus to insulin action upstream of PI3K, through modulation of IRS1 (insulin receptor substrate 1) phosphorylation. These data suggest that leptin resistance, as occurs in obesity, reduces the hypothalamic response to insulin and thereby impairs peripheral glucose homeostasis, contributing to the development of type 2 diabetes.
Article
It is well known that systemic infections cause flare-ups of disease in individuals with asthma and rheumatoid arthritis, and that relapses in multiple sclerosis can often be associated with upper respiratory-tract infections. Here we review evidence to support our hypothesis that in chronic neurodegenerative diseases such as Alzheimer's disease, with an ongoing innate immune response in the brain, systemic infections and inflammation can cause acute exacerbations of symptoms and drive the progression of neurodegeneration.
Article
In Reply: We agree with Dr Paz-Filho and colleagues that leptin levels show variation throughout the day,1 but we believe that this would lead to some random misclassification rather than confounding. It is unlikely that the degree of misclassification in leptin levels differs between participants who will and will not develop the outcome (dementia). Thus, this potential source of misclassification would likely bias the results toward the null hypothesis of no association between leptin levels and dementia. Our findings are also supported by data on 2871 adults in the Health ABC Study showing a correlation between higher leptin levels and a lower likelihood of decline on global cognitive test scores.2
Article
Overproduction of proinflammatory cytokines in the brains of transgenic animals causes brain pathology. To investigate the relationship between brain cytokines and pathology in the brains of animals with adult-onset, pathophysiologically induced brain cytokine expression, we studied rats infected with the parasite Trypanosoma brucei. Several weeks after infection, in situ hybridization histochemistry showed a pattern of chronic overexpression of the mRNAs for proinflammatory cytokines interleukin-1β and tumor necrosis factor-α in the brains of the animals. Similar spatiotemporal inductions of mRNAs for inhibitory factor κBα and interleukin-1β converting enzyme were found and quantified. The mRNAs for inducible nitric oxide synthase and interleukin-1 receptor antagonist were highly localized to the choroid plexus, which showed evidence of structural abnormalities associated with the parasites' presence there. The mRNAs for interleukin-6, interferon-γ, and inducible cyclooxygenase showed restricted induction patterns. Another set of animals was processed for degeneration-induced silver staining, TdT-mediated dUTP-digoxigenin nick end-labeling (TUNEL) staining, glial fibrillary acidic protein (GFAP) immunohistochemistry, and several other histological markers. Apoptosis of scattered small cells and degeneration of certain nerve fibers was found in patterns spatially related to the cytokine mRNA patterns and to cerebrospinal fluid diffusion pathways. Furthermore, striking cytoarchitectonically defined clusters of degenerating non-neuronal cells, probably astrocytes, were found. The results reveal chronic overexpression of potentially cytotoxic cytokines in the brain and selective histopathology patterns in this natural disease model. J. Comp. Neurol. 414:114–130, 1999. Published 1999 Wiley-Liss, Inc.
Article
It has been hypothesized that a central event in the early pathogenesis of sporadic Alzheimer disease (SAD) is the dysfunction of the neuronal insulin receptor signal transduction. To prove this, this receptor was inhibited by a triplicate icv application of STZ. Insulin binding sites were upregulated as in SAD. With respect to glucose transport proteins, detailed investigations are necessary.