PreprintPDF Available

The true nature of an autoimmune disease

Authors:
  • The Kinghorn Cancer Centre
Preprints and early-stage research may not have been peer reviewed yet.

Abstract

Abstract Here we propose a hypothetical model seeking to map the pathogenesis of a herpesviridae-positive serology subtype of Chronic Fatigue Syndrome / Myalgic Encephalomyelitis (CFS/ME) as a simultaneous α-ketoglutarate dehydrogenase (α-KGDH) and pyruvate dehydrogenase (PDH) deficiency, with degraded beta-adrenergic signalling cascade and impaired hepatic gluconeogenesis, phasic hyperlactatemia and hyperammonemia - as caused by herpesviridae-mediated antibodies and latent cell burden. For example, “M37GO37” targets dihydrolipoamide succinyltransferase from the α-ketoglutarate dehydrogenase complex (α-KGDC), creating an acute deficiency of α-KGDH, impairing Citric Acid Cycle (CAC) metabolites from succinyl-CoA / Complex V through malate, accumulating α-ketoglutarate (α-KG), reducing adenosine triphosphate (ATP), NAD+ and respiration. “M18GP8”, “M82GP8”, “M37GPl1” antibodies to pyruvate dehydrogenase complex (PDC), plus hypoxia, low physical activity and/or antibodies creating beta-adrenergic dysregulation can each cause a decrease in PDH, Cori Cycle efficiency and insulin resistance. When combined with succinate and argininosuccinate (ASA) deficiency, plus elevated α-KG, nitrogen disposal shunts to nitrogen retention via metabolism to L-glutamate and L-glutamine, triggering glutaminolysis. Sleeping, fasting and respiration decrease lactate and nitrogen retention via metabolic shunting, partially rescuing succinate availability for CAC, urea cycle metabolism via GABA. Each of these α-KGDH, PDH and beta-adrenergic cascade deficiencies are able to cause both of the others, adding additional complexity to diagnosis and treatment. These phases can be accompanied by debilitating symptoms associated with hyperammonemia, GABA deficiency, glutamate-induced excitotoxicity, uremia, hyperlactatemia, adrenergic and cortisol dysregulation, with accompanying hair, skin, GI, collagen, immune, sphingolipid, endocrine, sleep and neurological disorders. This further suggests investigating the herpesvirus family as causal for numerous disorders.
Review:
The true nature of an autoimmune
disease.
Authors: Joshua Leisk, Aline Noçon ©2021
Key words: Epstein-Barr Virus (EBV), hyperlactatemia, chronic fatigue syndrome, myalgic
encephalomyelitis, fibromyalgia, alpha-ketoglutarate dehydrogenase deficiency, pyruvate
dehydrogenase deficiency, herpesvirus autoimmune spectrum disorder
Abstract
Here we propose a hypothetical model seeking to map the pathogenesis of a
herpesviridae-positive serology subtype of Chronic Fatigue Syndrome / Myalgic
Encephalomyelitis (CFS/ME) as a simultaneous α-ketoglutarate dehydrogenase
(α-KGDH) and pyruvate dehydrogenase (PDH) deficiency, with degraded
beta-adrenergic signalling cascade and impaired hepatic gluconeogenesis, phasic
hyperlactatemia and hyperammonemia - as caused by herpesviridae-mediated
antibodies and latent cell burden.
For example, “M37GO37” targets dihydrolipoamide succinyltransferase from the
α-ketoglutarate dehydrogenase complex (α-KGDC), creating an acute deficiency of
α-KGDH, impairing Citric Acid Cycle (CAC) metabolites from succinyl-CoA / Complex V
through malate, accumulating α-ketoglutarate (α-KG), reducing adenosine triphosphate
(ATP), NAD+ and respiration.
“M18GP8”, “M82GP8”, “M37GPl1” antibodies to pyruvate dehydrogenase complex
(PDC), plus hypoxia, low physical activity and/or antibodies creating beta-adrenergic
dysregulation can each cause a decrease in PDH, Cori Cycle efficiency and insulin
resistance.
When combined with succinate and argininosuccinate (ASA) deficiency, plus elevated
α-KG, nitrogen disposal shunts to nitrogen retention via metabolism to L-glutamate and
L-glutamine, triggering glutaminolysis.
Sleeping, fasting and respiration decrease lactate and nitrogen retention via metabolic
shunting, partially rescuing succinate availability for CAC, urea cycle metabolism via
GABA.
-1-
Each of these α-KGDH, PDH and beta-adrenergic cascade deficiencies are able to
cause both of the others, adding additional complexity to diagnosis and treatment.
These phases can be accompanied by debilitating symptoms associated with
hyperammonemia, GABA deficiency, glutamate-induced excitotoxicity, uremia,
hyperlactatemia, adrenergic and cortisol dysregulation, with accompanying hair, skin, GI,
collagen, immune, sphingolipid, endocrine, sleep and neurological disorders.
This further suggests investigating the herpesvirus family as causal for numerous
disorders.
Introduction
The purpose of this paper is to stimulate discussion around the nature of the herpesvirus
family, including tick-borne variants such as MHV-68[123][124][125][127][136], their impact on
mitochondrial efficiency, metabolism, and their implied causal relationship for a very
large spectrum of diseases and disorders[95][100]. These disorders relate to the organs /
tissues infected and efficiency of both latent and lytic activity. This paper will primarily
focus on their role in CFS/ME and aim to provoke interest in further research around
applying this model to understanding and treating other diseases which may be directly
caused by the same family of viruses.
CFS/ME is a highly debilitating and poorly understood disorder that is regularly
associated with childhood EBV and less commonly with cytomegalovirus (CMV), human
herpesvirus-6 (HHV-6) and other infections[14]. It is a “bucket term” that broadly
describes a group of symptoms including extreme fatigue and myopathy which lasts for
more than six months[1].
EBV is the fourth member of the human-herpesvirus family, which is known for creating
a plethora of different antigens at different viral stages[109], ‘hijacking’ the host’s adaptive
immune response to replicate and maintain persistence. It has also been demonstrated
to replicate in epithelial cells via transcytosis[110].
Patients commonly report what can be described as cycles of symptoms, with initial
onset reported as a phase of [acute lymphadenopathy and flu-like symptoms], followed
by an overlapping phase of [acute lack of energy for movement, extreme fatigue,
neuralgia, myopathy, nausea, dizziness, neuropathy, increased sweating], with
idiopathic periods of [improved energy production, extreme afternoon exhaustion / post
exercise malaise (PEM), nausea, dizziness, dysautonomia, Raynaud’s Syndrome, liver
pain, gastroparesis, irritable bowel disorder, postural orthostatic tachycardia syndrome
(POTS) and encephalopathy] and confusion, anxiety, depression, restless leg syndrome
(RLS), involuntary muscle spasms, insomnia, endocrine disorders, alopecia, circadian
rhythm shift, edema, eating disorders, tinnitus and/or symptom onset induced by dietary
inputs[2][234].
-2-
Here we review current literature and propose a hypothetical model that attempts to map
and describe the aetiology and pathogenesis of a herpesviridae-positive serology
CFS/ME subtype, as a debilitating member of an (author proposed nomenclature)
Herpesvirus Autoimmune Spectrum Disorder (HASD)[95][100], also being a highly treatable
form of viral hepatitis.
HASD CFS/ME displays an array of simultaneous phasic metabolic and mitochondrial
impairments caused by:
1. an induced enzymatic deficiency of α-KGDH[22], initially impairing synthesis of
Complex V in the CAC, creating oxidative stress and unbalancing redox of
Nicotinamide Adenine Dinucleotide (NAD+) with its reduced form, Nicotinamide
Adenine Dinucleotide Hydrogen (NADH)[9][42][44][167][212][213], with
2. an induced enzymatic deficiency of PDH, initially impairing lactic acid clearance,
fatty acid synthesis, and
3. an induced dysregulation of beta2-adrenergic pathways leading to enzymatic
deficiency of activating monophosphate-activated protein kinase (AMPK),
dysregulating hepatic gluconeogenesis, along with many other pathway
alterations discussed further on.
In Japan, while studying primary biliary cirrhosis (PBC), Matsui et al., 1993[16] and
Fukushima et al., 1995[3] previously isolated and described a number of EBV-produced
mitochondrial antibodies, including “M37GO37”, which targets dihydrolipoamide
succinyltransferase (EC 2.3.1.61), an E2 component of α-KGDC[3[8][22] - completely
inhibiting production of α-KGDH in uninfected and presumably infected cells, which
already exhibit similar metabolic features[44].
Three additional antibodies “M18GP8”, “M82GP8”, “M37GPl1” target the PDC, causing
moderate impairment of pyruvate dehydrogenase[16].
Kojima et al. 1999[73] also reported an Epstein-Barr virus infection resembling
autoimmune hepatitis with lactate dehydrogenase (LDH) and alkaline phosphatase
(ALP) abnormalities.
Mitochondrial antibodies with similar targets have also been identified in PBC by a
number of other US-based studies[26][27][28][30], however those authors had not yet
attributed a causal link to any specific viral infection, possibly due to the language and
technological barriers around information sharing which existed during this decade.
-3-
It was initially considered that many of the observed metabolomic datasets in recent
studies[5][76][78][108] featured elevation of serum α-KG, known as Complex IV, produced by
oxidative decarboxylation of isocitrate via isocitrate dehydrogenase[8[11]].
Although α-KG can be also synthesised endogenously from glutamate via glutamate
dehydrogenase (GDH)[19] and/or from galacturonic acid by the microorganism
Agrobacterium tumefaciens[6], we did not consider these two sources to be a sufficient
explanation for the elevated serum α-KG in CFS/ME patients, when simultaneously
accompanied by an acute cellular energy crisis and other symptoms[1][2][77][156][167][180].
Based on reported urea cycle abnormalities, we hypothesised that elevated α-KG could
also be derived from a defect in the production of Complex V in the CAC, where
[Succinyl-CoA + NADH + H+ + CO2] is normally produced from oxidative
decarboxylation of [α-KG + NAD+ + CoA-SH] by [α-KGDH, thiamine pyrophosphate,
lipoic acid, Mg++, transsuccinytase][8][11][42][47].
This defect would cause an accumulation of α-KG produced in Complex IV of the
CAC[8][11][44] and an increased use of Branched-Chain Amino Acids (BCAAs)[10], as
observed in CFS/ME by Missailidis et al., 2020[9]. Each impaired CAC cycle, caused by
eg. glycolytic energy usage for muscle activation and brain function, would continue to
accumulate one additional unit of α-KG / Complex IV. Elevated α-KG is also associated
with degraded ATP Synthase, reducing ATP production and mTOR[39][56].
α-KGDH is a critical enzyme in regulating efficiency of the CAC and is highly sensitive to
oxidative stress, while paradoxically being a source of it - in the presence of reactive
oxygen species (ROS), such as hydrogen peroxide (H2O2) and 4-hydroxy-2-nonenal
(HNE), α-KGDH can be completely inhibited, acutely regulating energy production[167].
With α-KGDH deficiency, elevated α-KG / Complex IV can be metabolised to
L-glutamate and L-glutamine[4][21] thereby increasing nitrogen retention[20], while impairing
normal metabolism and renal excretion as urea through the UC[4][21][39]. This is relative to
high levels of α-KG[22], with low levels of succinate->[...]->argininosuccinate (ASA)[107][131]
and driven by CAC activity or glutaminolysis[216].
We considered that a Complex V production defect, via HHV-induced α-KGDH
deficiency was the highest probability vector for simultaneously explaining many of the
key CFS/ME features[10][14][24][42][44].
Some CFS/ME features, such as high lactate:pyruvate ratio and downstream cascading
effects, including lactic acidosis, insulin resistance, dysautonomia and glutaminolysis
were as yet explained[44][76]. We considered that this lactate:pyruvate ratio appeared likely
related to PDC/PDH abnormalities.
-4-
With similar characteristics to some cancers, α-KGDH deficiency causes depletion of
succinate and fumarate, thereby inducing hypoxia, insulin resistance, decreasing AMPK,
decreasing collagen production, bone density via a cascade of deficiencies downstream
from prolyl hydroxylases [P4H->hydroxyproline->[collagen]->prolidase->proline<->P5C]
and promoting carcinogenesis via disrupting epigenetic regulation via hypoxia-inducible
transcription factors (HIF)[60][78]. HIF affects hundreds of genes, including those
contributing to the Warburg effect, as well as DNA methylation and histone methylation.
Notably, it also affects expression of PDH[59][126][174].
However, while α-KGDH deficiency and/or “M18GP8”, “M82GP8”, “M37GPl1” antibodies
to PDC[16] would cause a direct deficiency of PDH[66][67], this seemed like a fairly simplistic
answer and we considered that other factors or alternative scenarios, involving different
antibodies, could create the same deficiencies.
Nilsson et al., 2020 performed related research and were unable to find antibodies to
PDC in 160 out of 161 CFS/ME patients. They were further unable to detect
antimitochondrial antibodies (AMA) in a spot-check of 29 random samples from the
CFS/ME cohort, however they did not specify if they also tested any PBC control
samples for AMA. Interestingly, the two methods they used for detecting antibodies to
PDC yielded different results on the same samples, suggesting the testing
methodologies are unreliable[128].
Relative to rescuing CAC efficiency from α-KGDH deficiency, Complex V / Succinyl CoA
can also be created from methylmalonyl CoA by the enzyme methylmalonyl-CoA
mutase, through the utilisation of deoxyadenosylcobalamin[79]. This may negatively
impact serum B12 levels and reduce PDH, which is needed to produce acetyl-CoA,
required as an irreversible, rate-limiting intermediate in the pathway to malonyl-CoA.
This reaction is also involved in the catabolism of some BCAAs and odd-chain fatty
acids[80].
GABA is another pathway to partially rescue Complex V / Succinyl CoA, via
succinate.The metabolism and depletion of GABA to partially restore succinate levels
and CAC efficiency can cause hypoxia and decreased PDH[70], sleeping disorders[50][51],
ulcerative colitis[158] and anxiety disorders[49][53].
During α-KGDH deficiency and/or when under oxidative stress, a further pathway to
rescue succinate has been shown to exist by decarboxylation of α-KG, pyruvate and
oxaloacetate[129][165][167].
The ratio of NAD+:NADH determines the direction and rate of critical enzymatic
reactions involved in mitochondrial activity. Relative to α-KGDH and PDH impairments,
NAD+:NADH redox balance could be impaired, depleting NAD+ and pooling NADH,
causing further impairment to many enzymatic reactions, impacting mitochondrial
efficiency[67][212][213].
-5-
Lactate is largely produced in muscle cells and is generally thought to be transported by
blood into the liver, where it is normally metabolised into glucose via the Cori Cycle and
transported back to the cells for reuse. Recent studies have shown that lactate can also
be used as a preferential source of energy and trigger for cell proliferation. Lactate can
also be shared between cells[7][61][77].
Lack of physical activity and/or carnitine both decrease PDH and lead to lactate
increase. Hepatic function, with a particular focus on lactate metabolism and
gluconeogenesis, is a key factor in maintaining systemic glucose:lactate homeostasis[68].
Even in the absence of viral antibodies, the forced sedentary lifestyle imposed on
CFS/ME patients could acutely contribute to ongoing symptoms of hyperlactatemia, with
or without lactic acidosis, as determined by transient urea cycle efficiency[191][237][238][239].
Another CFS/ME feature is hypoxia, which induces insulin insensitivity and
beta-adrenergic dysregulation[132] via increased G protein coupled receptor type 2
(GRK2). ß2-adrenergic signalling cascade dysregulation leads to impaired fatty acid
oxidation[55][71]. This cascade has implications for further impairing the immune response
to viral infections and downregulation of natural killer (NK) cell production[147] and altering
glutamate metabolism[23][37].
However, antibodies for ß2-adrenergic receptors, muscarinic acetylcholine receptors,
gamma-opioid receptors, dopamine receptors, serotonin and serotonin receptors have
also been discovered in a subset of CFS/ME patients, for whom removal /
immunoadsorption of IgG provided significant, yet temporary benefits. These antibodies
would also be able to cause the previously described CFS/ME pathway alterations, by
inducing the same three pivotal looping cascades of defects - α-KGDH deficiency,
PDH deficiency and impaired β2-adrenergic cascade. In addition, each identified
antibody target may benefit from additional consideration and therapeutic intervention
while these antibodies are present in the serum[35][37][62][128][129][130][132][147][165].
These three pivotal cascades of defects, including further disturbances to calcium
channels, described later[105][106], can cause increased insulin resistance, decreased
cyclic adenosine monophosphate (cAMP)[186], decreased adenosine
monophosphate-activated protein kinase (AMPK)[245], decreasing the Cori Cycle
efficiency and leading to hyperlactatemia or lactic acidosis[61][71]. cAMP pathway defects
can also disrupt normal cardiac function by dysregulating protein kinase A (PKA)[187][188].
The Cori Cycle defects result in post-exercise malaise (systemic inflammation, acute
hepatitis, neuropathy, myopathy, encephalopathy, psychosis)[64], gut microbiome
dysbiosis[155], increased digestive activity[154], depression, increased cancer risk and
pyramidal symptoms[43][44][45][52][63][72].
The described altered state of metabolism with hypoxia and hyperlactatemia also
remodels mitochondrial energy production towards glutaminolysis[25][45][65][216] which can
-6-
bypass currently degraded steps of the CAC (succinyl-CoA, succinate, fumarate,
malate), via transamination at oxaloacetate, while increasing metabolites of ammonia
and producing further lactate[76][77].
A “partial recovery” state, or reversal of described impairments, could be expected
intra-daily - during sleep[69] and resting periods, when energy production demands are
reduced and less than the maximum capability provided by the impaired beta-oxidation
cascade, thus sparing available succinate for CAC and UC metabolism needs. During
this “recovery state”, NAD+ biosynthesis and recovery via increased NAMPT normally
occurs as well[212][213]. Fasting and metabolic ketosis can also induce this “partial
recovery” state, by allowing additional mitochondrial efficiency via ketogenesis as an
alternate energy source and increased relative efficiency of hepatic gluconeogenesis.
Additional events may occur when (diurnal) GABA levels spike[51], which could be
depleted and metabolised to succinate[48][70], partially healing CAC and UC efficiency.
-7-
[Figure 1 - Pathway Diagram]
(High resolution included with supporting files)
This may also adversely affect sleep schedules[50][51][69], contribute to a number of anxiety
disorders[53][182] and eating disorders[181].
During sleep, elevated lactate levels and/or lactic acidosis could decline, reducing or
ameliorating lactatemia-related symptoms. However, with some relationship to ammonia
and nitrogen balance, if lactate levels decline and serum pH increases sufficiently during
eg. a typical 8 hour sleep cycle, mitochondrial energy production could revert from
glutaminolysis to impaired glycolytic metabolism or fatty acid oxidation.
In this state, mitochondrial efficiency could now be proportional to GABA->succinate[70]
and/or B12/methylmalonyl CoA->succinyl-CoA availability[79] and while impaired by
α-KGDH deficiency, cause a proportional “low energy state”, until enough impaired
mitochondrial activity, with insufficient PDH, again causes lactate to increase and serum
pH to decrease. This lactate elevation may retrigger glutaminolysis[7][65], allowing for a
small phase of increased mitochondrial efficiency, until lactate levels again reach a
debilitating level or acute lactic acidosis, or merely hyperlactatemia, as pH balance is
somewhat compensated for by the increased ammonia also created by the process.
Further, if UC efficiency is unable to match ammonia synthesis rates, generated by
glycolysis, glutaminolysis and/or α-KGDH deficiency, both minor and extended phases of
L-glutamate induced excitotoxicity[12][19], hyperammonemia, uremia[13] and symptoms
resembling multiple sclerosis[12][23][74] may be observed, following physical activity.
The stages described above can be expected to cyclically repeat[14][54][166] over days or
weeks. Relative to both renal clearance and hepatic function, some of these phasic
symptoms may overlap or be observed simultaneously.
PDH or acetyl-CoA abnormalities described earlier can also reduce conversion of
intracellular fatty acids into ceramides, perhaps favouring the use of sphingolipids in
ceramide synthesis, which are needed for other purposes[184].
The resulting decrease in sphingolipids[98], including sphingosine-1-phosphate and
sphingomyelin, are associated with muscle contraction dysfunction, mechanical pain
response, ion channel dysregulation, liver abnormalities, cognitive dysfunction, bipolar
disorder, anxiety, myelin formation abnormalities, schizophrenia (including dissociation,
derealization, delusions, and paranoia), mast cell dysregulation (further causing
histamine and serotonergic issues) and irritable bowel
syndrome[62][74][99][101][102][103][104][105]106][153][167][168][169][170].
PDH->acetyl-CoA deficiency is also a rate limiter in sterol production pathways, which
when combined with hypoxia and insulin resistance, has implications for a potential
cascade of endocrine alterations, sleep dysregulation issues, including circadian rhythm
alterations via cortisol[84] (and previously mentioned GABA [50][51]).
-8-
The endocrine alterations may also cause hirsutism, alopecia, ovarian follicle
disturbances, fibroids, endometriosis via compensatory upregulation of adrenal cortex
pathway hormone production, including 5-alpha reductase (5-AR) and
dihydrotestosterone (DHT) in response to dysregulated cortisol[85][185]. This effect would
be enhanced by mast cell dysregulation and gut / skin barrier degradation, with follicle
thinning/loss from the cortisol dysregulation and acute collagen synthesis impairments
downstream from α-KGDH deficiency, as previously
described[60][79][81][82][83][85][88][89][90][92][93][252].
This could be further impacted and sustained by a compensatory rebound effect of
hypothalamic–pituitary–gonadal (HPG)-axis suppression by DHT dominance, which
could be observable via pathology markers as “in range”, yet low luteinising hormone
(LH) and oestradiol (OE2) insufficiency (eg. in males - LH < 2.8IU/L, OE2 <70 pmol/L),
indicating hypothalamic feedback and gonadotropin hormone-releasing hormone
(GnRH) regulation is being governed by elevated 5-AR metabolites - allopregnanolone
or DHT[86][87].
These combined alterations would have further implications for hepatic function,
including lipid accumulation / non-alcoholic liver disease (NAFLD) and lactate
metabolism via gluconeogenesis[91][153].
-9-
At this time, it’s not at all clear which pathogens express the antigens associated with
each antibody. Early indications would suggest that EBV, MHV-68, CMV, HHV-6, HHV-7
would have the highest probability of being causal in CFS/ME, however this area has not
been adequately explored and it’s likely other common pathogens may have
successfully evolved through the use of similar vectors[157].
The model as described is extensible and may allow other pathogens or known
antibodies to be tested against it to predict pathophysiology.
For example:
A plausible pathophysiology for HHV-seronegative CFS/ME, based on a Clostridium
difficile
infection, which has been reportedly found in 22% of CFS/ME patients by Dr.
Jacob Teitelbaum.
-10-
[Figure 2 - Flowchart for HASD CFS/ME Model]
(High resolution included with supporting files)
Applying the HASD model, C.diff-secreted GDH, increasing serum GDH, further elevates
α-KG, thus degrading Complex V. This degradation of Complex V, as previously
described, is capable of causing inhibition of α-KGDH, leading to the entire looping
cascade of CFS/ME metabolic alterations. These symptoms are related to the
C.diff-secreted GDH, when combined with organ-specific elevated expression of GDH.
Notably, these organs are focused on synthesis tasks (eg. liver, gonads, etc)[39][56][214][217].
This further suggests that CFS/ME induced by c.diff infection, without a HHV
seropositive co-infection, could be treated by eg. Metformin and epigallocatechin gallate
(EGCG), which is found in green tea and acutely inhibits GDH (and therefore being
specifically contraindicated in HASD CFS/ME.)[38][215][217].
[This example has been annotated in blue on “Figure 2.”
]
This model also allows for speculation around the existence of eg. HHV-mediated or
other heterophile antibodies to GDH, as a further trigger for inducing CFS/ME.
Research into creating a comprehensive map of antigens / antibody targets for each
pathogen is sorely needed.
In previous years, latent cells have been traditionally thought of posing an insignificant
long-term health risk, however research has shown that these cells are programmed to
prefer glycolysis and glutaminolysis, further creating a lactate, hypoxia and ammonia
burden which can also trigger lytic reactivation and latent cell proliferation[7][44][65][216].
In addition to the well-known lytic replication phase, these cells have now been
demonstrated to replicate via transcytosis, having further implications for a number of
cancers[110].
In key ways, many herpesviridae-infected cells share common metabolic features with
senescent cells and cancer cells featuring the “Warburg” or “Reverse Warburg” effect -
hypoxia, lactate, elevated aerobic glycolysis and a preference for glutaminolysis, via
enhanced expression of KGA, GLS1, GLUD1, GLUD2 proteins[44][140][173][216].
Mirroring the behaviour of senescent cells and various cancer cells, these infected cells
are able to influence the behaviour of neighbouring cells and recruit them[110]. Various
suspected herpesviridae-related diseases and disorders show symptoms with strong
relevance to the location of these infected latent cells and their burden on neighbouring
cells and tissues[95][100][134[183].
It’s possible that hypoxic complications from SARS-CoV-2 infections / COVID-19 are
able to reactivate a latent EBV (or other HHV) infection, causing mononucleosis, chronic
fatigue, hair loss, headaches, dyspnea and other symptoms associated with “Long
COVID” syndrome[177][178].
-11-
As the herpesviridae-infected cells behave like senescent and various cancer cells, this
suggests they are also susceptible to the same treatment methods already being
explored, for example - selectively disabling glycolysis, glutaminolysis or
phosphodiesterase 4 (PDE4)[98][143][176][180][189][190].
This further suggests that induced necrosis of these cells could also be explored by
causing selective mitochondrial starvation, by perhaps creating a sustained
simultaneous state of [high lactate + low serum glucose + selective glycogen depletion +
glutamine depletion]
, eg. performing a “water fast” for 3 or more days and exercising,
while inhibiting both PDH kinase and GDH/GLS[38][98][140][175].
Implications and considerations for diagnostics and
management of the HASD CFS/ME model hypothesis
This HASD CFS/ME hypothesis model describes a complex array of disorders, which
suggest a number of challenges and a combination of therapeutic targets. We will further
review therapeutic options that may be appropriate for management and prevention or
remission of symptoms, based on this model.
As the model includes both a lytic and latent herpesviridae infection as the root cause,
these are logically the most desirable therapeutic targets for achieving long-term
remission, however symptomatic relief can also be provided and may further enhance
the patient’s immune response.
As with any infection, managing patient awareness around expected symptoms
associated with a sustained period of a heightened immune response could be advisable
- rashes, lymphadenopathy, fevers / chills would all be expected, especially in the early
stages of treatment.
The patient would be expected to go through various stages of recovery, from being
bed-ridden, through periods of afternoon fatigue and headaches, to periods of minor
limitations and then symptomatic relief, in a non-linear manner and relative to consistent
levels of appropriate physical activity.
Remission would come from a full arrest of lytic phase replication, combined with a
sufficient reduction of latent hepatic cell burden, allowing normal hepatic and other
metabolism, without interruption by neighbouring infected cell metabolism.
A key understanding of the proposed HASD CFS/ME model is that treatment MUST
include a comprehensive therapeutic intervention for each of the primary targets
described:
-12-
1. HHV Latent and Lytic Infection
2. α-KGDH Deficiency Cascade (including CAC, NAD+ depletion, nitrogen disposal)
3. PDH Deficiency Cascade (PDH/PDHK, carnitine pathways, NAD+)
4. ß2-Adrenergic Cascade (β2-AR->cAMP->AMPK-> Hepatic Gluconeogenesis)
5. Muscarinic Acetylcholine Receptor Homeostasis
A failure to treat each of these targets simultaneously will likely result in a short-term
improvement, followed by a delayed and intense, avoidable cascade of CFS/ME
symptoms.
Any treatment should also include therapeutic interventions to address the
secondary antibody targets, being mainly neurotransmitters and their receptors.
Diagnostics
Confirming a positive CFS/ME diagnosis has traditionally been problematic, as serology
markers can be inconclusive or altered by heterophile antibodies and viral behaviour,
where coinfections exist.
In the absence of a industry-standardised panel for assessing HHV infections and further
identifying antibodies present, a thorough approach to diagnostics would include
serology markers for the known family, with quantitative results. Simplified results
displaying “positive” or “negative” are not sufficient for a meaningful interpretation, as the
titres are critical tools in assessing infection status:
HSV-1 IgG, IgM
HSV-2 IgG, IgM
VZV IgG, IgM
EBV Early Antigen (EA),
EBV Viral Capsid Antigen (VCA) IgG, IgM
EBV Nuclear Antigen (EBNA) IgG
CMV IgG, IgM
HHV-6 IgG, IgM
HHV-7 IgG, IgM
KSHV IgG, IgM
Clinical Notes for EBV:
Interpreting serology for EBV has many complications. Due to the different stages of
infection and antigens expressed, EBV positive serology is normally interpreted via
multiple markers:
EBV EBNA IgG “positive” indicates a long-established latent infection and level of
burden. A “negative” results with any other EBV marker as “positive” indicates a recent
infection.
-13-
EBV EA anything other than “negative” indicates a recently active lytic infection,
regardless of other markers. In the presence of EBNA+, can indicate a chronic /
reactivating infection.
EBV VCA IgG “high” positive indicates an active or recent infection and severity | “low”
positive, in the absence of EBV EA or EBV VCA IgM indicates immunity.
EBV VCA IgM “high” positive indicates an active infection | “low” positive may indicate
the presence of a chronic coinfection, such as CMV, or HHV-6.
Heterophile antibodies can be a useful marker for diagnostics.
PCR tests - these are exceptionally useful for detecting and quantifying a lytic phase at
the time of the test
. Given the difficulties sometimes involved with patients being able to
travel on the day of a scheduled blood draw, owing to debilitating symptoms, PCR tests
can be an unreliable tool for initial screening, unlike lagging data provided by
immunoglobulin counts.
Faecal tests - testing for the presence of C.diff and other infections that may require
further treatment or interfere with HASD CFS/ME treatments.
Liver Function Tests (LFT) - the standard metabolic markers for inferring liver function
are not expected to be helpful in assessing impairments described in this model.
Some other early diagnostics ‘clues’ towards an undiagnosed chronic HHV infection may
also include:
1. Haematology markers showing a historical pattern of subclinical or low levels of
lymphocytes, without any obvious signs of infection.
2. T / B cell profiles on flow cytometry testing may also look unfavourable, with reference to
low CD4:CD8, as seen in HIV infections.
3. Blood urea levels may show a pattern of unusually low or high readings, unrelated to the
patient’s recall of dietary protein intake.
4. History of fussy eating disorder, low BMI and/or GI intolerances.
5. History of anxiety, depression, bipolar disorder or schizophrenia.
6. History of obsessive / compulsive behavioural disorders, including unusual levels of
dietary supplement usage.
7. Alopecia or other endocrine-related disorders.
8. History of connective tissue disorders or cysts.
9. History of nasal infection.
10. A positive “succinic acid challenge” (see “α-KGDH Deficiency Interventions / Succinate”)
,
where the patient chooses to consume up to 4 doses of succinic acid at 25mg per dose,
20 minutes apart, while monitoring for any physiological response, including a ‘surge of
energy’, ‘a sustained adrenaline spike’, or in other HASD patients, expected delayed
inflammation of infected tissues.
-14-
ANTIVIRAL THERAPY
Vaccination
There have been previous attempts to create a robust vaccine for EBV, however one
extremely promising piece of work was published in 2018.
van Zyl DG, Tsai M-H, Shumilov A, Schneidt V, Poirey R, Schlehe B et al., created
“Immunogenic particles with a broad antigenic spectrum stimulate cytolytic T cells and
offer increased protection against EBV infection ex vivo
and in mice”[121].
This research will hopefully provide a scalable, long-term solution. We wish the authors
the very best of luck in this endeavour and will continue to watch their work with interest.
Rituximab
Despite promising early indications for CFS/ME and despite demonstrating efficacy for
preventing similar EBV-infections following organ transplants, the monoclonal antibody
“rituximab” failed to show efficacy for CFS/ME in a phase 3 double-blind trial[29], where
the treatment was periodic b-cell depletion, with 2 infusions of rituximab, 500 mg/m2 of
body surface area, 2 weeks apart, followed by 4 maintenance infusions with a fixed dose
of 500 mg at 3, 6, 9, and 12 months.
It is possible this was an off-target treatment, related to high specificity against an
expressed protein and the variance amongst EBV mutations / strains, often presenting
different protein targets for monoclonal antibody therapies, including
CD19/20/21[31][32][33][34].
However, we would suggest that, unlike in posttransplant lymphoproliferative disease
(PT-LPD), the extensive latent hepatic infection and burden to surrounding cells in
CFS/ME patients is not being treated by periodic b-cell depletion therapy alone. This is
an incomplete approach. Lytic reactivation of latent cells could also be expected and
therefore any EBV-mediated antibodies created would persist for months in between
periodic treatments, allowing acute symptoms to continue.
Tenofovir
Tenofovir is a nucleotide reverse transcriptase inhibitor that shows strong efficacy
against EBV lytic replication[119] and a number of other problematic viruses, including
Human Immunovirus (HIV) and Hepatitis B (HBV). Tenofovir is commonly used even
during pregnancy and appears to be a fairly low-risk treatment.
Significant side effects reported include high blood lactate and an enlarged liver, which
has been previously attributed to drug-induced disease modifications for HBV[120],
however in retrospect, could also fit the expectations described in this paper for hepatic
infection by EBV or other HHV, where Tenofovir was used without simultaneous dosing
-15-
of a PDH kinase (PDHK) inhibitor. Long term use is associated with nephrotoxicity and
osteopenia.
Spironolactone
Recent advances in understanding HHV / EBV replication has identified that an existing
drug, “Spironolactone” (aldactone), can cause degradation of xeroderma pigmentosum
group B-complementing protein (XPB), in both B lymphocytes and epithelial cells,
inhibiting EBV SM-dependent late lytic gene transcription[17].
At lower doses, spironolactone has existing clinical uses as a potassium sparing diuretic
and at higher doses as an anti-androgen, providing some additional considerations if
used as a treatment for EBV infections[40][41]. At 100mg/day, 30% of males developed
symptoms of gynecomastia[113]. However, even at 200-400mg/day, male participants who
experienced anti-androgenic symptoms, such as gynecomastia[114], over 4-13 months
had complete reversal of these effects upon cessation of dosing[112]. A mild aromatase
(CYP19) inhibitor to rebalance the oestradiol to DHT ratio could be considered in
unusual cases - as HASD CFS/ME patients would be expected to already exhibit high
DHT and low oestradiol, owing to chronic cortisol elevation[115].
This discovery prompted an encouraging pilot study of patients with positive serology for
EBV (n=21)
, by Campo et al, 2020[18], with 5 out of 16 participants who tolerated
25mg/day of spironolactone and multivitamins showing full remission of CFS/ME and the
remaining participants showed a reduction of CFS/ME symptoms at the time of the
report. The duration of the study was not reported. We noted no mention of patient
hydration or electrolyte management was included, which may have contributed to some
of the participant intolerance / dropouts, if not an omission in reporting. Considering
spironolactone’s 1.4 hour (and metabolites’ 13.8 - 16.5 hour half-life)[110], the dosing
schedule used may not have been optimal in maintaining the serum levels needed for
antiviral efficacy and twice daily dosing of 25mg could be considered.
While spironolactone shows some early signs of strong efficacy at safely modulating
EBV and other HHV lytic infections, relative to the lifespan of b-lymphocytes and serum
antibodies, it’s reasonable to expect it would take months to reach a state of viral lytic
phase remission. Unlike the targeted b-cell depletion of rituximab, this treatment or
tenofovir allows the immune system to simply recover and remodel around maintaining a
robust, persistent response to EBV lytic events or reactivation.
After an expected 2 month “initial recovery” phase, as indicated by an unremarkable T /
B cell profile on flow cytometry testing, an extended “maintenance dose” of
spironolactone could be continued, while using lytic phase reactivating compounds, such
as histone deacetylase (HDAC) inhibitors[179] or compounds such as cordycepin[116][117]
and/or thymoquinone[118] (which appears to have additional effects against latent cells),
until full remission is achieved. Patients should be cautioned that additional attention to
-16-
their symptoms management protocol may be required to address increased levels of
antibodies.
We would caution this complementary approach could be ill-advised during the initial
treatment phase, as increasing an already elevated lytic activity may exceed
spironolactone’s dose efficacy and the capabilities of a still-recovering immune system.
Metabolic Shunting
As already described in previous studies for senescent cells, lymphomas and other
cancers, altering the metabolic parameters for infected cells, by eg. PDE4 inhibition
and/or PDHK inhibition and/or anaplerosis and/or GLS1 inhibition is sufficient to impact
cell proliferation and cause tumor shrinkage. The author’s opinion is that administration
of the therapeutic interventions described in “Symptoms Management” may have
selective, positive effects on infected cell viability and may also extend to targeting other
cells exhibiting the same metabolic features[98][140][143][144][147][149][150][151][175][176][179][180][189][190].
SYMPTOMS MANAGEMENT
The symptoms described in the HASD model are considered to be highly debilitating and
sometimes life-threatening, requiring an increased level of patient awareness around
expectations, timeline and management.
Due to the nature of the infection, the impact of HASD CFS/ME impairments would relate
to both lytic phase activity and separate latent cell location, burden and activity. In this
CFS/ME model, the primary burden revolves around hepatic impairments with other
organs, such as the brain, as secondary targets.
HASD CFS/ME should be considered as a chronic form of viral hepatitis and the
methods reviewed in this section could also be suitable for early stage intervention of
HHV infections, including mononucleosis.
The mitochondrial and metabolic impairments described previously suggest some key
targets for therapeutic intervention. The additional challenges for CFS/ME patients
involve the existing adaptations or long-term compensations around mitochondrial and
metabolism alterations, including lactic acid cycle efficiency, glutaminolysis and receptor
homeostasis, including GABA, dopamine, beta-adrenergic, muscarinic and others.
As previously stipulated, all primary targets described herein must be addressed
simultaneously to form a therapeutic intervention in this model.
It has been previously described that entering and exiting a ketogenic diet induces
temporary metabolic and mitochondrial impairments, while enzymatic adaptations inside
the cells and microbiome alterations are enacted. We would postulate that in HASD
-17-
CFS/ME, rehabilitation from adaptations to chronically altered hepatic metabolism would
be expected to have a similar, albeit longer transitional period with reduced efficiencies,
including expression of PDH and PDK[191][192][193][194].
Contraindications
Acetyl-CoA:CoA
The ratio of Acetyl-CoA to CoA acts as a rate limiter for critical enzymatic reactions
involved in pyruvate and carnitine metabolism pathways. These would have acute
effects for altering efficacy of therapeutic interventions and/or inducing preventable
symptoms. Caution is advised around co-administration of any medications or
supplements which further alter these ratios[159][160]161].
Anticholinergics
Based on the awareness of HHV-mediated antibodies and pathway alterations that
affect acetylcholine (ACh) receptor expression and homeostasis, the use of
anticholinergics would be expected to create cognitive and muscle activation
impairments in HASD CFS/ME patients[62][103][128].
Aspirin
Aspirin has a direct interaction with α-KGDH and α-KG binding. It has been
demonstrated to increase permeability of the mitochondrial inner membrane, as
demonstrated by acute intramitochondrial release of NAD(P)+[145][146]. This indicates
aspirin / acetylsalicylate / salicylate could add additional complications to HASD.
Berberine
Berberine is an over-the-counter (OTC) dietary supplement with similar properties to
Metformin that has been trialled as a successful therapeutic intervention in the
management of type-2 diabetes, with further benefits suggested for metabolic disorders
and related gut microbiome dysbiosis[219][220][222] and IgE based food allergies[223].
Berberine has been shown to have antidepressant effects by antagonising dopamine
receptors, also suppressing innate and adaptive immune responses in experimental
models of colitis[221]. It has also been shown to positively affect liver cancer cells by
suppressing glutamine uptake via SLC1A5[224], suggesting benefits against latent HHV
infection.
Unfortunately, due to the mechanisms of action beyond activating AMPK that affect
hepatic gluconeogenesis, berberine is contraindicated in HASD CFS/ME[246][247].
-18-
GDH
Epigallocatechin Gallate (EGCG) is found in green tea and acutely inhibits GDH,
affecting the metabolism of α-KG to glutamate and vice-versa. While this may provide
significant benefits for inhibiting latent cell proliferation in HHV or excess ammonia /
glutamate production from elevated α-KG in HASD CFS/ME, the effects of drinking
green tea and/or consuming extracts of green tea could create acute, temporary hepatic
impairments for HASD CFS/ME patients which could persist for days[38].
Metformin
Metformin is a commonly prescribed drug for managing type-2 diabetes, with similar
properties to Berberine. Intolerances to Metformin have been reported, complicit to
expected glucose:lactate alterations in the GI tract and related gut microbiome
dysbiosis[225].
Benefits against neurodegenerative diseases have been reported, as well as a reduction
in all-cause mortality for type-2 diabetics with cancers[226][227][228].
Unfortunately, due to the mechanisms of action beyond activating AMPK that affect
hepatic gluconeogenesis, metformin is contraindicated in HASD CFS/ME[246][247].
α-KGDH Deficiency Interventions
Resolving α-KGDH deficiency in the HASD CFS/ME model would allow for acute
improvements to mitochondrial efficiency and amelioration of the downstream cascade
of impairments. Resolving this deficiency via anaplerosis at succinate may also positively
alter immune response and infected cell viability, particularly when combined with the
“PDH Deficiency Interventions’ described.
Succinate
Succinate (succinic acid / amber acid / butanedioic acid)
has shown high efficacy at
reversing key menopausal symptoms in two randomised, double-blind,
placebo-controlled clinical trials[143][144]. In the first trial, the daily dose used was
10.54-11.4 mg/kg. The second trial used approximately half that dose, or 400mg/day.
The resulting commercialised product, Amberen, is available for these purposes. It has
shown benefits for specific types of cancer cell lines.
Succinic acid is available as both a dietary supplement and food additive (E363) in many
countries. In the HASD model, exogenous succinate provides a direct way of healing the
CAC, via anaplerosis, at the point where an α-KGDH deficiency creates the cascade of
impairments, leaving only a redox depletion of NAD+ to resolve exogenously and an
-19-
elevation of α-KG to be metabolised to an increased glutamate and glutamine load,
before being further excreted as urea.
Relative to patient activity levels, sufficient attention to hydration and urination sufficiency
would be necessary. Increasing GDH may possibly provide some benefits for increasing
a-KG metabolism (see: "β2-Adrenergic Cascade / Corydalis Ternata.")
In the herpesviridae seropositive CFS/ME model, succinate would rapidly restore
mitochondrial efficiency, however caution should be noted around using this “suddenly
available” energy without also simultaneously treating the PDH deficiency and
β2-adrenergic cascade impairments
, otherwise lactate will accumulate with physical
activity, leading to preventable myopathy, hepatitis, encephalopathy and general
inflammation. Relative to individual circumstances, painful symptoms from
hyperlactatemia could take hours or many days to resolve, also leading to latent cell
proliferation and lytic reactivation, in extreme circumstances.
Based on available clinical trial data and further addressing expectations around
CFS/ME dysfunctional beta-adrenergic signalling and dysautonomia, a suggested initial
assessment dose schedule of approximately 0.25mg/kg, repeated every 20 minutes and
up to 100mg, or until sustained “mild adrenergic overstimulation” is experienced.
A repeat dose of slightly less than the sum of the combined dosing to reach “mild
adrenergic overstimulation” could then be used as a therapeutic dose, every 3-4 hours,
with expectations of increasing this dose over 1-2 weeks, until no adrenergic
dysregulation is experienced by the patient at any dose, indicating metabolic adaptations
have been completed. This dose will need to be maintained until all related antibodies
have cleared and hepatic latent cell burden is resolved. Additional dosing may be
required, relative to significant increases in physical activity.
In HASD CFS/ME, before succinate treatment has restored normal metabolism, an
excess dose of succinate could be expected to cause temporary effects such as mild
dizziness and/or elevated heart rate, due to previous compensatory metabolic
adaptations, an effect which could be potentially useful as a blunt diagnostics tool.
During early treatment for HASD patients (with or without CFS/ME), acutely increased
immune response as indicated by skin rashes and inflammation in tissues with latent
cells would be expected.
GABA (+ Arginine)
GABA and arginine (NOT α-ketoglutarate form)
are both available as dietary
supplements in many countries and could help provide an exogenous route to help
restore GABA levels and receptor homeostasis[49], particularly during initial treatment.
These supplements also provide a backup path to synthesising succinate, should
physical activity levels exceed available intracellular succinate, ultimately caused by an
-20-
insufficient succinate dosing schedule. An excess dose of GABA may cause a temporary
flushing / skin tingling effect, or a mild sedative effect.
Based on the HASD CFS/ME model expectations for diurnal elevation of
α-KG->Glutamate, a speculative dose range could be 2 x 500mg - 750mg, taken in the
afternoon / evening, only if excess glutamate is observed. A fast-acting β-adrenergic
agonist could also assist with glutamate scavenging[37]. (see: "β2-Adrenergic Cascade /
β2-Adrenergic Agonists (short-acting)")
Arginine is required to assist transport of GABA across the blood-brain barrier[141]. Based
on published metabolomics data and HASD CFS/ME model expectations, if the patient is
already treating α-KGDH deficiency via anaplerosis at succinate, serum arginine may
already be sufficiently elevated. As indicated by “Plasma Amino Acids” pathology, a
suggested dose range is 250-500mg, taken in the afternoon / evening.
B12 / Methylcobalamin
Active + inactive serum B12 sufficiency should be assessed and resolved via diet and/or
therapeutic intervention, as B12 could be easily depleted during any metabolism to
succinate[80].
Alpha-Lipoic Acid
(see entry under “PDH Deficiency Interventions / Alpha-Lipoic Acid (ALA)”)
NAD+
Indications could include:
Absence of energy, where dietary sufficiency and dosing of succinate and β2-adrenergic
cascade therapeutic interventions are optimal.
NAD+, or precursors Nicotinamide Riboside (NR), Nicotinamide Mononucleotide (NMN)
and/or Niacin (Nicotinic Acid / Vitamin B3) would be a required cofactor in treating
α-KGDH deficiency via anaplerosis and the dosing schedule would relate to physical
activity levels. At this time, NMN has been suggested as providing a superior method of
maintaining NAD+ pools over a longer duration[67][212][213][218].
Based on the HASD CFS/ME model, a suggested dosing of NMN would be 125-250mg,
3 times per day and also before any sustained exercise periods.
PDH Deficiency Interventions
Resolving PDH deficiency will prevent unwanted lactate generation during energy
production. PDH kinase inhibition and/or PDH promotion, along with appropriate
-21-
progressive increases to daily activity will further increase PDH levels via metabolic
adaptations.
Excess lactate generated from an “inappropriate” amount of physical activity, relative to
PDH levels, would be first noticed as intense, burning muscle pains which linger. This
could be largely prevented, in future, by administration of a suitable PDH promoter or
PDHK inhibitor, such as ones described herein.
Burning muscle pain would be followed initially by delayed upper-right abdominal pain
(liver), headache swelling and general inflammation, if hepatic gluconeogenesis is
simultaneously impaired. (See also
: “β2-Adrenergic Cascade”
)
NB. The upper-right abdominal pain should be considered as a “critical state”, which
needs immediate remediation via a fast-acting beta2-adrenergic agonist to prevent lactic
acidosis and hyperammonemia symptoms - following any upper right abdominal pain, it
would be expected that headaches, neck inflammation, hot or swollen distal extremities
will be experienced.
Remarkably, a rapid intervention is already commonplace and is dosed via an inhaler,
for another autoimmune disease - asthma. (see: "β2-Adrenergic Cascade /
β2-Adrenergic Agonists (short-acting)")
A further psychological challenge, from a patient education perspective, is that
increasing PDH and improving liver metabolism function, to further progress their
recovery will require a lengthy adaptation time and appropriate levels of basic exercise,
such as walking. Exercise can induce excess lactate and cause systemic pains, in
absence of appropriate treatments. “Learning the limits”, then progressively and
appropriately challenging those limits each day / week / month is a necessary activity to
increase their lactate metabolism efficiency.
Using a ‘step-counter’ may be a useful tool in objectively tracking progress.
Dichloroacetate (DCA)
Dichloroacetate has been demonstrated as a highly effective PDHK inhibitor in clinical
trials for cancers and has been successfully used for congenital lactic acidosis since the
1990s[149][151].
Common side effects reported include peripheral neuropathy, delusions, cognitive
impairment, mood swings and gastrointestinal upset, at doses of 6.25-12.5mg/kg, twice
per day. It has been reported that patients with GSTZ1 polymorphisms metabolise DCA
twice as fast and therefore require twice the standard dose for cancer treatments,
however it was mentioned that PDHK inhibition was still achieved[96][97][148]. This also
suggests a lower dose of DCA may be effective for HASD CFS/ME, with a more
side-effect friendly profile.
-22-
It has been suggested from case reports that mitigations for these side effects can be
achieved by use of vitamin B1 (as benfotiamine / thiamine), alpha lipoic acid, L-carnitine
and a proton pump, such as pantoprazole[68][133][148][150]. The anaplerotic healing of CAC
efficiency / proton pumps as provided by succinate would further suggest additional use
of pantoprazole could be unnecessary.
Resveratrol
Resveratrol has been shown to be a potent PDHK inhibitor and tumor suppressor, with a
mild side-effect profile. It has also demonstrated some ability to increase expression of
proteins required to form Complexes I-V, suggesting therapeutic benefit for some
inherited mitochondrial diseases. High doses have been known to cause diarrhea and
neuropathy[98][142][152][163].
Based on available pharmacokinetics data, dosing at 500-750mg, three times a day with
meals would yield a suitable steady-state for PDK inhibition, providing gastrointestinal
upsets or neuropathy are not experienced[142][152].
L-Carnitine
(Not to be confused with Acetyl-L-Carnitine.)
L-Carnitine deficiency in mitochondrial myopathy disorders is a well-known cause for
reduced PDH and lactate clearance issues[160]. L-Carnitine and acetyl-CoA is reversibly
converted to acetyl-carnitine by carnitine acetyltransferase (CAT)[159]. The ratio of CoA to
acetyl-CoA limits PDH kinase.
Care should be taken, as the acetylated form “Acetyl-L-Carnitine” would induce the
opposite effect
[161].
Common dosing schedules for L-Carnitine range from 7-15mg/kg/day.
Alpha-Lipoic Acid (ALA)
(Not to be confused with flaxseed derivatives / linolenic acid or linoleic acid.)
ALA is available in multiple forms - of the two enantiomers, (R)-(+)-lipoic acid (RLA) and
(S)-(-)-lipoic acid (SLA), RLA is suggested as superior. It’s also available as a mixture,
(R/S)-lipoic acid (R/S-LA). ALA is noted for being a PDH promoter, tumor metastasis
suppressor and also protects α-KGDH from oxidative stress[36][150][162][171].
Case reports and pharmacokinetic data would suggest an optimal dose range in HASD
CFS/E is 250-400mg, 3 times per day and before sustained exercise.
-23-
Exercise
Exercise plays a crucial role in favourable metabolic adaptations to PDH and PDHK
expression. Studies have shown 1 week of aerobic exercise is insufficient for these
adaptations, whereas 8 weeks of consistently exercising conferred beneficial results[191].
Hyperlactatemia Crisis Intervention
Indications could include:
Onset of upper right abdomen pain (liver), followed by neuralgia, systemic inflammation
and/or hot, swollen hands and feet.
B2-Adrenergic Agonists
ß2-adrenergic agonists provide rapid intervention and prevention to hyperlactatemia.
Therapeutic options are readily available via inhalers, as short-acting interventions for
the treatment of asthma. (see: “β2-Adrenergic Cascade / β2-Adrenergic Agonists
(short-acting).”)
Sodium Bicarbonate
Sodium bicarbonate is routinely used in the treatment of lactic and metabolic acidosis,
however in the HASD CFS/ME model, would likely provide little benefit, as the pH levels
may be already balanced by concomitant ammonia production[75].
β2-Adrenergic Cascade
Indications could include:
“Insensitivity to β-oxidation” - “feeling cold” / no energy on waking or fasting.
“Hyperlactatemia” - swelling, inflammation, liver pain, headache / encephalopathy.
β2-Adrenergic Agonists (short-acting)
For autoimmune diseases, such as asthma, there are a number of short-acting
beta-adrenergic agonists available for rapid and convenient dosing, in the form of
inhalers, with effects that typically last 4-6 hours[229]. These are sometimes supplied as
prescription or OTC in different countries.
Commonly used β2-Adrenergic Agonists include Salbutamol (albuterol), levalbuterol and
adrenaline. They provide metered doses at typically 100-200 mcg and are used 3-5
times a day, or as needed.
Common side-effects can include over-stimulation, agitation, panic, tachycardia.
In HASD CFS/ME, dosing would likely approximate schedules for asthma.
Therapeutic intervention targeting, or combined with, cAMP and/or AMPK may be
preferable.
-24-
β2-Adrenergic Agonists (long-acting)
Complementary to short-acting β2-adrenergic agonists, long-acting β2-adrenergic
agonists provide extended period of agonism. Commonly prescribed examples are
salmeterol, formoterol, with metered doses[230].
Common side-effects can include over-stimulation, agitation, panic, tachycardia.
In HASD CFS/ME, dosing would likely approximate schedules for asthma.
Therapeutic intervention targeting, or combined with, cAMP and/or AMPK may be
preferable.
cAMP
Forskolin
Forskolin, an extract of coleus forskohlii, is a highly potent cAMP promoter and
has been demonstrated to significantly enhance nerve regeneration and
positively alter immune responses related to asthma and chronic obstructive
pulmonary disorder. Important to the HASD CFS/ME model, downstream of
cAMP, forskolin increases hepatic gluconeogenesis and mitochondrial biogenesis
via PGC-1α[233]. Forskolin’s effects are significantly improved when combined with
a PDE4 inhibitor[231][232][234]. Forskolin also increases CYP3A activity, which may
be a consideration for some drug-drug interactions[235].
Commonly used doses range from 2.5 - 60mg of the active extract, up to 3 times
per day, with positive effects on body composition[236].
In the HASD CFS/ME model, a suggested dose could be 5-15mg of the active
extract, 3 times per day.
Exercise
Exercise plays a significant role in positively altering fatty acid oxidation efficiency via
beta-adrenergic signalling. An appropriate, consistent, daily exercise routine is a critical
requirement for metabolic recovery from the impairments induced by CFS/ME and may
take many months[237][238].
Transport protein CD36 appears to also be a critical player in these adaptations and
could be worthy of further exploration[239].
The challenge in HASD CFS / ME is self-identifying the appropriate amount of exercise
and progressively increasing it. Excessive activity would induce ammonia and glutamate
burden, relative to UC efficiency. Afternoon naps may be initially required. Without
appropriate symptomatic management, it would also induce hyperlactatemia and other
challenges.
-25-
H1 Antagonists
Ketotifen
Ketotifen is a H1 antagonist and mast cell stabilizer that has been demonstrated
to upregulate expression of β2-adrenergic receptors and be an effective drug in
treating asthma. Ketotifen is available both as OTC ocular drops and as tablets,
for treating allergic reactions[240][241][242][243].
In HASD CFS/ME, ketotifen could be useful as a means of resensitising
β2-adrenergic receptors following dysregulation by antibodies, metabolic
impairments or excessive use of β2-adrenergic agonists.
Common side effects include drowsiness, at higher doses. An evening dosing
schedule could be considered.
Corydalis Ternata
Benzylisoquinoline alkaloids and other compounds from corydalis ternata, such as
berberine, coptisine and protopine have been demonstrated to antagonise H1 receptors
(increasing β2 receptor expression), plus increase GDH and AMPK activity[202][203][204].
While protopine may be beneficial for altering Cori Cycle and α-KG metabolism
efficiency, it exhibits mild anticholinergic effects, which may prove problematic as a
therapeutic intervention in HASD CFS/ME[209].
Muscarinic Acetylcholine Receptors
Indications could include:
1. Muscle spasms / cramping.
2. Myopathy other than post-exercise lactate.
Choline
In the general adult population and relative to gender, the recommended daily choline
intake is approximately 400-550mg / day with a reported tolerable upper limit of >3.5g /
day. Frequent anecdotal reports indicate excessive intake of choline has been
associated with depressive effects, in a subset of the population. Dietary choline comes
mostly from organ meats, eggs, and soy lecithin[153].
In the HASD CFS/ME model, daily choline intake should be relative to activity levels,
with the minimum intake increased to 750mg and consumed across 3-4 servings per day
to maintain optimal systemic levels against metabolic half-life and expected 3-5 hour
digestion + absorption delays[62][210].
Choline from dietary supplements like choline bitartrate may be useful in maintaining
sufficiency. Attention should be given to any digestion and pharmacokinetic data of any
supplement to maintain systemic sufficiency.
-26-
Endocrine Disruption - Resolving Adrenal Cortex Dominance
Human Chorionic Gonadotropin (hCG)
It is expected that the described HASD therapy over 6-8 weeks would restore normal
endocrine function via amelioration of the impairment cascade, as observable by a
remission of symptoms relating to endocrine disruption, including those resembling PFS
and remission of tissue and follicle abnormalities such alopecia, PCOS or endometriosis.
This would be quantified in male patients by testing 24 hour urine cortisol, DHT,
Adrenocorticotropic Hormone (ACTH), Dehydroepiandrosterone (DHEAS), LH, FSH,
Testosterone (T), Sex-Hormone Binding Globulin (SHBG), Free T, OE2, Thyroid
Stimulating Hormone (TSH) and Prolactin (PRL).
In males, if 24 hour urine cortisol, serum DHEAS and DHT are in mid-normal / normal
range, yet LH and OE2 have not concomitantly increased to normal ‘mid-range’ values
(LH > 4-7IU/L, OE2 > 90-120pmol/l), a therapeutic endocrine “challenge” could be
performed, via a 4 week intervention of parenteral injections of the LH analogue, hCG, to
restore oestrogen dominance over other HPG feedback inputs - overriding
allopregnanolone and DHT[68].
Extrapolation from older “min/max” peer-reviewed literature correlates with more recently
published anecdotal case report data[201] suggesting that a single 150IU dose of hCG
provides a serum LH analogue with a Cmax of approximately 1IU/L and a Tmax of 8 hours.
Further, a 500IU dose of hCG provides a serum LH analogue with a Cmax of
approximately 3IU/L and a Tmax of 36 hours[195][196][197][198][199][200].
Therefore, in males, a suggested therapeutic serum hCG Cmax target for each of the 4
weeks could be:
Week 1 - 6IU/L (as 1000IU, once, eg. on day 1)
Weeks 2,3 - 3IU/L (as 500IU hCG, every 3.5 days, eg. on days 7, 10.5, 14, 17.5)
Week 4 - 1.5IU/L (as 250IU, every 2 days, eg. days 21, 23, 25, 27)
Mildly elevated oestradiol (130-150pmol/L) during week 1 is expected and desired. Minor
and temporary symptoms, such as nipple sensitivity and alterations to libido may be
noticed during week 1 and 2. Retest serum markers at 3 weeks following cessation of
treatment.
Neurotransmitter Homeostasis
Normalisation of Glutamate, Glutamine and GABA
Cabrera-Pastor et al., 2019 reported on the effects of increasing cyclic guanosine
monophosphate (cGMP) to reverse a plethora of neurotransmitter imbalances caused by
“hyperammonemia, hepatic encephalopathy or Alzheimer's disease”[49].
-27-
Their work indicated therapeutic uses for:
1. Phosphodiesterase 5 (PDE5) inhibitors, such as tadalafil, sildenafil, zaprinast, to
reduce metabolism of cGMP. These could be incompatible in HASD CFS/ME due
to decreased gluconeogenesis[249].
2. GABA modulating neurosteroids, such as pregnenolone, allopregnanolone -
which are also increased by endogenous LH levels or hCG therapy.
3. GABA-A receptors antagonists (bicuculline).
4. Inhibitors of cyclooxygenase, such as ibuprofen, for reducing neuroinflammation.
Polygala Tenuifolia
Research has shown that Polygala can exert agonistic effects on β-adrenergic,
α-adrenergic, D & D dopamine and 5-HT(2A) receptors[205][206]. This may prove
beneficial against impairments caused by HASD CFS/ME antibodies for these or related
targets[128].
Taurine
Taurine has been demonstrated to reduce production of catecholamines in hypertensive
patients, which may be relevant to some HASD CFS/ME patients[244].
Tyrosine
Tyrosine is a key intermediate in catecholamine synthesis pathways. It has also been
identified as a target for heterophile antibodies in CFS/ME[128]. Exogenous tyrosine exerts
pressure on catecholamine production and may be beneficial in restoring impaired
neurotransmitter homeostasis[207][208].
-28-
Further Research is Needed for Potentially Related Diseases
This paper is intended to allow a better understanding of herpesviridae, which can be used and
extended by all. This model outlines key pathways only and more research is needed.
If this model is validated and proven robust through clinical trials, then the HASD signature of
mitochondrial and metabolic disturbances described in this paper suggests significant research
is needed into exploring a large number of diseases and disorders that display a related profile
of metabolic alterations and impairments.
Based on the evidence presented in this paper, it is the authors’ opinion that many common
diseases and disorders are simply the same ubiquitous herpesviridae infection(s), presenting
different symptoms based on the location and burden of the latent cells and any level of lytic
activity.
We hope to see further research in attempting to apply this model to -
Multiple Sclerosis[74][94][138], Parkinson’s Disease[94][260][261], Alzheimer’s Disease[94][183][15], ALS[94][266],
bipolar disorder[63][105], schizophrenia[101][102][103][105][248], anxiety disorders[256], depression[128][257],
narcolepsy[128], sleeping disorders[49][50][51], eating disorders[49], fibromyalgia[43], fibrosis[267],
rheumatoid arthritis[44][138][264], psoriatic arthritis[44][264], polycystic ovary syndrome[185],
endometriosis[96], mast cell activation syndrome (MCAS)[99][243], eczema and other skin
disorders[44][253][254][255], alopecia[92][185], hirsutism[252], colitis[44][258], irritable bowel
disorder[44][65][154][155][262], coeliac disease[259], gut microbiome disorders with elevated butyrate[154],
Raynaud’s Syndrome[265], Ehlers Danlos Syndrome[250][251], Addison’s Disease[268], Hashimoto’s
Disease[269], Lupus / SLE[138][265], myasthenia gravis[270], primary biliary cirrhosis[3][16][26][27][30],
NAFLD[54][107][168], Type-1[271] diabetes, Leigh Disease[45][135], Reye Syndrome[45][273], Fumarase
Deficiency Syndrome[249], mitochondrial diseases where genomic sequencing is negative,
lymphomas[33][44][72][179][189][190], multiple myeloma[44][148], food allergies[272], lactic acidosis[45],
congenital lactic acidosis[45][263], Sudden Infant Death Syndrome (SIDS)[45][172][263],
colic[45][154][155][263], cancers featuring the “Warburg” or “Reverse Warburg” effect[44], “Long COVID”
syndrome[177][178], as well as dislipidemia, insulin resistance, hypogonadism, Post Finasteride
Syndrome and Post-SSRI sexual dysfunction.
-29-
Acknowledgements
With special thanks to J Carlson and S Asnani.
Data Availability Statement
All materials used have been cited. Images were created in Draw.IO and source files to extend
this model are available upon request to the corresponding author.
Author Contributions
JL conceived the design and authored the manuscript. AN reviewed methodology, provided
oversight and guidance, expert lab experience, analysis skills, audited / validated citations and
edited the manuscript. All authors critically reviewed the final manuscript.
Funding
This work was unfunded and conducted out of human interest.
Conflict of Interest
The authors declare that the research was conducted in the absence of any commercial or
financial relationships that could be construed as a potential conflict of interest. Authors have
joint-filed a patent relating to the formulation of a treatment protocol.
-30-
Reference List
-31-
1. Avellaneda Fernández A, Pérez Martín Á, Izquierdo
Martínez M et al. Chronic fatigue syndrome: aetiology,
diagnosis and treatment. BMC Psychiatry
. 2009;9(Suppl 1):S1.
doi:10.1186/1471-244x-9-s1-s1
2. Bell D. Illness Onset Characteristics in Children with Chronic
Fatigue Syndrome and Idiopathic Chronic Fatigue. Journal of
Chronic Fatigue Syndrome
. 1997;3(2):43-51.
doi:10.1300/j092v03n02_05
3. Fukushima N, Nakamura M, Matsui M et al. Establishment
and structural analysis of human mAb to the E2 component of
the 2-oxoglutarate dehydrogenase complex generated from a
patient with primary biliary cirrhosis. Int Immunol
.
1995;7(7):1047-1055. doi:10.1093/intimm/7.7.1047
4. Glasgow A. Clinical Application of Blood Ammonia
Determinations. Lab Med
. 1981;12(3):151-157.
doi:10.1093/labmed/12.3.151
5. Germain A, Ruppert D, Levine S, Hanson M. Prospective
Biomarkers from Plasma Metabolomics of Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome Implicate Redox
Imbalance in Disease Symptomatology. Metabolites
.
2018;8(4):90. doi:10.3390/metabo8040090
6. Richard P, Hilditch S. d-Galacturonic acid catabolism in
microorganisms and its biotechnological relevance. Appl
Microbiol Biotechnol
. 2009;82(4):597-604.
doi:10.1007/s00253-009-1870-6
7. Pötzsch A, Zocher S, Bernas S, Leiter O, Rünker A,
Kempermann G. L-lactate exerts a pro-proliferative effect on
adult hippocampal precursor cells in vitro. iScience
.
2021;24(2):102126. doi:10.1016/j.isci.2021.102126
8. Berg J, Tymoczko J, Stryer L. Biochemistry
. 5th ed. New
York: W.H. Freeman and Co.; 2002:Chapter 17. Available
from: https://www.ncbi.nlm.nih.gov/books/NBK21163/
9. Missailidis D, Annesley S, Allan C et al. An Isolated
Complex V Inefficiency and Dysregulated Mitochondrial
Function in Immortalized Lymphocytes from ME/CFS Patients.
Int J Mol Sci
. 2020;21(3):1074. doi:10.3390/ijms21031074
10. OTT P, CLEMMESEN O, LARSEN F. Cerebral metabolic
disturbances in the brain during acute liver failure: From
hyperammonemia to energy failure and proteolysis.
Neurochem Int
. 2005;47(1-2):13-18.
doi:10.1016/j.neuint.2005.04.002
11. The Role of Citric Acid in Intermediate Metabolism in
Animal Tissues. A Source Book in Chemistry, 1900-1950
.
1968:383-390. doi:10.4159/harvard.9780674366701.c143
12. Thomson A. THE ROYAL COLLEGE OF PHYSICIANS
REPORT ON ALCOHOL: GUIDELINES FOR MANAGING
WERNICKE'S ENCEPHALOPATHY IN THE ACCIDENT AND
EMERGENCY DEPARTMENT. Alcohol and Alcoholism
.
2002;37(6):513-521. doi:10.1093/alcalc/37.6.513
13. Meyer T, Hostetter T. Approaches to Uremia. Journal of the
American Society of Nephrology
. 2014;25(10):2151-2158.
doi:10.1681/asn.2013121264
14. Kimura H, Cohen J. Chronic Active Epstein–Barr Virus
Disease. Front Immunol
. 2017;8.
doi:10.3389/fimmu.2017.01867
15. Xu S, Gaskin F. Increased incidence of anti-β-amyloid
autoantibodies secreted by Epstein-Barr virus transformed B
cell lines from patients with Alzheimer's disease. Mech Ageing
Dev
. 1997;94(1-3):213-222.
doi:10.1016/s0047-6374(96)01861-1
16. Matsui M, Nakamura M, Ishibashi H, Koike K, Kudo J, Niho
Y. Human monoclonal antibodies from a patient with primary
biliary cirrhosis that recognize two distinct autoepitopes in the
E2 component of the pyruvate dehydrogenase complex.
Hepatology
. 1993;18(5):1069-1077.
doi:10.1002/hep.1840180509
17. Verma D, Church T, Swaminathan S. Epstein–Barr virus
co-opts TFIIH component XPB to specifically activate essential
viral lytic promoters. Proceedings of the National Academy of
Sciences
. 2020;117(23):13044-13055.
doi:10.1073/pnas.2000625117
18. SPIRONOLACTONE AS TREATMENT FOR CHRONIC
FATIGUE SYNDROME IN PATIENTS WITH POSITIVE
EPSTEIN BAR VIRUS SEROLOGY. Intern Med J.
2020;50(S1):19-19. doi:10.1111/imj.11_14849
19. Walker M, van der Donk W. The many roles of glutamate in
metabolism. J Ind Microbiol Biotechnol
. 2015;43(2-3):419-430.
doi:10.1007/s10295-015-1665-y
20. Gropman A, Prust M, Breeden A, Fricke S, VanMeter J.
Urea cycle defects and hyperammonemia: effects on functional
imaging. Metab Brain Dis
. 2012;28(2):269-275.
doi:10.1007/s11011-012-9348-0
21. Cruzat V, Macedo Rogero M, Noel Keane K, Curi R,
Newsholme P. Glutamine: Metabolism and Immune Function,
Supplementation and Clinical Translation. Nutrients
.
2018;10(11):1564. doi:10.3390/nu10111564
22. Qi F, Pradhan R, Dash R, Beard D. Detailed kinetics and
regulation of mammalian 2-oxoglutarate dehydrogenase. BMC
Biochem
. 2011;12(1):53. doi:10.1186/1471-2091-12-53
23. Al Gawwam G, Sharquie I. Serum Glutamate Is a Predictor
for the Diagnosis of Multiple Sclerosis. The Scientific World
Journal
. 2017;2017:1-5. doi:10.1155/2017/9320802
-32-
24. Faff-Michalak L, Albrecht J. The two catalytic components
of the 2-oxoglutarate dehydrogenase complex in rat cerebral
synaptic and nonsynaptic mitochondria: Comparison of the
response to in vitro treatment with ammonia,
hyperammonemia, and hepatic encephalopathy. Neurochem
Res
. 1993;18(2):119-123. doi:10.1007/bf01474673
25. Smith B, Clotfelter L, Cheung J, LaNoue K. Differences in
2-oxoglutarate dehydrogenase regulation in liver and kidney.
Biochemical Journal
. 1992;284(3):819-826.
doi:10.1042/bj2840819
26. Bassendine M, Jones D, Yeaman S. Biochemistry and
Autoimmune Response to the 2-Oxoacid Dehydrogenase
Complexes in Primary Biliary Cirrhosis. Semin Liver Dis
.
1997;17(01):49-60. doi:10.1055/s-2007-1007182
27. Teoh K, Rowley M, Zafirakis H et al. Enzyme inhibitory
autoantibodies to pyruvate dehydrogenase complex in primary
biliary cirrhosis: Applications of a semiautomated assay.
Hepatology
. 1994;20(5):1220-1224.
doi:10.1002/hep.1840200518
28. Moteki S, Amaki S, Nakano E. [The association between
autoantibodies to enzyme and diseases--with special reference
to antibodies to pyruvate dehydrogenase complex (PDH)].
Rinsho Byori. 1998 Apr;46(4):317-23. Japanese. PMID:
9594621.
29. Fluge Ø, Rekeland I, Lien K et al. B-Lymphocyte Depletion
in Patients With Myalgic Encephalomyelitis/Chronic Fatigue
Syndrome. Ann Intern Med
. 2019;170(9):585.
doi:10.7326/m18-1451
30. Mutimer D, Fussey S, Yeaman S, Kelly P, James O,
Bassendine M. Frequency of IgG and IgM autoantibodies to
four specific M2 mitochondrial autoantigens in primary biliary
cirrhosis. Hepatology
. 1989;10(4):403-407.
doi:10.1002/hep.1840100402
31. Chiu Y, Tung C, Lee Y et al. A comprehensive library of
mutations of Epstein–Barr virus. Journal of General Virology
.
2007;88(9):2463-2472. doi:10.1099/vir.0.82881-0
32. Roberts M, Luxembourg A, Cooper N. Epstein--Barr virus
binding to CD21, the virus receptor, activates resting B cells
via an intracellular pathway that is linked to B cell infection.
Journal of General Virology
. 1996;77(12):3077-3085.
doi:10.1099/0022-1317-77-12-3077
33. Elsayed A, Asano N, Ohshima K, Izutsu K, Kinoshita T,
Nakamura S. Prognostic significance of CD20 expression and
Epstein-Barr virus (EBV) association in classical Hodgkin
lymphoma in Japan: A clinicopathologic study. Pathol Int
.
2014;64(7):336-345. doi:10.1111/pin.12175
34. Curran K, Kernan N, Wang X et al. CD19 Targeted
Allogeneic EBV-Specific T Cells for the Treatment of Relapsed
ALL in Pediatric Patients Post HSCT. Blood
.
2012;120(21):353-353. doi:10.1182/blood.v120.21.353.353
35. Wirth K, Scheibenbogen C. A Unifying Hypothesis of the
Pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue
Syndrome (ME/CFS): Recognitions from the finding of
autoantibodies against ß2-adrenergic receptors. Autoimmun
Rev
. 2020;19(6):102527. doi:10.1016/j.autrev.2020.102527
36. McLain A, Cormier P, Kinter M, Szweda L.
Glutathionylation of α-ketoglutarate dehydrogenase: The
chemical nature and relative susceptibility of the cofactor lipoic
acid to modification. Free Radical Biology and Medicine
.
2013;61:161-169. doi:10.1016/j.freeradbiomed.2013.03.020
37. Leibowitz A, Boyko M, Shapira Y, Zlotnik A. Blood
Glutamate Scavenging: Insight into Neuroprotection. Int J Mol
Sci
. 2012;13(8):10041-10066. doi:10.3390/ijms130810041
38. Pournourmohammadi S, Grimaldi M, Stridh M et al.
Epigallocatechin-3-gallate (EGCG) activates AMPK through
the inhibition of glutamate dehydrogenase in muscle and
pancreatic ß-cells: A potential beneficial effect in the
pre-diabetic state?. Int J Bioc
2hem Cell Biol
. 2017;88:220-225.
doi:10.1016/j.biocel.2017.01.01
39. Chin R, Fu X, Pai M et al. The metabolite α-ketoglutarate
extends lifespan by inhibiting ATP synthase and TOR. Nature
.
2014;510(7505):397-401. doi:10.1038/nature13264
40. Hussain S. Is spironolactone safe for dialysis patients?.
Nephrology Dialysis Transplantation
. 2003;18(11):2364-2368.
doi:10.1093/ndt/gfg413
41. Matsumoto Y, Kageyama S, Yakushigawa T et al.
Long-Term Low-Dose Spironolactone Therapy Is Safe in
Oligoanuric Hemodialysis Patients. Cardiology
.
2009;114(1):32-38. doi:10.1159/000210553
42. Sweetman E, Kleffmann T, Edgar C, de Lange M, Vallings
R, Tate W. A SWATH-MS analysis of Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome peripheral blood
mononuclear cell proteomes reveals mitochondrial dysfunction.
J Transl Med
. 2020;18(1). doi:10.1186/s12967-020-02533-3
43. Ribeiro G, Scola R, Piovesan E et al. A importância de
ácido láctico na enxaqueca e na fibromialgia. Rev Bras
Reumatol
. 2015;55(6):471-476. doi:10.1016/j.rbr.2015.02.002
44. Krishna G, Soman Pillai V, Valiya Veettil M. Upregulation of
GLS1 Isoforms KGA and GAC Facilitates Mitochondrial
Metabolism and Cell Proliferation in Epstein–Barr Virus
Infected Cells. Viruses
. 2020;12(8):811.
doi:10.3390/v12080811
45. Guffon N, Lopez-Mediavilla C, Dumoulin R et al.
2-Ketoglutarate dehydrogenase deficiency, a rare cause of
primary hyperlactataemia: Report of a new case. J Inherit
Metab Dis
. 1993;16(5):821-830. doi:10.1007/bf00714273
46. Gibson G, Park L, Sheu K, Blass J, Calingasan N. The
α-ketoglutarate dehydrogenase complex in neurodegeneration.
Neurochem Int
. 2000;36(2):97-112.
doi:10.1016/s0197-0186(99)00114-x
47. Kiss G, Konrad C, Doczi J et al. The negative impact of α
-ketoglutarate dehydrogenase complex deficiency on matrix
substrate-level phosphorylation. The FASEB Journal
.
2013;27(6):2392-2406. doi:10.1096/fj.12-220202
48. Liang S, Carlson G, Coulter D. Dynamic Regulation of
Synaptic GABA Release by the Glutamate-Glutamine Cycle in
Hippocampal Area CA1. Journal of Neuroscience
.
2006;26(33):8537-8548. doi:10.1523/jneurosci.0329-06.2006
-33-
49. Cabrera-Pastor A, Arenas Y, Taoro-Gonzalez L, Montoliu
C, Felipo V. Chronic hyperammonemia alters extracellular
glutamate, glutamine and GABA and membrane expression of
their transporters in rat cerebellum. Modulation by extracellular
cGMP. Neuropharmacology
. 2019;161:107496.
doi:10.1016/j.neuropharm.2019.01.011
50. Ono D, Honma K, Yanagawa Y, Yamanaka A, Honma S.
GABA in the suprachiasmatic nucleus refines circadian output
rhythms in mice. Commun Biol
. 2019;2(1).
doi:10.1038/s42003-019-0483-6
51. Cardinali D, Golombek D. The Rhythmic GABAergic
System. Neurochem Res
. 1998;23(5):607-614.
doi:10.1023/a:1022426519297
52. Certo M, Tsai C, Pucino V, Ho P, Mauro C. Lactate
modulation of immune responses in inflammatory versus
tumour microenvironments. Nature Reviews Immunology
.
2020. doi:10.1038/s41577-020-0406-2
53. M J, Stachowicz K, Nowak G, Pilc A. The Loss of
Glutamate-GABA Harmony in Anxiety Disorders. Anxiety
Disorders
. 2011. doi:10.5772/19919
54. Holecek M. Evidence of a vicious cycle in glutamine
synthesis and breakdown in pathogenesis of hepatic
encephalopathy–therapeutic perspectives. Metab Brain Dis
.
2013;29(1):9-17. doi:10.1007/s11011-013-9428-9
55. Ciccarelli M, Santulli G, Pascale V, Trimarco B, Iaccarino
G. Adrenergic receptors and metabolism: role in development
of cardiovascular disease. Front Physiol
. 2013;4.
doi:10.3389/fphys.2013.00265
56. Katewa S, Khanna A, Kapahi P. Mitobolites: The Elixir of
Life. Cell Metab
. 2014;20(1):8-9.
doi:10.1016/j.cmet.2014.06.013
57. Kim M, Kim H. The Roles of Glutamine in the Intestine and
Its Implication in Intestinal Diseases. Int J Mol Sci
.
2017;18(5):1051. doi:10.3390/ijms18051051
58. Krishna Rao R. Role of Glutamine in Protection of Intestinal
Epithelial Tight Junctions. Journal of Epithelial Biology and
Pharmacology
. 2012;5(1):47-54.
doi:10.2174/1875044301205010047
59. Cascarano J, Ades I, O'Connor J. Hypoxia: A
succinate-fumarate electron shuttle between peripheral cells
and lung. Journal of Experimental Zoology
.
1976;198(2):149-153. doi:10.1002/jez.1401980204
60. Zdzisińska B, Żurek A, Kandefer-Szerszeń M.
Alpha-Ketoglutarate as a Molecule with Pleiotropic Activity:
Well-Known and Novel Possibilities of Therapeutic Use. Arch
Immunol Ther Exp (Warsz)
. 2016;65(1):21-36.
doi:10.1007/s00005-016-0406-x
61. Brooks G. The Science and Translation of Lactate Shuttle
Theory. Cell Metab
. 2018;27(4):757-785.
doi:10.1016/j.cmet.2018.03.008
62. Kuo I, Ehrlich B. Signaling in Muscle Contraction. Cold
Spring Harb Perspect Biol
. 2015;7(2):a006023.
doi:10.1101/cshperspect.a006023
63. Yoshimi N, Futamura T, Kakumoto K et al. Blood
metabolomics analysis identifies abnormalities in the citric acid
cycle, urea cycle, and amino acid metabolism in bipolar
disorder. BBA Clin
. 2016;5:151-158.
doi:10.1016/j.bbacli.2016.03.008
64. van Campen C, Rowe P, Verheugt F, Visser F. Numeric
Rating Scales Show Prolonged Post-exertional Symptoms
After Orthostatic Testing of Adults With Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome. Front Med
(Lausanne)
. 2021;7. doi:10.3389/fmed.2020.602894
65. Pérez-Escuredo J, Dadhich R, Dhup S et al. Lactate
promotes glutamine uptake and metabolism in oxidative cancer
cells. Cell Cycle
. 2016;15(1):72-83.
doi:10.1080/15384101.2015.1120930
66. Patel K, O'Brien T, Subramony S, Shuster J, Stacpoole P.
The spectrum of pyruvate dehydrogenase complex deficiency:
Clinical, biochemical and genetic features in 371 patients. Mol
Genet Metab
. 2012;106(3):385-394.
doi:10.1016/j.ymgme.2012.03.017
67. Sugden M, Holness M. Recent advances in mechanisms
regulating glucose oxidation at the level of the pyruvate
dehydrogenase complex by PDKs. American Journal of
Physiology-Endocrinology and Metabolism
.
2003;284(5):E855-E862. doi:10.1152/ajpendo.00526.2002
68. Constantin-Teodosiu D. Regulation of Muscle Pyruvate
Dehydrogenase Complex in Insulin Resistance: Effects of
Exercise and Dichloroacetate. Diabetes Metab J
.
2013;37(5):301. doi:10.4093/dmj.2013.37.5.301
69. Aalling N, Nedergaard M, DiNuzzo M. Cerebral Metabolic
Changes During Sleep. Curr Neurol Neurosci Rep
. 2018;18(9).
doi:10.1007/s11910-018-0868-9
70. Ravasz D, Kacso G, Fodor V, Horvath K, Adam-Vizi V,
Chinopoulos C. Catabolism of GABA, succinic semialdehyde
or gamma-hydroxybutyrate through the GABA shunt impair
mitochondrial substrate-level phosphorylation. Neurochem Int
.
2017;109:41-53. doi:10.1016/j.neuint.2017.03.008
71. Lessard S, Rivas D, Chen Z et al. Impaired Skeletal Muscle
β-Adrenergic Activation and Lipolysis Are Associated with
Whole-Body Insulin Resistance in Rats Bred for Low Intrinsic
Exercise Capacity. Endocrinology
. 2009;150(11):4883-4891.
doi:10.1210/en.2009-0158
72. Mo X, Wei F, Tong Y et al. Lactic Acid Downregulates Viral
MicroRNA To Promote Epstein-Barr Virus-Immortalized B
Lymphoblastic Cell Adhesion and Growth. J Virol
. 2018;92(9).
doi:10.1128/jvi.00033-18
73. Kojima K, Nagayama R, Hirama S et al. Epstein-Barr virus
infection resembling autoimmune hepatitis with lactate
dehydrogenase and alkaline phosphatase anomaly. J
Gastroenterol
. 1999;34(6):706-712.
doi:10.1007/s005350050324
-34-
74. Buljevac D. Epstein-Barr virus and disease activity in
multiple sclerosis. Journal of Neurology, Neurosurgery &
Psychiatry
. 2005;76(10):1377-1381.
doi:10.1136/jnnp.2004.048504
75. Adamczak M, Masajtis-Zagajewska A, Mazanowska O,
Madziarska K, Stompór T, Więcek A. Diagnosis and Treatment
of Metabolic Acidosis in Patients with Chronic Kidney Disease
– Position Statement of the Working Group of the Polish
Society of Nephrology. Kidney and Blood Pressure Research
.
2018;43(3):959-969. doi:10.1159/000490475
76. Fluge Ø, Mella O, Bruland O et al. Metabolic profiling
indicates impaired pyruvate dehydrogenase function in myalgic
encephalopathy/chronic fatigue syndrome. JCI Insight
.
2016;1(21). doi:10.1172/jci.insight.89376
77. Chinopoulos C. From Glucose to Lactate and Transiting
Intermediates Through Mitochondria, Bypassing Pyruvate
Kinase: Considerations for Cells Exhibiting Dimeric PKM2 or
Otherwise Inhibited Kinase Activity. Front Physiol
. 2020;11.
doi:10.3389/fphys.2020.543564
78. Naviaux R, Naviaux J, Li K et al. Metabolic features of
chronic fatigue syndrome. Proceedings of the National
Academy of Sciences
. 2016;113(37):E5472-E5480.
doi:10.1073/pnas.1607571113
79. Halarnkar P, Blomquist G. Comparative aspects of
propionate metabolism. Comparative Biochemistry and
Physiology Part B: Comparative Biochemistry
.
1989;92(2):227-231. doi:10.1016/0305-0491(89)90270-8
80. Tracey T, Steyn F, Wolvetang E, Ngo S. Neuronal Lipid
Metabolism: Multiple Pathways Driving Functional Outcomes in
Health and Disease. Front Mol Neurosci
. 2018;11.
doi:10.3389/fnmol.2018.00010
81. Yamaoka M, Hara T, Kusaka M. Overcoming Persistent
Dependency on Androgen Signaling after Progression to
Castration-Resistant Prostate Cancer: Fig. 1. Clinical Cancer
Research
. 2010;16(17):4319-4324.
doi:10.1158/1078-0432.ccr-10-0255
82. Lee E, Nam Y, Kang S et al. The local
hypothalamic–pituitary–adrenal axis in cultured human dermal
papilla cells. BMC Mol Cell Biol
. 2020;21(1).
doi:10.1186/s12860-020-00287-w
83. Bouissou P, Fiet J, Guezennec C, Pesquies P. Plasma
adrenocorticotrophin and cortisol responses to acute hypoxia
at rest and during exercise. Eur J Appl Physiol Occup Physiol
.
1988;57(1):110-113. doi:10.1007/bf00691248
84. COSTE O, BEERS P, BOGDAN A, CHARBUY H,
TOUITOU Y. Hypoxic alterations of cortisol circadian rhythm in
man after simulation of a long duration flight. Steroids
.
2005;70(12):803-810. doi:10.1016/j.steroids.2005.05.003
85. Toufexis D, Wilson M. Dihydrotestosterone differentially
modulates the cortisol response of the
hypothalamic–pituitary–adrenal axis in male and female rhesus
macaques, and restores circadian secretion of cortisol in
females. Brain Res
. 2012;1429:43-51.
doi:10.1016/j.brainres.2011.10.024
86. Bodó E, van Beek N, Naumann V et al. Modulation of
Chemotherapy-Induced Human Hair Follicle Damage by 17-β
Estradiol and Prednisolone: Potential Stimulators of Normal
Hair Regrowth by “Dystrophic Catagen” Promotion?. Journal of
Investigative Dermatology
. 2009;129(2):506-509.
doi:10.1038/jid.2008.228
87. Acevedo-Rodriguez A, Kauffman A, Cherrington B, Borges
C, Roepke T, Laconi M. Emerging insights into
hypothalamic-pituitary-gonadal axis regulation and interaction
with stress signalling. J Neuroendocrinol
. 2018;30(10):e12590.
doi:10.1111/jne.12590
88. Matsumura H, Mohri Y, Binh N et al. Hair follicle aging is
driven by transepidermal elimination of stem cells via
COL17A1 proteolysis. Science (1979)
.
2016;351(6273):aad4395-aad4395.
doi:10.1126/science.aad4395
89. Choe S, Kim D, Kim E et al. Psychological Stress
Deteriorates Skin Barrier Function by Activating
11β-Hydroxysteroid Dehydrogenase 1 and the HPA Axis. Sci
Rep
. 2018;8(1). doi:10.1038/s41598-018-24653-z
90. Barrault C, Garnier J, Pedretti N et al. Androgens induce
sebaceous differentiation in sebocyte cells expressing a stable
functional androgen receptor. J Steroid Biochem Mol Biol
.
2015;152:34-44. doi:10.1016/j.jsbmb.2015.04.005
91. Nasiri M, Nikolaou N, Parajes S et al. 5α-Reductase Type
2 Regulates Glucocorticoid Action and Metabolic Phenotype in
Human Hepatocytes. Endocrinology
. 2015;156(8):2863-2871.
doi:10.1210/en.2015-1149
92. Sharpley C, McFarlane J, Slominski A. Stress-linked
cortisol concentrations in hair: what we know and what we
need to know. Rev Neurosci
. 2012;23(1).
doi:10.1515/rns.2011.058
93. Lee E, Nam Y, Kang S et al. The local
hypothalamic–pituitary–adrenal axis in cultured human dermal
papilla cells. BMC Mol Cell Biol
. 2020;21(1).
doi:10.1186/s12860-020-00287-w
94. Chen H, Denton T, Xu H, Calingasan N, Beal M, Gibson G.
Reductions in the mitochondrial enzyme α-ketoglutarate
dehydrogenase complex in neurodegenerative disease -
beneficial or detrimental?. J Neurochem
. 2016;139(5):823-838.
doi:10.1111/jnc.13836
95. Harley J, Chen X, Pujato M et al. Transcription factors
operate across disease loci, with EBNA2 implicated in
autoimmunity. Nat Genet
. 2018;50(5):699-707.
doi:10.1038/s41588-018-0102-3
96. Horne A, Ahmad S, Carter R et al. Repurposing
dichloroacetate for the treatment of women with endometriosis.
Proceedings of the National Academy of Sciences
.
2019;116(51):25389-25391. doi:10.1073/pnas.1916144116
97. Michelakis E, Webster L, Mackey J. Dichloroacetate (DCA)
as a potential metabolic-targeting therapy for cancer. Br J
Cancer
. 2008;99(7):989-994. doi:10.1038/sj.bjc.6604554
-35-
98. Saunier E, Antonio S, Regazzetti A et al. Resveratrol
reverses the Warburg effect by targeting the pyruvate
dehydrogenase complex in colon cancer cells. Sci Rep
.
2017;7(1). doi:10.1038/s41598-017-07006-0
99. Hagemann P, Nsiah-Dosu S, Hundt J, Hartmann K,
Orinska Z. Modulation of Mast Cell Reactivity by Lipids: The
Neglected Side of Allergic Diseases. Front Immunol
. 2019;10.
doi:10.3389/fimmu.2019.01174
100. Hong T, Parameswaran S, Donmez O et al. Epstein-Barr
virus nuclear antigen 2 (EBNA2) extensively rewires the
human chromatin landscape at autoimmune risk loci. 2020.
PREPRINT - doi:10.1101/2020.04.15.043612
101. Esaki K, Balan S, Iwayama Y et al. Evidence for Altered
Metabolism of Sphingosine-1-Phosphate in the Corpus
Callosum of Patients with Schizophrenia. Schizophr Bull
.
2020;46(5):1172-1181. doi:10.1093/schbul/sbaa052
102. Fantini J, Barrantes F. Sphingolipid/cholesterol regulation
of neurotransmitter receptor conformation and function.
Biochimica et Biophysica Acta (BBA) - Biomembranes
.
2009;1788(11):2345-2361. doi:10.1016/j.bbamem.2009.08.016
103. Baier C, Barrantes F. Sphingolipids are necessary for
nicotinic acetylcholine receptor export in the early secretory
pathway. J Neurochem
. 2007;101(4):1072-1084.
doi:10.1111/j.1471-4159.2007.04561.x
104. The signaling lipid sphingosine 1-phosphate regulates
mechanical pain. eLife 2018;7:e33285
. 2018.
doi:10.7554/elife.33285.001
105. Olsen A, Færgeman N. Sphingolipids: membrane
microdomains in brain development, function and neurological
diseases. Open Biol
. 2017;7(5):170069.
doi:10.1098/rsob.170069
106. Rosenhouse-Dantsker A, Mehta D, Levitan I. Regulation
of Ion Channels by Membrane Lipids. Compr Physiol
. 2012.
doi:10.1002/cphy.c110001
107. Ivanovski I, Ješić M, Ivanovski A, Garavelli L, Ivanovski P.
Metabolically based liver damage pathophysiology in patients
with urea cycle disorders - A new hypothesis. World J
Gastroenterol
. 2017;23(44):7930-7938.
doi:10.3748/wjg.v23.i44.7930
108. Yamano E, Sugimoto M, Hirayama A et al. Index markers
of chronic fatigue syndrome with dysfunction of TCA and urea
cycles. Sci Rep
. 2016;6(1). doi:10.1038/srep34990
109. Taylor G, Long H, Brooks J, Rickinson A, Hislop A. The
Immunology of Epstein-Barr Virus–Induced Disease. Annu Rev
Immunol
. 2015;33(1):787-821.
doi:10.1146/annurev-immunol-032414-112326
110. Hutt-Fletcher L. Epstein–Barr virus replicating in epithelial
cells. Proceedings of the National Academy of Sciences
.
2014;111(46):16242-16243. doi:10.1073/pnas.1418974111
111. Sica D. Pharmacokinetics and Pharmacodynamics of
Mineralocorticoid Blocking Agents and their Effects on
Potassium Homeostasis. Heart Fail Rev
. 2005;10(1):23-29.
doi:10.1007/s10741-005-2345-1
112. Nuttall F, Warrier R, Gannon M. Gynecomastia and drugs:
a critical evaluation of the literature. Eur J Clin Pharmacol
.
2015;71(5):569-578. doi:10.1007/s00228-015-1835-x
113. Huffman D, Kampmann J, Hignite C, Azarnoff D.
Gynecomastia induced in normal males by spironolactone.
Clinical Pharmacology & Therapeutics
. 1978;24(4):465-473.
doi:10.1002/cpt1978244465
114. Polat S, Cuhaci N, Evranos B, Ersoy R, Cakir B.
Gynecomastia: Clinical evaluation and management. Indian J
Endocrinol Metab
. 2014;18(2):150.
doi:10.4103/2230-8210.129104
115. de Ronde W, de Jong F. Aromatase inhibitors in men:
effects and therapeutic options. Reproductive Biology and
Endocrinology
. 2011;9(1):93. doi:10.1186/1477-7827-9-93
116. Du Y, Yu J, Du L, Tang J, Feng W. Cordycepin enhances
Epstein–Barr virus lytic infection and Epstein–Barr
virus-positive tumor treatment efficacy by doxorubicin. Cancer
Lett
. 2016;376(2):240-248. doi:10.1016/j.canlet.2016.04.001
117. Ryu E, Son M, Lee M et al. Cordycepin is a novel
chemical suppressor of Epstein-Barr virus replication.
Oncoscience
. 2014;1(12):866-881.
doi:10.18632/oncoscience.110
118. Zihlif M, Mahmoud I, Ghanim M et al. Thymoquinone
Efficiently Inhibits the Survival of EBV-Infected B Cells and
Alters EBV Gene Expression. Integr Cancer Ther
.
2012;12(3):257-263. doi:10.1177/1534735412458827
119. Drosu N, Edelman E, Housman D. Tenofovir prodrugs
potently inhibit Epstein–Barr virus lytic DNA replication by
targeting the viral DNA polymerase. Proceedings of the
National Academy of Sciences
. 2020;117(22):12368-12374.
doi:10.1073/pnas.2002392117
120. Wassner C, Bradley N, Lee Y. A Review and Clinical
Understanding of Tenofovir: Tenofovir Disoproxil Fumarate
versus Tenofovir Alafenamide. Journal of the International
Association of Providers of AIDS Care (JIAPAC)
.
2020;19:232595822091923. doi:10.1177/2325958220919231
121. van Zyl D, Tsai M, Shumilov A et al. Immunogenic
particles with a broad antigenic spectrum stimulate cytolytic T
cells and offer increased protection against EBV infection ex
vivo and in mice. PLoS Pathog
. 2018;14(12):e1007464.
doi:10.1371/journal.ppat.1007464
122. Giorgi-Coll S, Amaral A, Hutchinson P, Kotter M,
Carpenter K. Succinate supplementation improves metabolic
performance of mixed glial cell cultures with mitochondrial
dysfunction. Sci Rep
. 2017;7(1).
doi:10.1038/s41598-017-01149-w
123. Rajčáni J, Kúdelová M. Gamma herpesviruses:
Pathogenesis of infection and cell signaling. Folia Microbiol
(Praha)
. 2003;48(3):291-318. doi:10.1007/bf02931360
124. Kúdelová M, Štibrániová I. Murine Gammaherpesvirus 68
(MHV-68), a Newly Discovered Tick Borne Virus. Ticks and
Tick-Borne Pathogens
. 2019. doi:10.5772/intechopen.81025
-36-
125. Hricová M, Mistríková J. Murine gammaherpesvirus 68
serum antibodies in general human population. Acta Virol
.
2007;51(4):283-287.
126. Losman J, Koivunen P, Kaelin W.
2-Oxoglutarate-dependent dioxygenases in cancer. Nature
Reviews Cancer
. 2020;20(12):710-726.
doi:10.1038/s41568-020-00303-3
127. Chen H, Bartee M, Yaron J et al. Mouse Gamma
Herpesvirus MHV-68 Induces Severe Gastrointestinal (GI)
Dilatation in Interferon Gamma Receptor-Deficient Mice
(IFNγR−/−) That Is Blocked by Interleukin-10. Viruses
.
2018;10(10):518. doi:10.3390/v10100518
128. Nilsson I, Palmer J, Apostolou E et al. Metabolic
Dysfunction in Myalgic Encephalomyelitis/Chronic Fatigue
Syndrome Not Due to Anti-mitochondrial Antibodies. Front Med
(Lausanne)
. 2020;7. doi:10.3389/fmed.2020.00108
129. Fedotcheva N, Sokolov A, Kondrashova M. Nonezymatic
formation of succinate in mitochondria under oxidative stress.
Free Radical Biology and Medicine
. 2006;41(1):56-64.
doi:10.1016/j.freeradbiomed.2006.02.012
130. Scheibenbogen C, Loebel M, Freitag H et al.
Immunoadsorption to remove ß2 adrenergic receptor
antibodies in Chronic Fatigue Syndrome CFS/ME. PLoS One
.
2018;13(3):e0193672. doi:10.1371/journal.pone.0193672
131. Pall M. Levels of Nitric Oxide Synthase Product Citrulline
Are Elevated in Sera of Chronic Fatigue Syndrome Patients.
Journal of Chronic Fatigue Syndrome
. 2002;10(3-4):37-41.
doi:10.1300/j092v10n03_04
132. Fuhrmann D, Olesch C, Kurrle N et al. Chronic Hypoxia
Enhances β-Oxidation-Dependent Electron Transport via
Electron Transferring Flavoproteins. Cells
. 2019;8(2):172.
doi:10.3390/cells8020172
133. Khan A, Andrews D, Blackburn A. Long-term stabilization
of stage 4 colon cancer using sodium dichloroacetate therapy.
World J Clin Cases
. 2016;4(10):336.
doi:10.12998/wjcc.v4.i10.336
134. Pollard P, Brière J, Alam N et al. Accumulation of Krebs
cycle intermediates and over-expression of HIF1α in tumours
which result from germline FH and SDH mutations. Hum Mol
Genet
. 2005;14(15):2231-2239. doi:10.1093/hmg/ddi227
135. Soreze Y, Boutron A, Habarou F et al. Mutations in
human lipoyltransferase gene LIPT1 cause a Leigh disease
with secondary deficiency for pyruvate and alpha-ketoglutarate
dehydrogenase. Orphanet J Rare Dis
. 2013;8(1):192.
doi:10.1186/1750-1172-8-192
136. Hoffman B, Wang Y, Feldman E, Tibbetts S. Epstein–Barr
virus EBER1 and murine gammaherpesvirus TMER4 share
conserved in vivo function to promote B cell egress and
dissemination. Proceedings of the National Academy of
Sciences
. 2019;116(51):25392-25394.
doi:10.1073/pnas.1915752116
137. Harley JB, James JA. Epstein-Barr virus infection induces
lupus autoimmunity. Bull NYU Hosp Jt Dis.
2006;64(1-2):45-50. PMID: 17121489.
138. Lossius A, Johansen J, Torkildsen Ø, Vartdal F, Holmøy
T. Epstein-Barr Virus in Systemic Lupus Erythematosus,
Rheumatoid Arthritis and Multiple Sclerosis—Association and
Causation. Viruses
. 2012;4(12):3701-3730.
doi:10.3390/v4123701
139. James J, Robertson J. Lupus and Epstein-Barr. Curr Opin
Rheumatol
. 2012;24(4):383-388.
doi:10.1097/bor.0b013e3283535801
140. Song Z, Wei B, Lu C, Li P, Chen L. Glutaminase sustains
cell survival via the regulation of glycolysis and glutaminolysis
in colorectal cancer. Oncol Lett
. 2017;14(3):3117-3123.
doi:10.3892/ol.2017.6538
141. Shyamaladevi N, Jayakumar A, Sujatha R, Paul V,
Subramanian E. Evidence that nitric oxide production
increases γ-amino butyric acid permeability of blood-brain
barrier. Brain Res Bull
. 2002;57(2):231-236.
doi:10.1016/s0361-9230(01)00755-9
142. SERGIDES C, CHIRILĂ M, SILVESTRO L, PITTA D,
PITTAS A. Bioavailability and safety study of resveratrol 500
mg tablets in healthy male and female volunteers. Exp Ther
Med
. 2015;11(1):164-170. doi:10.3892/etm.2015.2895
143. Maevsky EI, Peskov AB, Uchitel ML, Pogorelov AG,
Saharova NY, Vihlyantseva EF, Bogdanova LA, Kondrashova
MN. A succinate-based composition reverses menopausal
symptoms without sex hormone replacement therapy. Adv
Gerontol. 2008;21(2):298-305. PMID: 18942377.
144. Radzinsky V, Uspenskaya Y, Shulman L, Kuznetsova I.
Succinate-Based Dietary Supplement for Menopausal
Symptoms: A Pooled Analysis of Two Identical Randomized,
Double-Blind, Placebo-Controlled Clinical Trials. Obstet
Gynecol Int
. 2019;2019:1-9. doi:10.1155/2019/1572196
145. Nulton-Persson A, Szweda L, Sadek H. Inhibition of
Cardiac Mitochondrial Respiration by Salicylic Acid and
Acetylsalicylate. J Cardiovasc Pharmacol
. 2004;44(5):591-595.
doi:10.1097/00005344-200411000-00012
146. Martens M, Lee C. Reye's syndrome: Salicylates and
mitochondrial functions. Biochem Pharmacol
.
1984;33(18):2869-2876. doi:10.1016/0006-2952(84)90209-0
147. Wieduwild E, Girard-Madoux M, Quatrini L et al.
β2-adrenergic signals downregulate the innate immune
response and reduce host resistance to viral infection. Journal
of Experimental Medicine
. 2020;217(4).
doi:10.1084/jem.20190554
148. Tian D, Bennett S, Coupland L et al. GSTZ1 genotypes
correlate with dichloroacetate pharmacokinetics and chronic
side effects in multiple myeloma patients in a pilot phase 2
clinical trial. Pharmacol Res Perspect
. 2019;7(6).
doi:10.1002/prp2.526
149. Tataranni T, Piccoli C. Dichloroacetate (DCA) and
Cancer: An Overview towards Clinical Applications. Oxid Med
Cell Longev
. 2019;2019:1-14. doi:10.1155/2019/8201079
150. Khan A, Andrews D, Shainhouse J, Blackburn A.
Long-term stabilization of metastatic melanoma with sodium
dichloroacetate. World J Clin Oncol
. 2017;8(4):371.
doi:10.5306/wjco.v8.i4.371
-37-
151. Stacpoole P, Barnes C, Hurbanis M, Cannon S, Kerr D.
Treatment of congenital lactic acidosis with dichloroacetate.
Arch Dis Child
. 1997;77(6):535-541. doi:10.1136/adc.77.6.535
152. Wang P, Sang S. Metabolism and pharmacokinetics of
resveratrol and pterostilbene. BioFactors
. 2018;44(1):16-25.
doi:10.1002/biof.1410
153. Fischer L, daCosta K, Kwock L et al. Sex and
menopausal status influence human dietary requirements for
the nutrient choline. Am J Clin Nutr
. 2007;85(5):1275-1285.
doi:10.1093/ajcn/85.5.1275
154. Bourriaud C, Robins R, Martin L et al. Lactate is mainly
fermented to butyrate by human intestinal microfloras but
inter-individual variation is evident. J Appl Microbiol
.
2005;99(1):201-212. doi:10.1111/j.1365-2672.2005.02605.x
155. Pham V, Lacroix C, Braegger C, Chassard C.
Lactate-utilizing community is associated with gut microbiota
dysbiosis in colicky infants. Sci Rep
. 2017;7(1).
doi:10.1038/s41598-017-11509-1
156. McLain A, Szweda P, Szweda L. α-Ketoglutarate
dehydrogenase: A mitochondrial redox sensor. Free Radic
Res
. 2010;45(1):29-36. doi:10.3109/10715762.2010.534163
157. Schreiner P, Harrer T, Scheibenbogen C et al. Human
Herpesvirus-6 Reactivation, Mitochondrial Fragmentation, and
the Coordination of Antiviral and Metabolic Phenotypes in
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome.
Immunohorizons
. 2020;4(4):201-215.
doi:10.4049/immunohorizons.2000006
158. Zizzo M, Serio R. Therapeutic Potential of the Gabaergic
System in Ulcerative Colitis: Current Status and Perspectives.
Dig Dis Sci
. 2017;62(10):2780-2780.
doi:10.1007/s10620-017-4709-5
159. Ferrari R., Visioli O. (1995) Carnitine and lactate
metabolism. In: De Jong J.W., Ferrari R. (eds) The Carnitine
System. Developments in Cardiovascular Medicine, vol 162.
Springer, Dordrecht. doi:10.1007/978-94-011-0275-9_15
160. 8. Campos Y, Huertas R, Bautista J et al. Muscle carnitine
deficiency and lipid storage myopathy in patients with
mitochondrial myopathy. Muscle Nerve
. 1993;16(7):778-781.
doi:10.1002/mus.880160713
161. Claessens Y, Cariou A, Chiche J, Dauriat G, Dhainaut J.
l-Carnitine as a treatment of life-threatening lactic acidosis
induced by nucleoside analogues. AIDS
. 2000;14(4):472.
doi:10.1097/00002030-200003100-00029
162. Feuerecker B, Pirsig S, Seidl C et al. Lipoic acid inhibits
cell proliferation of tumor cells in vitro and in vivo. Cancer Biol
Ther
. 2012;13(14):1425-1435. doi:10.4161/cbt.22003
163. Lopes Costa A, Le Bachelier C, Mathieu L et al. Beneficial
effects of resveratrol on respiratory chain defects in patients'
fibroblasts involve estrogen receptor and estrogen-related
receptor alpha signaling. Hum Mol Genet
.
2013;23(8):2106-2119. doi:10.1093/hmg/ddt603
165. Tretter L. Generation of Reactive Oxygen Species in the
Reaction Catalyzed by -Ketoglutarate Dehydrogenase.
Journal of Neuroscience
. 2004;24(36):7771-7778.
doi:10.1523/jneurosci.1842-04.2004
166. Dunckelmann R, Ebinger F, Schulze A, Wanders R,
Rating D, Mayatepek E. 2-Ketoglutarate Dehydrogenase
Deficiency with Intermittent 2-Ketoglutaric Aciduria.
Neuropediatrics
. 2000;31(1):35-38. doi:10.1055/s-2000-15295
167. Tretter L, Adam-Vizi V. Alpha-ketoglutarate
dehydrogenase: a target and generator of oxidative stress.
Philosophical Transactions of the Royal Society B: Biological
Sciences
. 2005;360(1464):2335-2345.
doi:10.1098/rstb.2005.1764
168. Oliveira C, da Costa Gayotto L, Tatai C et al. Vitamin C
and Vitamin E in Prevention of Nonalcoholic Fatty Liver
Disease (NAFLD) in Choline Deficient Diet Fed Rats. Nutr J
.
2003;2(1). doi:10.1186/1475-2891-2-9
169. Roccamo A, Pediconi M, Aztiria E, Zanello L,
Wolstenholme A, Barrantes F. Cells defective in sphingolipids
biosynthesis express low amounts of muscle nicotinic
acetylcholine receptor. European Journal of Neuroscience
.
1999;11(5):1615-1623. doi:10.1046/j.1460-9568.1999.00574.x
170. Fantini J, Barrantes F. Sphingolipid/cholesterol regulation
of neurotransmitter receptor conformation and function.
Biochimica et Biophysica Acta (BBA) - Biomembranes
.
2009;1788(11):2345-2361. doi:10.1016/j.bbamem.2009.08.016
171. Teichert J, Hermann R, Ruus P, Preiss R. Plasma
Kinetics, Metabolism, and Urinary Excretion of Alpha-Lipoic
Acid following Oral Administration in Healthy Volunteers. The
Journal of Clinical Pharmacology
. 2003;43(11):1257-1267.
doi:10.1177/0091270003258654
172. Olpin S. The metabolic investigation of sudden infant
death. Ann Clin Biochem
. 2004;41(4):282-293.
doi:10.1258/0004563041201473
173. Luczkowska K, Stekelenburg C, Sloan-Béna F et al.
Hyperinsulinism associated with GLUD1 mutation: allosteric
regulation and functional characterization of p.G446V
glutamate dehydrogenase. Hum Genomics
. 2020;14(1).
doi:10.1186/s40246-020-00262-8
174. Green H, Brewer A. Dysregulation of
2-oxoglutarate-dependent dioxygenases by hyperglycaemia:
does this link diabetes and vascular disease?. Clin
Epigenetics
. 2020;12(1). doi:10.1186/s13148-020-00848-y
175. Zhu M, Fang J, Zhang J et al. Biomolecular Interaction
Assays Identified Dual Inhibitors of Glutaminase and
Glutamate Dehydrogenase That Disrupt Mitochondrial
Function and Prevent Growth of Cancer Cells. Anal Chem
.
2017;89(3):1689-1696. doi:10.1021/acs.analchem.6b03849
176. Johmura Y, Yamanaka T, Omori S et al. Senolysis by
glutaminolysis inhibition ameliorates various age-associated
disorders. Science (1979)
. 2021;371(6526):265-270.
doi:10.1126/science.abb5916
-38-
177. Sumbria D, Berber E, Mathayan M, Rouse B. Virus
Infections and Host Metabolism—Can We Manage the
Interactions?. Front Immunol
. 2021;11.
doi:10.3389/fimmu.2020.594963
178. Lopez-Leon S, Wegman-Ostrosky T, Perelman C et al.
More than 50 Long-term effects of COVID-19: a systematic
review and meta-analysis. 2021.
doi:10.1101/2021.01.27.21250617
179. Ghosh S, Perrine S, Williams R, Faller D. Histone
deacetylase inhibitors are potent inducers of gene expression
in latent EBV and sensitize lymphoma cells to nucleoside
antiviral agents. Blood
. 2012;119(4):1008-1017.
doi:10.1182/blood-2011-06-362434
180. Matés J, Di Paola F, Campos-Sandoval J, Mazurek S,
Márquez J. Therapeutic targeting of glutaminolysis as an
essential strategy to combat cancer. Semin Cell Dev Biol
.
2020;98:34-43. doi:10.1016/j.semcdb.2019.05.012
181. Guardia D, Rolland B, Karila L, Cottencin O. GABAergic
and Glutamatergic Modulation in Binge Eating: Therapeutic
Approach. Curr Pharm Des
. 2011;17(14):1396-1409.
doi:10.2174/138161211796150828
182. Grados M, Atkins E, Kovacikova G, McVicar E. A
selective review of glutamate pharmacological therapy in
obsessive&ndash;compulsive and related disorders. Psychol
Res Behav Manag
. 2015:115. doi:10.2147/prbm.s58601
183. Brothers H, Gosztyla M, Robinson S. The Physiological
Roles of Amyloid-β Peptide Hint at New Ways to Treat
Alzheimer's Disease. Front Aging Neurosci
. 2018;10.
doi:10.3389/fnagi.2018.00118
184. Sugden M, Holness M. The pyruvate
carboxylase-pyruvate dehydrogenase axis in islet pyruvate
metabolism: Going round in circles?. Islets
. 2011;3(6):302-319.
doi:10.4161/isl.3.6.17806
185. Kurzrock R, Cohen P. Polycystic ovary syndrome in men:
Stein–Leventhal syndrome revisited. Med Hypotheses
.
2007;68(3):480-483. doi:10.1016/j.mehy.2006.03.057
186. Yu HJ, Ma H, Green RD. Calcium entry via L-type calcium
channels acts as a negative regulator of adenylyl cyclase
activity and cyclic AMP levels in cardiac myocytes. Mol
Pharmacol. 1993 Oct;44(4):689-93. PMID: 7694067.
187. Charnet P. cAMP-dependent phosphorylation of the
cardiac L-type Ca channel: A missing link?. Biochimie
.
1995;77(12):957-962. doi:10.1016/0300-9084(95)80008-5
188. Liu G, Papa A, Katchman A et al. Mechanism of
adrenergic CaV1.2 stimulation revealed by proximity
proteomics. Nature
. 2020;577(7792):695-700.
doi:10.1038/s41586-020-1947-z
189. Ogawa R, Streiff M, Bugayenko A, Kato G. Inhibition of
PDE4 phosphodiesterase activity induces growth suppression,
apoptosis, glucocorticoid sensitivity, p53, and p21WAF1/CIP1
proteins in human acute lymphoblastic leukemia cells. Blood
.
2002;99(9):3390-3397. doi:10.1182/blood.v99.9.3390
190. Moon E, Lerner A. PDE4 inhibitors activate a
mitochondrial apoptotic pathway in chronic lymphocytic
leukemia cells that is regulated by protein phosphatase 2A.
Blood
. 2003;101(10):4122-4130.
doi:10.1182/blood-2002-10-3208
191. LeBlanc P, Peters S, Tunstall R, Cameron-Smith D,
Heigenhauser G. Effects of aerobic training on pyruvate
dehydrogenase and pyruvate dehydrogenase kinase in human
skeletal muscle. J Physiol
. 2004;557(2):559-570.
doi:10.1113/jphysiol.2003.058263
192. Kennedy A, Pissios P, Otu H et al. A high-fat, ketogenic
diet induces a unique metabolic state in mice. American
Journal of Physiology-Endocrinology and Metabolism
.
2007;292(6):E1724-E1739. doi:10.1152/ajpendo.00717.2006
193. Ogura Y, Kakehashi C, Yoshihara T et al. Ketogenic diet
feeding improves aerobic metabolism property in extensor
digitorum longus muscle of sedentary male rats. PLoS One
.
2020;15(10):e0241382. doi:10.1371/journal.pone.0241382
194. Ang Q, Alexander M, Newman J et al. Ketogenic Diets
Alter the Gut Microbiome Resulting in Decreased Intestinal
Th17 Cells. Cell
. 2020;181(6):1263-1275.e16.
doi:10.1016/j.cell.2020.04.027
195. Coviello A, Matsumoto A, Bremner W et al. Low-Dose
Human Chorionic Gonadotropin Maintains Intratesticular
Testosterone in Normal Men with Testosterone-Induced
Gonadotropin Suppression. The Journal of Clinical
Endocrinology & Metabolism
. 2005;90(5):2595-2602.
doi:10.1210/jc.2004-0802
196. Lee J, Ramasamy R. Indications for the use of human
chorionic gonadotropic hormone for the management of
infertility in hypogonadal men. Transl Androl Urol
.
2018;7(S3):S348-S352. doi:10.21037/tau.2018.04.11
197. Madhusoodanan V, Patel P, Ramasamy R. 233 Human
Chorionic Gonadotropin (hCG) Monotherapy for the Treatment
of Symptoms of Hypogonadism in Men with Total Testosterone
> 300 ng/Dl. J Sex Med
. 2019;16(4):S113.
doi:10.1016/j.jsxm.2019.01.240
198. Roth M, Page S, Lin K et al. Dose-Dependent Increase in
Intratesticular Testosterone by Very Low-Dose Human
Chorionic Gonadotropin in Normal Men with Experimental
Gonadotropin Deficiency. The Journal of Clinical
Endocrinology & Metabolism
. 2010;95(8):3806-3813.
doi:10.1210/jc.2010-0360
199. SMALS A, PIETERS G, BOERS G et al. Differential Effect
of Single High Dose and Divided small Dose Administration of
Human Chorionic Gonadotropin on Leydig Cell Steroidogenic
Desensitization *. The Journal of Clinical Endocrinology &
Metabolism
. 1984;58(2):327-331. doi:10.1210/jcem-58-2-327
200. Martikainen H, Alén M, Rahkila P, Vihko R. Testicular
responsiveness to human chorionic gonadotrophin during
transient hypogonadotrophic hypogonadism induced by
androgenic/anabolic steroids in power athletes. J Steroid
Biochem
. 1986;25(1):109-112.
doi:10.1016/0022-4731(86)90288-8
-39-
201. Saya D. Quantitative Serum hCG Case Studies. Defy
Medical.
https://web.archive.org/web/20200924015444/https://www.defy
medical.com/blog/quantitative-serum-hcg-case-studies-150iu-a
nd-500iu. Published 2016.
202. Lee K, Huh J, Choi M et al. Regulation of glutamate level
in rat brain through activation of glutamate dehydrogenase by
Corydalis ternata. Exp Mol Med
. 2005;37(4):371-377.
doi:10.1038/emm.2005.47
203. Kim K, Lee I, Piao C et al. Benzylisoquinoline alkaloids
from the tubers of Corydalis ternata and their cytotoxicity.
Bioorg Med Chem Lett
. 2010;20(15):4487-4490.
doi:10.1016/j.bmcl.2010.06.035
204. Kim K, Lee I, Choi S et al. New Triterpenoids from the
Tubers of Corydalis ternata:Structural Elucidation and
Bioactivity Evaluation. Planta Med
. 2011;77(13):1555-1558.
doi:10.1055/s-0030-1270781
205. Bettio L, Machado D, Cunha M et al. Antidepressant-like
effect of extract fromPolygala paniculata: Involvement of the
monoaminergic systems. Pharm Biol
. 2011;49(12):1277-1285.
doi:10.3109/13880209.2011.621958
206. Chung I, Moore N, Oh W et al. Behavioural pharmacology
of polygalasaponins indicates potential antipsychotic efficacy.
Pharmacology Biochemistry and Behavior
.
2002;71(1-2):191-195. doi:10.1016/s0091-3057(01)00648-7
207. Daubner S, Le T, Wang S. Tyrosine hydroxylase and
regulation of dopamine synthesis. Arch Biochem Biophys.
2011;508(1):1-12. doi:10.1016/j.abb.2010.12.017
208. Fernstrom J, Fernstrom M. Tyrosine, Phenylalanine, and
Catecholamine Synthesis and Function in the Brain. J Nutr
.
2007;137(6):1539S-1547S. doi:10.1093/jn/137.6.1539s
209. Üstünes L, Laekeman G, Gözler B, Vlietinck A, Özer A,
Herman A. In Vitro Study of the Anticholinergic and
Antihistaminic Activities of Protopine and Some Derivatives. J
Nat Prod
. 1988;51(5):1021-1022. doi:10.1021/np50059a043
210. Zeisel S, da Costa K. Choline: an essential nutrient for
public health. Nutr Rev
. 2009;67(11):615-623.
doi:10.1111/j.1753-4887.2009.00246.x
211. Wiedeman A, Barr S, Green T, Xu Z, Innis S, Kitts D.
Dietary Choline Intake: Current State of Knowledge Across the
Life Cycle. Nutrients
. 2018;10(10):1513.
doi:10.3390/nu10101513
212. Stein L, Imai S. The dynamic regulation of NAD
metabolism in mitochondria. Trends in Endocrinology &
Metabolism
. 2012;23(9):420-428.
doi:10.1016/j.tem.2012.06.005
213. Xiao W, Wang R, Handy D, Loscalzo J. NAD(H) and
NADP(H) Redox Couples and Cellular Energy Metabolism.
Antioxid Redox Signal
. 2018;28(3):251-272.
doi:10.1089/ars.2017.7216
214. Girinathan B, Braun S, Govind R. Clostridium difficile
glutamate dehydrogenase is a secreted enzyme that confers
resistance to H2O2. Microbiology (N Y). 2014;160(1):47-55.
doi:10.1099/mic.0.071365-0
215. Yun B, Oh S, Song M et al. Inhibitory Effect of
Epigallocatechin Gallate on the Virulence ofClostridium
difficilePCR Ribotype 027. J Food Sci
.
2015;80(12):M2925-M2931. doi:10.1111/1750-3841.13145
216. Chambers J, Maguire T, Alwine J. Glutamine Metabolism
Is Essential for Human Cytomegalovirus Infection. J Virol
.
2009;84(4):1867-1873. doi:10.1128/jvi.02123-09
217. Plaitakis A, Kalef-Ezra E, Kotzamani D, Zaganas I,
Spanaki C. The Glutamate Dehydrogenase Pathway and Its
Roles in Cell and Tissue Biology in Health and Disease.
Biology (Basel)
. 2017;6(4):11. doi:10.3390/biology6010011
218. Poddar S, Sifat A, Haque S, Nahid N, Chowdhury S,
Mehedi I. Nicotinamide Mononucleotide: Exploration of Diverse
Therapeutic Applications of a Potential Molecule.
Biomolecules
. 2019;9(1):34. doi:10.3390/biom9010034
219. Lan J, Zhao Y, Dong F et al. Meta-analysis of the effect
and safety of berberine in the treatment of type 2 diabetes
mellitus, hyperlipemia and hypertension. J Ethnopharmacol
.
2015;161:69-81. doi:10.1016/j.jep.2014.09.049
220. Tian Y, Cai J, Gui W et al. Berberine Directly Affects the
Gut Microbiota to Promote Intestinal Farnesoid X Receptor
Activation. Drug Metabolism and Disposition
.
2018;47(2):86-93. doi:10.1124/dmd.118.083691
221. Kawano M, Takagi R, Kaneko A, Matsushita S. Berberine
is a dopamine D1- and D2-like receptor antagonist and
ameliorates experimentally induced colitis by suppressing
innate and adaptive immune responses. J Neuroimmunol
.
2015;289:43-55. doi:10.1016/j.jneuroim.2015.10.001
222. Feng X, Sureda A, Jafari S et al. Berberine in
Cardiovascular and Metabolic Diseases: From Mechanisms to
Therapeutics. Theranostics
. 2019;9(7):1923-1951.
doi:10.7150/thno.30787
223. Yang N, Song Y, Liu C, Cho S, Li X. Pharmacokinetics Of
Berberine, a Bioactive Compound In Butanol Purified Food
Allergy Herbal Formula-2. Journal of Allergy and Clinical
Immunology
. 2014;133(2):AB28.
doi:10.1016/j.jaci.2013.12.127
224. Zhang P, Wang Q, Lin Z, Yang P, Dou K, Zhang R.
Berberine Inhibits Growth of Liver Cancer Cells by
Suppressing Glutamine Uptake. Onco Targets Ther
.
2020;Volume 12:11751-11763. doi:10.2147/ott.s235667
225. McCreight L, Bailey C, Pearson E. Metformin and the
gastrointestinal tract. Diabetologia
. 2016;59(3):426-435.
doi:10.1007/s00125-015-3844-9
226. Rotermund C, Machetanz G, Fitzgerald J. The
Therapeutic Potential of Metformin in Neurodegenerative
Diseases. Front Endocrinol (Lausanne)
. 2018;9.
doi:10.3389/fendo.2018.00400
227. Perez-Lopez F, Pasupuleti V, Gianuzzi X, Palma-Ardiles
G, Hernandez-Fernandez W, Hernandez A. Systematic review
and meta-analysis of the effect of metformin treatment on
overall mortality rates in women with endometrial cancer and
type 2 diabetes mellitus. Maturitas
. 2017;101:6-11.
doi:10.1016/j.maturitas.2017.04.001
-40-
228. Derosa G, D'Angelo A, Romano D, Maffioli P. Effects of
metformin extended release compared to immediate release
formula on glycemic control and glycemic variability in patients
with type 2 diabetes. Drug Des Devel Ther
. 2017;Volume
11:1481-1488. doi:10.2147/dddt.s131670
229. Ameredes B, Calhoun W. Levalbuterol versus albuterol.
Curr Allergy Asthma Rep
. 2009;9(5):401-409.
doi:10.1007/s11882-009-0058-6
230. Stynes G, Svedsater H, Wex J et al. Once-daily
fluticasone furoate/vilanterol 100/25 mcg versus twice daily
combination therapies in COPD – mixed treatment
comparisons of clinical efficacy. Respir Res
. 2015;16(1).
doi:10.1186/s12931-015-0184-8
231. Kilmer S, Carlsen R. Forskolin activation of adenylate
cyclase in vivo stimulates nerve regeneration. Nature
.
1984;307(5950):455-457. doi:10.1038/307455a0
232. Hertz A, Bender A, Smith K et al. Elevated cyclic AMP
and PDE4 inhibition induce chemokine expression in human
monocyte-derived macrophages. Proceedings of the National
Academy of Sciences
. 2009;106(51):21978-21983.
doi:10.1073/pnas.0911684106
233. Lustig Y, Ruas J, Estall J et al. Separation of the
gluconeogenic and mitochondrial functions of PGC-1 through
S6 kinase. Genes Dev
. 2011;25(12):1232-1244.
doi:10.1101/gad.2054711
234. Lim E, Son C. Review of case definitions for myalgic
encephalomyelitis/chronic fatigue syndrome (ME/CFS). J
Transl Med
. 2020;18(1). doi:10.1186/s12967-020-02455-0
235. Ding X, Staudinger J. Induction of Drug Metabolism by
Forskolin: The Role of the Pregnane X Receptor and the
Protein Kinase A Signal Transduction Pathway. Journal of
Pharmacology and Experimental Therapeutics
.
2004;312(2):849-856. doi:10.1124/jpet.104.076331
236. Godard M, Johnson B, Richmond S. Body Composition
and Hormonal Adaptations Associated with Forskolin
Consumption in Overweight and Obese Men. Obes Res
.
2005;13(8):1335-1343. doi:10.1038/oby.2005.162
237. Mika A, Macaluso F, Barone R, Di Felice V, Sledzinski T.
Effect of Exercise on Fatty Acid Metabolism and Adipokine
Secretion in Adipose Tissue. Front Physiol
. 2019;10.
doi:10.3389/fphys.2019.00026
238. Purdom T, Kravitz L, Dokladny K, Mermier C.
Understanding the factors that effect maximal fat oxidation. J
Int Soc Sports Nutr
. 2018;15(1).
doi:10.1186/s12970-018-0207-1
239. Yoshida Y, Jain S, McFarlan J, Snook L, Chabowski A,
Bonen A. Exercise- and training-induced upregulation of
skeletal muscle fatty acid oxidation are not solely dependent
on mitochondrial machinery and biogenesis. J Physiol
.
2012;591(18):4415-4426. doi:10.1113/jphysiol.2012.238451
240. Poller U. Terbutaline-induced desensitization of human
cardiac β2-adrenoceptor-mediated positive inotropic effects:
attenuation by ketotifen. Cardiovasc Res
. 1998;40(1):211-222.
doi:10.1016/s0008-6363(98)00101-1
241. Polson J, Lockey R, Bukantz S, Lowitt S, Krzanowski J,
Szentivanyi A. Effects of ketotifen on the responsiveness of
peripheral blood lymphocyte β-adrenergic receptors. Int J
Immunopharmacol
. 1988;10(6):657-663.
doi:10.1016/0192-0561(88)90019-7
242. Kabra S, Pandey R, Singh R, Seth V. Ketotifen for asthma
in children aged 5 to 15 years: a randomized
placebo-controlled trial. Annals of Allergy, Asthma &
Immunology
. 2000;85(1):46-52.
doi:10.1016/s1081-1206(10)62433-7
243. Nurmatov U, Rhatigan E, Simons F, Sheikh A.
H1-antihistamines for primary mast cell activation syndromes:
a systematic review. Allergy
. 2015;70(9):1052-1061.
doi:10.1111/all.12672
244. Fujita T, Ando K, Noda H, Ito Y, Sato Y. Effects of
increased adrenomedullary activity and taurine in young
patients with borderline hypertension. Circulation
.
1987;75(3):525-532. doi:10.1161/01.cir.75.3.525
245. Omar B, Zmuda-Trzebiatowska E, Manganiello V,
Göransson O, Degerman E. Regulation of AMP-activated
protein kinase by cAMP in adipocytes: Roles for
phosphodiesterases, protein kinase B, protein kinase A, Epac
and lipolysis. Cell Signal
. 2009;21(5):760-766.
doi:10.1016/j.cellsig.2009.01.015
246. Li A, Liu Q, Li Q, Liu B, Yang Y, Zhang N. Berberine
Reduces Pyruvate-driven Hepatic Glucose Production by
Limiting Mitochondrial Import of Pyruvate through
Mitochondrial Pyruvate Carrier 1. EBioMedicine
.
2018;34:243-255. doi:10.1016/j.ebiom.2018.07.039
247. Xu M, Xiao Y, Yin J et al. Berberine Promotes Glucose
Consumption Independently of AMP-Activated Protein Kinase
Activation. PLoS One
. 2014;9(7):e103702.
doi:10.1371/journal.pone.0103702
248. Dickerson F, Jones-Brando L, Ford G et al. Schizophrenia
is Associated With an Aberrant Immune Response to
Epstein–Barr Virus. Schizophr Bull
. 2018;45(5):1112-1119.
doi:10.1093/schbul/sby164
249. Ryder B, Moore F, Mitchell A, Thompson S, Christodoulou
J, Balasubramaniam S. Fumarase Deficiency: A Safe and
Potentially Disease Modifying Effect of High Fat/Low
Carbohydrate Diet. JIMD Rep
. 2017:77-83.
doi:10.1007/8904_2017_65
250. Rowe P, Barron D, Calkins H, Maumenee I, Tong P,
Geraghty M. Orthostatic intolerance and chronic fatigue
syndrome associated with Ehlers-Danlos syndrome. J Pediatr
.
1999;135(4):494-499. doi:10.1016/s0022-3476(99)70173-3
251. Rowe P, Marden C, Flaherty M et al. Impaired Range of
Motion of Limbs and Spine in Chronic Fatigue Syndrome. J
Pediatr. 2014;165(2):360-366. doi:10.1016/j.jpeds.2014.04.051
252. MOORE A, MAGEE F, CUNNINGHAM S, CULLITON M,
McKENNA T. ADRENAL ABNORMALITIES IN IDIOPATHIC
HIRSUTISM. Clin Endocrinol (Oxf)
. 1983;18(4):391-399.
doi:10.1111/j.1365-2265.1983.tb00584.x
-41-
253. Sangueza-Acosta M, Sandoval-Romero E. Epstein-Barr
virus and skin. An Bras Dermatol
. 2018;93(6):786-799.
doi:10.1590/abd1806-4841.20187021
254. Rystedt I, Strannegård I, Strannegård Ö. Increased
Serum Levels of Antibodies to Epstein-Barr Virus in Adults with
History of Atopic Dermatitis. Int Arch Allergy Immunol
.
1984;75(2):179-183. doi:10.1159/000233610
255. Samoylikov P, Gervazieva V, Kozhevnikov S. Association
between autoimmune reactions and severity of atopic
dermatitis in children with herpes virus infection. World Allergy
Organization Journal
. 2013;6:8. doi:10.1186/1939-4551-6-8
256. Fagundes C, Jaremka L, Glaser R et al. Attachment
anxiety is related to Epstein–Barr virus latency. Brain Behav
Immun
. 2014;41:232-238. doi:10.1016/j.bbi.2014.04.002
257. Ford J, Stowe R. Depressive symptoms are associated
with salivary shedding of Epstein-Barr virus in female
adolescents: The role of sex differences.
Psychoneuroendocrinology
. 2017;86:128-133.
doi:10.1016/j.psyneuen.2017.09.009
258. Xu S, Chen H, Zu X et al. Epstein-Barr virus infection in
ulcerative colitis: a clinicopathologic study from a Chinese
area. Therap Adv Gastroenterol
. 2020;13:175628482093012.
doi:10.1177/1756284820930124
259. Perfetti V, Baldanti F, Lenti M et al. Detection of Active
Epstein–Barr Virus Infection in Duodenal Mucosa of Patients
With Refractory Celiac Disease. Clinical Gastroenterology and
Hepatology
. 2016;14(8):1216-1220.
doi:10.1016/j.cgh.2016.03.022
260. Caggiu E, Arru G, Hosseini S et al. Inflammation,
Infectious Triggers, and Parkinson's Disease. Front Neurol
.
2019;10. doi:10.3389/fneur.2019.00122
261. Bu X, Wang X, Xiang Y et al. The association between
infectious burden and Parkinson's disease: A case-control
study. Parkinsonism Relat Disord
. 2015;21(8):877-881.
doi:10.1016/j.parkreldis.2015.05.015
262. Pezhouh M, Miller J, Sharma R et al. Refractory
inflammatory bowel disease: is there a role for Epstein-Barr
virus? A case-controlled study using highly sensitive
Epstein-Barr virus–encoded small RNA1 in situ hybridization.
Hum Pathol
. 2018;82:187-192.
doi:10.1016/j.humpath.2018.08.001
263. Kim Y, Kim H, Park J, Kim C, Kim W. Identification of
Epstein-Barr Virus in the Human Placenta and Its Pathologic
Characteristics. J Korean Med Sci
. 2017;32(12):1959.
doi:10.3346/jkms.2017.32.12.1959
264. Mehraein Y, Lennerz C, Ehlhardt S, Remberger K, Ojak
A, Zang K. Latent Epstein–Barr virus (EBV) infection and
cytomegalovirus (CMV) infection in synovial tissue of
autoimmune chronic arthritis determined by RNA- and DNA-in
situ hybridization. Modern Pathology
. 2004;17(7):781-789.
doi:10.1038/modpathol.3800119
265. Draborg A, Duus K, Houen G. Epstein-Barr Virus in
Systemic Autoimmune Diseases. Clinical and Developmental
Immunology
. 2013;2013:1-9. doi:10.1155/2013/535738
266. Nociti V, Frisullo G, Marti A et al. Epstein-Barr virus
antibodies in serum and cerebrospinal fluid from Multiple
sclerosis, Chronic Inflammatory Demyelinating
Polyradiculoneuropathy and Amyotrophic Lateral Sclerosis. J
Neuroimmunol
. 2010;225(1-2):149-152.
doi:10.1016/j.jneuroim.2010.04.007
267. Marzouk K, Corate L, Saleh S, Sharma O.
Epstein-Barr-virus-induced interstitial lung disease. Curr Opin
Pulm Med
. 2005;11(5):456-460.
doi:10.1097/01.mcp.0000176678.38539.9a
268. Hertel N, Jacobsen B, Pedersen F, Heilmann C.
Adrenocortical insufficiency associated with Epstein-Barr virus
infection in a patient with the Wiskott-Aldrich syndrome. Eur J
Pediatr
. 1987;146(6):603-604. doi:10.1007/bf02467365
269. Assaad S, Meheissen M, Elsayed E, Alnakhal S, Salem T.
Study of Epstein–Barr virus serological profile in Egyptian
patients with Hashimoto’s thyroiditis: A case-control study. J
Clin Transl Endocrinol
. 2020;20:100222.
doi:10.1016/j.jcte.2020.100222
270. Cavalcante P, Serafini B, Rosicarelli B et al. Epstein-barr
virus persistence and reactivation in myasthenia gravis
thymus. Ann Neurol
. 2009:NA-NA. doi:10.1002/ana.21902
271. Yamaguchi Y, Chikuba N, Nakanishi T et al. Production of
islet cell antibodies from Epstein-Barr virus-transformed
peripheral blood lymphocytes in Type 1 (insulin-dependent)
diabetic patients. Diabetologia
. 1991;34(7):511-514.
doi:10.1007/bf00403288
272. Pan Y, Nie Z, Zhang Y, Zhang K, Li J, Wang L.
Identification of EBV infection in adults with egg specific food
allergy. Virol J
. 2013;10(1):9. doi:10.1186/1743-422x-10-9
273. Fleisher G, Schwartz J, Lennette E. Primary Epstein-Barr
virus infection in association with Reye syndrome. J Pediatr
.
1980;97(6):935-937. doi:10.1016/s0022-3476(80)80424-0
FOOTER
-42-
... We recently shared our novel understanding of a rather complicated disease model by submitting a few articles for peer-review and journal publication purposes [1] [2] . This model appears to accurately describe the pathophysiology of a very large number of diseases and disorders in a way that allows them to be treated and potentially cured [1] [2] . ...
... We recently shared our novel understanding of a rather complicated disease model by submitting a few articles for peer-review and journal publication purposes [1] [2] . This model appears to accurately describe the pathophysiology of a very large number of diseases and disorders in a way that allows them to be treated and potentially cured [1] [2] . Largescale testing has not yet been performed. ...
... There have been some early challenges in communicating our understanding of this model, as the language used was unfortunately as complicated as the disease model itself [1] [2] . ...
Preprint
Full-text available
Here we describe the root cause of Chronic Fatigue Syndrome / Myalgic Encephalomyelitis, the growing list of related diseases in the proposed spectrum, related cancers, senescent cells, their previously described mitochondrial / metabolic disturbances and their related pattern of metabolite depletions and compensations as being virally-induced by upregulated expression of protein levels for “GLS1 - KGA, GAC” and “GLUD1”, “GLUD2”. We further describe some practical solutions to this problem, in plain language. [PLEASE NOTE: This is a "special edition", intended for a wider audience and hyperbole contained here may not be present in the final version.] {Errata, second paragraph on Page 9 of 39 should read: First, if α-KG levels are low - converting glutamate into α-KG (while importantly making ammonia and NADH) - this was incorrectly written as NAD+}
... In "The true nature of an autoimmune disease" we hypothesised a complex, extensible model of impaired mitochondrial and metabolic disorders in human herpesvirus (HHV) seropositive CFS/ME as part of a Herpesvirus Autoimmune Spectrum Disorder (HASD) [1] . ...
... The HASD CFS/ME model is primarily based around metabolic impairments caused by alterations to metabolic behaviour in HHV-infected latent hepatic cells, with further complications associated with any lytic activity [1] . ...
... Aspartate, or aspartic acid is a key metabolite for many enzymatic reactions, including those in transamination of α-KG to oxaloacetate [1] [60] . ...
Preprint
Full-text available
Abstract In cells, where mitochondria are utilising glutaminolysis and transamination - whether caused by normal function or when specifically altered by eg. virally induced overexpression of KGA, GLS1, GLUD1 and/or GLUD2 - key enzymatic reactions for energy production are reversed during the transamination of aspartate=>oxaloacetate and α-KG=>glutamate + ammonia, consuming additional pyridoxal 5-phosphate (P5P). Excessive glutaminolysis and transamination increases nitrogen / urea burden, while consuming aspartate, before further consuming acetate and butyrate for increased α-KG=>[..]=>glutamine disposal via the phenylacetylglutamine (PAGN) pathway to urine. Further, this elevates lactate and affects neighbouring cell metabolism via the lactate shuttle. Depletion of acetate could lead to many impaired reactions involving acetyl-CoA, with implications to beta-oxidation pathways, mitochondrial function, pyruvate:lactate balance and any pathways that require an acetyl donor, such as choline=>acetylcholine and carnitine=>acetyl-L-carnitine. Depletion of acetate would cause further dysregulation to the urea cycle (UC), creating an elevation of glutamate, acting as a rate limiting factor for glutaminolysis and further causing impairment of mitochondrial Nicotinamide Adenine Dinucleotide (NAD+):NADH redox, favouring NADH. Aspartate depletion also dysregulates production of reactive oxygen species (ROS), sex hormones, thyroid hormones, alpha-melanocyte-stimulating hormone, GABA and dopamine release. Depletion of butyrate dysregulates immune and mitochondrial apoptotic regulation via Nuclear Factor Kappa B (NF-κB) signalling. It further impacts nitrogen metabolism. The depletion of NAD+ acts as a rate limiting factor for many enzymatic reactions, including glutamate dehydrogenase (GDH), further impairing glutamate<=>α-KG metabolism. We further describe a role for acetate, aspartate, butyrate, P5P and Vitamin B5 in a therapeutic intervention against the HASD model for CFS/ME.
Article
Full-text available
L-lactate has energetic and signaling properties and its availability is modulated by activity-dependent stimuli, which also regulate adult hippocampal neurogenesis. Studying the effects of L-lactate on neural precursor cells (NPCs) in vitro, we found that L-lactate is pro-proliferative and that this effect is dependent on the active lactate transport by monocarboxylate transporters. Increased proliferation was not linked to amplified mitochondrial respiration. Instead, L-lactate deviated glucose metabolism to the pentose phosphate pathway, indicated by increased glucose-6-phosphate dehydrogenase activity while glycolysis decreased. Knockout of Hcar1 revealed that the pro-proliferative effect of L-lactate was not dependent on receptor activity although phosphorylation of ERK1/2 and Akt was increased following L-lactate treatment. Together, we show that availability of L-lactate is linked to the proliferative potential of NPCs and add evidence to the hypothesis that lactate influences cellular homeostatic processes in the adult brain, specifically in the context of adult hippocampal neurogenesis.
Article
Full-text available
When viruses infect cells, they almost invariably cause metabolic changes in the infected cell as well as in several host cell types that react to the infection. Such metabolic changes provide potential targets for therapeutic approaches that could reduce the impact of infection. Several examples are discussed in this review, which include effects on energy metabolism, glutaminolysis and fatty acid metabolism. The response of the immune system also involves metabolic changes and manipulating these may change the outcome of infection. This could include changing the status of herpesviruses infections from productive to latency. The consequences of viral infections which include coronavirus disease 2019 (COVID-19), may also differ in patients with metabolic problems, such as diabetes mellitus (DM), obesity, and endocrine diseases. Nutrition status may also affect the pattern of events following viral infection and examples that impact on the pattern of human and experimental animal viral diseases and the mechanisms involved are discussed. Finally, we discuss the so far few published reports that have manipulated metabolic events in-vivo to change the outcome of virus infection. The topic is expected to expand in relevance as an approach used alone or in combination with other therapies to shape the nature of virus induced diseases.
Preprint
Full-text available
COVID-19, caused by SARS-CoV-2, can involve sequelae and other medical complications that last weeks to months after initial recovery, which has come to be called Long-COVID or COVID long-haulers. This systematic review and meta-analysis aims to identify studies assessing long-term effects of COVID-19 and estimates the prevalence of each symptom, sign, or laboratory parameter of patients at a post-COVID-19 stage. LitCOVID (PubMed and Medline) and Embase were searched by two independent researchers. All articles with original data for detecting long-term COVID-19 published before 1st of January 2021 and with a minimum of 100 patients were included. For effects reported in two or more studies, meta-analyses using a random-effects model were performed using the MetaXL software to estimate the pooled prevalence with 95% CI. Heterogeneity was assessed using I2 statistics. The Preferred Reporting Items for Systematic Reviewers and Meta-analysis (PRISMA) reporting guideline was followed. A total of 18,251 publications were identified, of which 15 met the inclusion criteria. The prevalence of 55 long-term effects was estimated, 21 meta-analyses were performed, and 47,910 patients were included. The follow-up time ranged from 15 to 110 days post-viral infection. The age of the study participants ranged between 17 and 87 years. It was estimated that 80% (95% CI 65-92) of the patients that were infected with SARS-CoV-2 developed one or more long-term symptoms. The five most common symptoms were fatigue (58%), headache (44%), attention disorder (27%), hair loss (25%), and dyspnea (24%). All meta-analyses showed medium (n=2) to high heterogeneity (n=13). In order to have a better understanding, future studies need to stratify by sex, age, previous comorbidities, severity of COVID-19 (ranging from asymptomatic to severe), and duration of each symptom. From the clinical perspective, multi-disciplinary teams are crucial to developing preventive measures, rehabilitation techniques, and clinical management strategies with whole-patient perspectives designed to address long COVID-19 care.
Article
Full-text available
Introduction: Muscle pain, fatigue, and concentration problems are common among individuals with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). These symptoms are commonly increased as part of the phenomenon of postexertional malaise (PEM). An increase in the severity of these symptoms is described following physical or mental exercise in ME/CFS patients. Another important symptom of ME/CFS is orthostatic intolerance, which can be detected by head-up tilt testing (HUT). The effect of HUT on PEM has not been studied extensively. For this purpose, we assessed numeric rating scales (NRS) for pain, fatigue, and concentration pre- and post-HUT. As pain is a core symptom in fibromyalgia (FM), we subgrouped ME/CFS patients by the presence or absence of FM. Methods and Results: In eligible ME/CFS patients who underwent HUT, NRS of pain, fatigue, and concentration were obtained pre-HUT, immediately after HUT, at 24 and 48 h, and at 7 days posttest. We studied 174 ME/CFS patients with FM, 104 without FM, and 30 healthy controls (HC). Values for all symptoms were unchanged for HC pre- and post-HUT. Compared with pre-HUT, the three NRS post-HUT were significantly elevated in both ME/CFS patient groups even after 7 days. NRS pain was significantly higher at all time points measured in the ME/CFS patients with FM compared with those without FM. In ME/CFS patients, the maximum fatigue and concentration scores occurred directly post-HUT, whereas pain perception reached the maximum 24 h post-HUT. Conclusion: NRS scores of pain, fatigue, and concentration were significantly increased even at 7 days post-HUT compared with pre-HUT in ME/CFS patients with and without FM, suggesting that orthostatic stress is an important determinant of PEM.
Article
Full-text available
A metabolic hallmark of many cancers is the increase in glucose consumption coupled to excessive lactate production. Mindful that L-lactate originates only from pyruvate, the question arises as to how can this be sustained in those tissues where pyruvate kinase activity is reduced due to dimerization of PKM2 isoform or inhibited by oxidative/nitrosative stress, posttranslational modifications or mutations, all widely reported findings in the very same cells. Hereby 17 pathways connecting glucose to lactate bypassing pyruvate kinase are reviewed, some of which transit through the mitochondrial matrix. An additional 69 converging pathways leading to pyruvate and lactate, but not commencing from glucose, are also examined. The minor production of pyruvate and lactate by glutaminolysis is scrutinized separately. The present review aims to highlight the ways through which L-lactate can still be produced from pyruvate using carbon atoms originating from glucose or other substrates in cells with kinetically impaired pyruvate kinase and underscore the importance of mitochondria in cancer metabolism irrespective of oxidative phosphorylation.
Article
Full-text available
Recent studies of the ketogenic diet, an extremely high-fat diet with extremely low carbohydrates, suggest that it changes the energy metabolism properties of skeletal muscle. However, ketogenic diet effects on muscle metabolic characteristics are diverse and sometimes countervailing. Furthermore, ketogenic diet effects on skeletal muscle performance are unknown. After male Wistar rats (8 weeks of age) were assigned randomly to a control group (CON) and a ketogenic diet group (KD), they were fed for 4 weeks respectively with a control diet (10% fat, 10% protein, 80% carbohydrate) and a ketogenic diet (90% fat, 10% protein, 0% carbohydrate). After the 4-week feeding period, the extensor digitorum longus (EDL) muscle was evaluated ex vivo for twitch force, tetanic force, and fatigue. We also analyzed the myosin heavy chain composition, protein expression of metabolic enzymes and regulatory factors, and citrate synthase activity. No significant difference was found between CON and KD in twitch or tetanic forces or muscle fatigue. However, the KD citrate synthase activity and the protein expression of Sema3A, citrate synthase, succinate dehydrogenase, cytochrome c oxidase subunit 4, and 3-hydroxyacyl-CoA dehydrogenase were significantly higher than those of CON. Moreover, a myosin heavy chain shift occurred from type IIb to IIx in KD. These results demonstrated that the 4-week ketogenic diet improves skeletal muscle aerobic capacity without obstructing muscle contractile function in sedentary male rats and suggest involvement of Sema3A in the myosin heavy chain shift of EDL muscle.
Article
Full-text available
Abstract Background Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a serious and complex physical illness that affects all body systems with a multiplicity of symptoms, but key hallmarks of the disease are pervasive fatigue and ‘post-exertional malaise’, exacerbation after physical and/or mental activity of the intrinsic fatigue and other symptoms that can be highly debilitating and last from days to months. Although the disease can vary widely between individuals, common symptoms also include pain, cognitive deficits, sleep dysfunction, as well as immune, neurological and autonomic symptoms. Typically, it is a very isolating illness socially, carrying a stigma because of the lack of understanding of the cause and pathophysiology. Methods To gain insight into the pathophysiology of ME/CFS, we examined the proteomes of peripheral blood mononuclear cells (PBMCs) by SWATH-MS analysis in a small well-characterised group of patients and matched controls. A principal component analysis (PCA) was used to stratify groups based on protein abundance patterns, which clearly segregated the majority of the ME/CFS patients (9/11) from the controls. This majority subgroup of ME/CFS patients was then further compared to the control group. Results A total of 60 proteins in the ME/CFS patients were differentially expressed (P 0.2 and
Article
Selective destruction of senescent cells Senescent cells are associated with a variety of age-related medical conditions and thus have been proposed as potential targets for therapy, but we do not yet have a full understanding of the underlying mechanisms. Johmura et al. used RNA interference to screen for enzymes essential to the survival of senescent cells (see the Perspective by Pan and Locasale). The authors identified a key role for glutamine metabolism, particularly the enzyme glutaminase 1, and demonstrated that inhibition of this pathway induced the death of senescent cells. Glutaminase targeting also ameliorated aging-related organ dysfunction and obesity-related disorders in mouse models, suggesting the potential therapeutic value of this approach. Science , this issue p. 265 ; see also p. 234
Article
2-Oxoglutarate-dependent dioxygenases (2OGDDs) are a superfamily of enzymes that play diverse roles in many biological processes, including regulation of hypoxia-inducible factor-mediated adaptation to hypoxia, extracellular matrix formation, epigenetic regulation of gene transcription and the reprogramming of cellular metabolism. 2OGDDs all require oxygen, reduced iron and 2-oxoglutarate (also known as α-ketoglutarate) to function, although their affinities for each of these co-substrates, and hence their sensitivity to depletion of specific co-substrates, varies widely. Numerous 2OGDDs are recurrently dysregulated in cancer. Moreover, cancer-specific metabolic changes, such as those that occur subsequent to mutations in the genes encoding succinate dehydrogenase, fumarate hydratase or isocitrate dehydrogenase, can dysregulate specific 2OGDDs. This latter observation suggests that the role of 2OGDDs in cancer extends beyond cancers that harbour mutations in the genes encoding members of the 2OGDD superfamily. Herein, we review the regulation of 2OGDDs in normal cells and how that regulation is corrupted in cancer.