ArticlePDF Available

Morphological evidence for the existence of nitric oxide and carbon monoxide pathways in the rat islets of Langerhans: An immunocytochemical and confocal microscopical study

Authors:

Abstract and Figures

To map the cellular location of inducible and constitutive nitric oxide synthase and haem oxygenase in rat islets to clarify the morphological background to putative nitric oxide and carbon monoxide pathways. Immunocytochemistry and confocal microscopy. After treatment with endotoxin, immunoreactivity for inducible nitric oxide synthase was expressed in a large number of islet cells, most of which were insulin-immunoreactive beta cells and in single glucagon-immunoreactive and pancreatic polypeptide-immunoreactive cells. Somatostatin-immunoreactive cells lacked immunoreactivity for inducible nitric oxide synthase. In untreated rats, immunoreactivity for constitutive nitric oxide synthase occurred in the majority of insulin-immunoreactive and glucagon-immunoreactive cells, in most pancreatic polypeptide-immunoreactive and somatostatin-immunoreactive cells and in islet nerves. Similarly, immunoreactivity for constitutive haem oxygenase was detected in all four types of islet cells. Endotoxin treatment did not change the pattern of immunoreactivity for constitutive and inducible haem oxygenase. After treatment with alloxan, insulin-immunoreactivity was observed only in single islet cells, being almost devoid of immunoreactivity for constitutive nitric oxide synthase and haem oxygenase. In vivo endotoxin-induced expression of inducible nitric oxide synthase in insulin-producing and in scattered glucagon-producing and pancreatic polypeptide-producing cells strengthens previous suggestions of a pathophysiological role for inducible nitric oxide synthase in the development of insulin-dependent diabetes mellitus. The presence of constitutive nitric oxide synthase and haem oxygenase in all four types of islet cells, together with recent functional data of ours support roles for nitric oxide and carbon monoxide as intracellular, paracrine or neurocrine modulators of islet hormone secretion.
Content may be subject to copyright.
Diabetologia (1999) 42: 978±986
Morphological evidence for the existence of nitric oxide and
carbon monoxide pathways in the rat islets of Langerhans:
An immunocytochemical and confocal microscopical study
P. A l m 1, P. Ekström2, R. Henningsson3, I. Lundquist3
1Department of Pathology, University of Lund, Sweden
2Department of Zoology, University of Lund, Sweden
3Department of Pharmacology, University of Lund, Sweden
ÓSpringer-Verlag 1999
Abstract
Aims/hypothesis. To map the cellular location of in-
ducible and constitutive nitric oxide synthase and
haem oxygenase in rat islets to clarify the morpholog-
ical background to putative nitric oxide and carbon
monoxide pathways.
Methods. Immunocytochemistry and confocal mi-
croscopy.
Results. After treatment with endotoxin, immunore-
activity for inducible nitric oxide synthase was ex-
pressed in a large number of islet cells, most of which
were insulin-immunoreactive beta cells and in single
glucagon-immunoreactive and pancreatic polypep-
tide-immunoreactive cells. Somatostatin-immunore-
active cells lacked immunoreactivity for inducible ni-
tric oxide synthase. In untreated rats, immunoreactiv-
ity for constitutive nitric oxide synthase occurred in
the majority of insulin-immunoreactive and gluca-
gon-immunoreactive cells, in most pancreatic
polypeptide-immunoreactive and somatostatin-im-
munoreactive cells and in islet nerves. Similarly, im-
munoreactivity for constitutive haem oxygenase was
detected in all four types of islet cells. Endotoxin
treatment did not change the pattern of immunoreac-
tivity for constitutive and inducible haem oxygenase.
After treatment with alloxan, insulin-immunoreactiv-
ity was observed only in single islet cells, being almost
devoid of immunoreactivity for constitutive nitric ox-
ide synthase and haem oxygenase.
Conclusion/interpretation. In vivo endotoxin-induced
expression of inducible nitric oxide synthase in insu-
lin-producing and in scattered glucagon-producing
and pancreatic polypeptide-producing cells strength-
ens previous suggestions of a pathophysiological role
for inducible nitric oxide synthase in the develop-
ment of insulin-dependent diabetes mellitus. The
presence of constitutive nitric oxide synthase and
haem oxygenase in all four types of islet cells, togeth-
er with recent functional data of ours support roles
for nitric oxide and carbon monoxide as intracellular,
paracrine or neurocrine modulators of islet hormone
secretion. [Diabetologia (1999) 42: 978±986]
Keywords Pancreatic islets, nitric oxide synthase,
haem oxygenase, imunocytochemistry, confocal mi-
croscopy.
Received: 19 November 1998 and in revised form: 22 March
1999
Corresponding author: P. Alm, MD, PhD, Department of Pa-
thology, University Hospital, S-22185 Lund, Sweden
Abbreviations: CO, Carbon monoxide; HO, haem oxygenase;
HO-1, inducible haem oxygenase; HO-2, constitutive haem ox-
ygenase; NO, nitric oxide; NOS, nitric oxide synthase; GLUC,
glucagon; IG, immunoglobulins; INS, insulin; IR, immunore-
active; LPS, lipopolysaccharide (endotoxin); iNOS, inducible
nitric oxide synthase; cNOS, constitutive nitric oxide synthase;
eNOS, endothelial nitric oxide synthase; nNOS, neuronal ni-
tric oxide synthase; PP, pancreatic polypeptide; SOM, soma-
tostatin; ZnPP, zinc protoporphyrin; FITC, fluorescein isothio-
cyanate conjugated.
Nitric oxide (NO) is a free radical gas that conveys
biological information in a way greatly differing
from that of the classical transmitters. In the nervous
system NO does not act on conventional receptors
but through effects on various regulatory processes,
intracellularly or in the membrane [1]. The formation
of NO is catalysed by the enzyme nitric oxide syn-
thase (NOS), in a reaction in which l-arginine and
oxygen are converted to NO and citrulline. There
are two major types of NOS enzymes; one inducible
isoform iNOS, originally described in macrophages
and also shown to be expressed in a variety of mam-
malian tissues among which are the islets of Langer-
hans [2, 3], and constitutive isoforms (cNOS) present
in neurons (nNOS) and endothelial cells (eNOS) [1].
We and others [4±8] have shown previously that the
pancreatic islets contain a constitutive NOS as deter-
mined by histochemical, immunocytochemical and
biochemical methods. Islet iNOS has been implicated
as an important factor in the pathogenesis of Type I
(insulin-dependent) diabetes mellitus [2, 3], whereas
islet cNOS has been suggested to be involved in the
physiological regulation of insulin and glucagon se-
cretion [4±15]. Thus, both cNOS and iNOS seem to
be of great physiological and pathophysiological im-
portance in the pancreatic islets. Constitutive nitric
oxide synthase was localised by the use of an antiser-
um to the neuronal isoform of cNOS.
There are diverging results as to the cellular loca-
tion of the NOS isoforms within the islets, i.e. whe-
ther only the insulin producing beta cells or, in addi-
tion, other endocrine cell types such as glucagon-pro-
ducing, somatostatin-producing, and pancreatic
polypeptide (PP)-producing cells also contain NOS
activity [4±8, 16±18]. Further, in this context, atten-
tion has also been drawn to another gaseous mole-
cule, carbon monoxide (CO), since a number of re-
cent studies have shown that CO may serve as a neu-
ronal messenger molecule similar to NO [19±21].
Carbon monoxide is produced by the action of haem
oxygenase (HO), at which haem from haemoglobin
is degraded to CO and biliverdin [19±22]. The latter
compound can then be converted to bilirubin, which
is an important antioxidant, the reaction being cataly-
sed by the enzyme biliverdin reductase. Similar to
NOS, HO consists of at least two isoenzymes, an in-
ducible (HO-1), and a constitutively expressed iso-
form (HO-2) [19±21]. Expression of HO-1 is induced
by various stress factors, e.g. fever, starvation, oxida-
tive injury. A cytokine-induced expression of a pro-
tein (presumably HO-1) has previously been ob-
served in islet tissue, and suggested to be a protective
mechanism against oxidative stress [2, 3, 23±25]. Con-
stitutive haem oxygenase is highly expressed in ner-
vous tissue, in which CO may have a transmitter-like
function [19, 20] and recent findings of ours suggest
a role for CO in the regulation of the release of islet
hormones in rats [26].
To further clarify the morphological background
to the putative NO and CO functional pathways in
the islets of Langerhans, the aim of this study was to
map the cellular location of iNOS, cNOS, HO-1 and
HO-2 in islets of normal and alloxan diabetic rats by
means of combined immunocytochemical and confo-
cal microscopical methods.
Materials and methods
Tissue handling. Female Sprague-Dawley rats (300±400 g body
weight, aged about 3±4 months) were purchased from B&K
Universal, Stockholm, Sweden. The animals had free access
to water and standard pellets and were used in different exper-
imental groups consisting of four to six animals. One group
consisted of rats that received no treatment. One group of
rats was given LPS (lipopolysaccharide endotoxin from salmo-
nella typhimurium, Sigma, St Louis, Mo., USA; 10 mg/kg i. p.,
dissolved in saline) and used after 6 h, at which time there is a
high expression of iNOS [34]. One group was treated with al-
loxan (Sigma; 60 mg/kg i.v., dissolved in saline with the addi-
tion of a drop of 0.1 N acetic acid to acidify the solution) and
killed after 5 days. Plasma glucose was determined by a glu-
cose oxidase method [18, 27] to ensure that the animals had be-
come diabetic. The concentrations of plasma glucose in freely
fed diabetic animals were 15.1±38.7 mmol/l (total range) com-
pared with 8.9±11.7 mmol/l in normal rats. The rats were an-
aesthesized with ketamine (100 mg/kg intramuscular; Ketalar,
Parke Davis, Barcelona, Spain) and xylazin (15 mg/kg intra-
muscular; Rompun, Bayer, Leverkusen, Germany) and perfus-
ed transcardially through the ascending aorta, first with 100 ml
of ice-cold calcium-free Krebs buffer (containing 0.5 g/l sodi-
um nitrite and 10.000 iU/l of heparin), and then with 300 ml of
an ice-cold, freshly prepared solution of 4% formaldehyde in
phosphate buffered saline (PBS, 0.1 mol/l, pH 7.4). The pan-
creatic glands were then rapidly dissected out and divided
into pieces, which were fixed in the same fixative for 4 hours.
After this they were rinsed in ice-cold 15 % sucrose in PBS
(three rinses during 48 h). The tissue specimens were frozen
in isopentane at ±40°C and then stored at ±70°C. Principles of
laboratory animal care (NIH publication No 85±23 1985)
were followed and the experimental design was also approved
by the animal ethics committee of the University of Lund,
Lund, Sweden.
Immunocytochemistry. Cryostat section were cut at a thickness
of 8 mm and thaw-mounted onto chrom alum-coated glass
slides and air dried for 30 min to 1 h. To show iNOS, nNOS,
HO-1 and HO-2, sections were pre-incubated in PBS with
0.2% Triton X-100 for about 2 h, and then incubated for
2 days in the presence of rabbit antisera to iNOS, nNOS, HO-
1 or HO-2. The antiserum to iNOS (1:500) was generated in
rabbits against a 25 amino acid peptide of a cloned inducible
NOS from a murine macrophage cell line [28, 29]. The antisera
to nNOS were generated in rabbits against a 15 amino acid se-
quence (nNOS-15, 1:1280) [30], or a 21 amino acid sequence
(nNOS-21, peptide 58; 1:2000) [31] from the C-terminal part
of a cloned rat cerebellar NOS [32]. The antiserum to HO-1
(1:500; code OSA 100, StressGen Biotechnol, Victoria, Cana-
da) was generated in rabbits against rat liver HO-1. The rabbit
HO-2 antiserum (1:1000, code OSA-200; StressGen) was gen-
erated in rabbits against rat testes HO-2. After rinsing in PBS
(three rinses during 10 min), the sections were incubated for
90 min with fluorescein isothiocyanate conjugated (FITC)
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase 979
swine anti-rabbit immunoglobulins (IG) (1:80; Dakopatts,
Stockholm, Sweden) or Texas red-conjugated affinity purified
F(ab¢)2fragments of donkey anti-rabbit IG (1:80; code
711±076±132; Jackson Immuno Research, West Grove, Pa.,
USA). After rinsing, the sections were mounted in PBS/glycer-
ol with p-phenylenediamine to prevent fluorescence fading
[33].
To show two antigens simultaneously [34], sections were in-
cubated overnight with iNOS, nNOS, or HO-2 antisera (see
above), rinsed and then incubated overnight with antisera gen-
erated in guinea-pigs to insulin (1:16 000), glucagon (1:4000)
and pancreatic polypeptide (1:500) (the latter antisera pur-
chased from Linco Res, St Louis, Mo., USA), and a mouse
monoclonal antiserum to somatostatin (1:5 of a prediluted an-
tiserum; cat no 8330±0496, Biogenesis, Poole, England). After
rinsing, the sections were incubated for 90 min with FITC
goat anti-guinea-pig IG or goat anti-mouse IG (1:80; Sigma,
St Louis, Mo., USA), rinsed and then incubated with Texas
red conjugated affinity purified F(ab¢)2fragments of donkey
anti-rabbit IG (1:80; see above). The sections were rinsed and
mounted as described above. An Olympus 3 ´50 fluorescence
microscope (LRI Instrument AB, Lund, Sweden) equipped
with epi-illumination and appropriate filter settings for Texas
Red-immunofluorescence and FITC-immunofluorescence
was used for the examinations of the sections [35].
The primary and secondary antisera were diluted in PBS. In
control experiments no immunoreactivity could be detected in
sections incubated in the absence of the primary antisera or
with nNOS-15, HO-2, glucagon, somatostatin or pancreatic
polypeptide antisera absorbed with excess of the correspond-
ing immunizing antigen (100 mg/ml). No absorption controls
could be done with the iNOS or nNOS-21 antisera as antigenic
substances were not available. The characteristics of the iNOS
and the nNOS antisera have been presented previously
[29±31]. In control experiments iNOS-immunoreactivity was
only observed in LPS-induced tissues (macrophages in lung
and liver), in which no nNOS-immunoreactivity could be
seen. As cross reactions to antigens sharing similar amino acid
sequences cannot be completely excluded the structures shown
are referred to as iNOS-, nNOS-, HO-1-, HO-2-, insulin-(INS-),
glucagon-(GLUC-), pancreatic polypeptide-(PP-), or soma-
tostatin-(SOM-) immunoreactive (IR).
Confocal microscopy. To evaluate whether two immunoreac-
tivities were colocalized within the same cellular structures,
sections were analysed in a confocal laser scanning microscope
(Multiprobe 2001 TM CLSM; Molecular Dynamics) equipped
with an Ar/Kr laser and an inverted Nikon Diaphot TMD mi-
croscope as described elsewhere [35].
Results
No iNOS-immunoreactivity could be detected in is-
lets of untreated animals. After LPS treatment,
iNOS-immunoreactivity was expressed in a large
number of islet cells, which were diffusely spread
over the islets (Fig.1A, D, G, J). Double immuno-
staining showed that these cells were also INS-IR,
which was further verified by confocal microscopy
(Fig.1C).
Moreover, single iNOS-IR cells also displayed
GLUC-immunoreactivity and PP-immunoreactivity
(Fig.1F, I) but in most of the GLUC-IR and PP-IR
cells iNOS-immunoreactivity was lacking. No iNOS-
IR cells expressed SOM-immunoreactivity (Fig.1L).
In comparison, treatment with LPS did not seem to
change the nNOS-immunolabelling pattern with
both the nNOS antisera used.
Constitutive NOS expressed as nNOS-immunore-
activity could be detected in the cytoplasm of most is-
let cells of untreated animals (Fig.2A, D, G). The
number and distribution patterns of nNOS-IR cells
were similar with the two NOS antisera used, al-
though the intensity of the NOS-immunofluores-
cence was weaker with the nNOS-21 than with the
nNOS-15 antiserum. Vessels of capillary size, be-
tween the trabecula of islet cells (Fig.2G) and be-
tween exocrine acini, were accompanied by varicose
NOS-IR nerve terminals, which were also found
around arteries of various sizes. No nNOS-immu-
noreactivity could be detected in endothelial cells of
the islet microvasculature.
Double immunolabelling showed that nNOS-im-
munoreactivity was in most INS-IR cells (Fig.2A±C).
In the periphery of the islets there were, however,
several nNOS-IR cells which lacked INS-immunore-
activity. Most GLUC-IR cells, which were located
along the periphery of the islets, were also nNOS-IR
(Fig.2D±F), although single GLUC-IR cells were dis-
covered which lacked nNOS-immunoreactivity. Pan-
creatic polypeptide-IR and some SOM-IR cells,
which were also located in the periphery of the islets,
also expressed nNOS-immunoreactivity (data not
shown), which in the SOM-IR cells were as far as to
the ends of their long and gracile dendritic processes.
In the cytoplasm of almost all islet cells HO-2 im-
munoreactivity could be detected (Fig.3 A, D, G,
L). Double immunolabelling in combination with
confocal microscopy revealed that most INS-IR cells
also showed HO-2 immunoreactivity, although there
were HO-2 IR cell in the periphery of the islets that
lacked INS-immunoreactivity (Fig.3A±C). Further,
along the periphery of the islets there was a broad
ring of GLUC-IR, which also were HO-2 IR (Fig. 3
D±F) and dispersed PP-cells and SOM-IR cells with
extended processes. These also expressed HO-2 im-
munoreactivity (Fig.3G±K, L±N). No HO-1 immu-
noreactivity could be detected in any type of islet
cells.
After treatment with alloxan almost no specific
nNOS-immunoreactivity and HO-2 immunoreactivi-
ty was discovered in most of the damaged beta cells
and INS-immunoreactivity was only seen in single is-
let cells, which were vacuolized and enlarged. In com-
parison, nNOS-immunoreactivity and HO-2 immu-
noreactivity was well preserved only in non beta cells
(GLUC-IR cells and others) (data not shown).
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase980
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase 981
Fig. 1 A±L. Confocal microscopy of rat islets of Langerhans af-
ter treatment with LPS. Left panel: red fluorescence in A,D,G
and Jindicating expression of iNOS-immunoreactivity (Texas
red immunofluorescence). Middle panel: green fluorescence
(FITC-immunofluorescence) shows immunoreactivities for in-
sulin (B), glucagon (E), pancreatic polypeptide (H) and soma-
tostatin (K). Right panel: overlay picture of A+B (=C),
D+E(=F), G+H(=I) and J+K(=L). Cells showing yel-
lowish fluorescence (arrowheads) indicate colocalization of
iNOS/insulin (C), iNOS/glucagon (F), and iNOS/pancreatic
polypeptide (I). Bars with numerals indicate lengths (mm)
Discussion
A characteristic of Type I diabetes is a local inflam-
matory reaction in the pancreatic islets that are un-
dergoing autoimmune destruction [2, 3]. Nitric oxide
has been proposed as a possible mediator in the dam-
age process to the insulin producing beta cells and
there is ample in vitro evidence that IL-1 and other
cytokines are able to induce iNOS expression in islet
tissue [2, 3]. The pancreatic islet consists, however,
of a heterogeneous cell population, making it difficult
to localize the cellular source of iNOS expression and
NO production. It was recently shown that rat islets
exposed to cytokine in vitro expressed iNOS in their
insulin cells whereas the glucagon cells seemed unaf-
fected [7]. No data on the possible existence of iNOS
in somatostatin cells or PP-cells have so far appeared
in the literature.
Lipopolysaccharide (endotoxin) is known to stim-
ulate cytokine production [2, 3]. It is important, how-
ever, to note that apart from cytokines, other factors
also have been suggested to serve as direct or indirect
mediators of effects of LPS. Thus, it is known that
LPS may cause the synthesis of reactive oxygen spe-
cies such as superoxide and hydrogen peroxide, and
that NO can combine with superoxide to form the po-
tent oxidizing agent peroxynitrite [2, 3]. Hence, our
immunocytochemical data cannot be extrapolated to
answer questions on the intimate mechanisms of im-
mune destruction of the islet beta cells. Our results
show that after treatment with LPS in vivo iNOS is
expressed in most INS-IR cells and also, although to
a much lesser extent, in scattered GLUC-IR and PP-
IR cells. No iNOS was detected in cells immunostain-
ed for somatostatin. Our in vivo data of iNOS expres-
sion in INS-IR cells, agree with findings of previous
studies of islets exposed to cytokine in vitro [2, 3, 7].
The observation that iNOS expression can be elicited
in GLUC-IR cells and PP-IR cells has not been de-
scribed previously. This may be explained by differ-
ences between the in vitro and the in vivo situation
and the possible induction by LPS of unknown
iNOS stimulatory factors. On the other hand, since
the fraction of iNOS positive cells among the
GLUC-IR cell and PP-IR cells is much smaller than
among the INS-IR cells, it is not inconceivable that
refined techniques such as confocal microscopy are
required to observe these iNOS positive GLUC-IR
cell and PP-IR cells. It is difficult to explain why
SOM-IR cells are not influenced by LPS. It has
been shown [36, 37] that cells producing somatostatin
in contrast to those producing insulin, glucagon and
PP are not members of the group of endocrine cells
belonging to the amine precursor uptake and decar-
boxylation series. Whether the ability of endocrine
cells to store amine is coupled to that of expressing
iNOS remains to be explained. If NO finally turns
out, however, to be of pathophysiological importance
in the development of Type I diabetes, it is notable
that not only insulin cells but also GLUC-IR cells
and PP-IR cells are able to express iNOS. Whether
the number of these non-beta cells observed express-
ing iNOS is, however, enough to actually contribute
to damaging the beta cells remains to be explained.
It is also tempting to speculate that NO derived by
the action of iNOS in the glucagon cells is at least
partly responsible for the increased glucagon secre-
tion in the diabetic condition. Indirectly, such a
mechanism might also explain why there is an in-
creased somatostatin response to glucagon in diabe-
tes.
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase982
Fig. 2 A±G. Confocal microscopy of islets of Langerhans of
untreated rats. Left panel: red fluorescence in Aand D(Texas
Red immunoflurescence) indicates nNOS-immunoreactivity.
Middle panel: green fluorescence (FITC-immunofluores-
cence) shows immunoreactivities for insulin (B) and glucagon
(E). Right panel: overlay picture of A+B(=C), and D+E
(=F). Yellowish fluorescent cells indicate colocalization of
nNOS/insulin (C) and nNOS/glucagon (F). Bars with numerals
indicate lengths (mm). G. Rat islet of Langerhans with nNOS-
IR varicose nerve terminals (arrowheads) running along ves-
sels of capillary size between endocrine cells. FITC-immuno-
fluorescence. Bar = 100 mm
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase 983
Fig. 3 A±L. Confocal microscopy of islets of Langerhans of un-
treated rats. Left panel: red fluorescence in A,D,G and Jdis-
plays expression of HO-2 immunoreactivity (Texas red immu-
nofluorescence). Middle panel: green fluorescence (FITC-im-
munofluorescence) shows immunoreactivities for insulin (B),
glucagon (E), pancreatic polypeptide (H) and somatostatin
(K). Right panel: overlay picture of A+B (=C), D+E
(=F), G+H(=I) and J+K (=L). Cells showing yellowish
fluorescence indicate colocalization of HO-2/insulin (C), HO-
2/glucagon (F), HO-2/pancreatic polypeptide (I), and HO-2/
somatostatin (L). Bars with numerals indicate lengths (mm)
Our data show that all four endocrine cell types,
i.e. those containing insulin, glucagon, somatostatin
and PP showed immunoreactivity for two different
antisera of the neuronal isoform of constitutive
NOS. As previously discussed it has been known for
several years [2, 3] that rat pancreatic islets are able
to express the inducible NOS isoform after treatment
in vitro with different cytokines. The possibility of a
constitutive NOS isoform being present has, howev-
er, been a matter of debate. We and others [4, 5, 7, 8,
16, 38] have previously observed cNOS activity in rat
and mouse islet cells by both histochemical (NAD-
PH-diaphorase activity) and immunocytochemical
methods. In a recent study, nNOS immunoreactivity
was shown in rat islets with a weak intensity [38] that
was apparently lower than the findings in this study.
This is possibly due to differences in methodology
and nNOS antisera used but also to differences in
brightness of the fluorophores of the secondary anti-
bodies [39]. In comparison, other studies carried out
with other NOS antisera as well as with the NAD-
PH-diaphorase method were either unable to show
cNOS activity in rat islet cells or reported that only
cells containing somatostatin or islet nerves or both
were positive for NOS [40, 41, 42]. The reasons for
these discrepancies are not known. The existence of
different local isoforms, differences between antisera
in recognizing epitopes or differences in methodolo-
gy are possible explanations. Further, the recent ob-
servation that a constitutive eNOS is localized to glu-
cagon and somatostatin (but not to insulin cells) in rat
islets suggests the possibility that both eNOS and
nNOS are localized in the same cell type [5, 18]. In
the present study using two antisera of different
sources directed against neuronal NOS, we observed
that nNOS-immunoreactivity was located in all four
types of endocrine cells. Moreover, confocal micros-
copy disclosed coinciding profiles between cells ex-
pressing nNOS-immunoreactivity and immunoreac-
tivity for insulin, glucagon, pancreatic polypeptide
and somatostatin, respectively. Taken together with
the results from previous studies [4±8, 16] this data
strongly suggest that a constitutive nNOS resides in
all these four cell types and in nervous structures as
well. Moreover, results of most functional studies on
the influence of NOS-inhibitors and NO donors on
insulin and glucagon secretion favour NO as an im-
portant modulator of the secretory processes of these
hormones [4±6, 8±11, 13±15, 43]. Although some ear-
ly studies [8, 10] and a recent one (using islets from
newborn rats) [18] suggested that NO had a positive
effect on glucose-stimulated and l-arginine stimulat-
ed insulin release, we [4±6, 9, 14, 15, 44±46] and oth-
ers [13, 43, 47] have repeatedly shown that NO is
strongly inhibitory to insulin secretion induced by
these secretagogues. The inhibitory effect of NO on
insulin release induced by nutrients is probably due
to the formation of S-nitrosothiols [48], which impair
important regulatory thiol groups. Thiol groups have
long been shown to be essential for stimulus-secre-
tion coupling induced by glucose [49, 50]. In contrast,
NO is a positive modulator of glucagon secretion,
probably acting by stimulating the cyclic GMP system
[5, 6, 44±46].
It has previously been shown that cytotoxicity me-
diated by cytokine can induce expression of HO-1 in
cultured islets [23±25]. That we did not observe any
immunocytochemical evidence for HO-1 expression
in rats islets after injection with endotoxin does not
exclude that HO-1 could be detected by other more
sensitive methods but not by immunocytochemistry
which might be too insensitive in the present situa-
tion. We have very recently obtained evidence for
the presence of mechanisms mediated by HO-2 and
CO in the release of insulin and glucagon from rat is-
lets [26]. Thus, islet tissue was found to produce large
amounts of CO [26]. As this CO production was
strongly suppressed by the HO-inhibitor zinc proto-
porphyrin-IX (ZnPP-IX), and since both insulin and
glucagon secretion from intact islets could be sup-
pressed by ZnPP-IX and stimulated by the HO-sub-
strate haemin, we concluded that CO should be re-
cognized as a putative physiologic stimulator of insu-
lin and glucagon release [26]. The data in this study
strongly suggest that HO-2 resides in all four types
of endocrine cells in rat pancreatic islets and confocal
microscopy showed coinciding profiles between cells
expressing HO-2 immunoreactivity and immunoreac-
tivities for insulin and glucagon as well as for PP and
somatostatin. Hence, the CO-pathway could be of
functional significance as an intracellular modulator
system, not only for the release of insulin and gluca-
gon [26] but also for the secretion of PP and soma-
tostatin. Thus, from the most recent data and those
from this study it seems likely that NO as well as CO
formed within the islets of Langerhans are capable
of acting both within their cells of origin and also as
paracrine, neurocrine or even as endocrine media-
tors.
In conclusion, we have shown the existence of a
morphological substrate for a putative functional
role of iNOS, nNOS and HO-2 as important regulato-
ry enzymes in the physiology and pathophysiology of
hormone secretion from the islets of Langerhans.
Acknowledgements. The technical help of L. Thuresson and
the secretarial help of E. Björkbom is gratefully acknow-
ledged. This study was supported by the Swedish Medical Re-
search Council (12X-11205, 14X-4286), the foundations of
Crafoord, Magnus Bergvall, Albert Påhlsson, Thelma Zoga
and ke Wiberg, the Swedish Diabetes Association and the
Medical Faculty, University of Lund, Lund, Sweden. The gen-
erous supply of iNOS and nNOS-21 antisera by V. Riveros-
Moreno, Wellcome Research Laboratories, Beckenham,
England is gratefully appreciated.
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase984
References
1. Knowles RG, Moncada S (1994) Nitric oxide synthases in
mammals. Biochem J 298: 249±258
2. Eizirik DL, Flodström M, Karlsen AE, Welsh N (1996) The
harmony of the spheres: inducible nitric oxide synthase and
related genes in pancreatic beta cells. Diabetologia 39:
875±890
3. Mandrup-Poulsen T (1996) The role of interleukin-1 in the
pathogenesis of IDDM. Diabetologia 39: 1005±1029
4. Panagiotidis G, Alm P, Lundquist I (1992) Inhibition of is-
let nitric oxide synthase increases arginine-induced insulin
release. Eur J Pharmacol 229: 277±278
5. Panagiotidis G, kesson B, Alm P, Lundquist I (1994) The
nitric oxide system in the endocrine pancreas induces dif-
ferential effects on the secretion of insulin and glucagon.
Endocr 2: 787±792
6. Salehi A, Carlberg M, Henningson R, Lundquist I (1996)
Islet constitutive nitric oxide synthase: biochemical deter-
mination and regulatory function. Am J Physiol 270 (Cell
Physiol 39): C1634±C1641
7. Corbett JA, McDaniel ML (1995) Intra islet release of inter-
leukin 1 inhibits bcell function by inducing B cell expression
of inducible nitric oxide synthase. J Exp Med 181: 559±568
8. Schmidt HHHW, Warner TD, Ishii K, Scheng H, Murad F
(1992) Insulin secretion from pancreatic B cells caused by
L-arginine-derived nitrogen oxides. Science 255: 721±723
9. Panagiotidis G. kesson B, Rydell EL, Lundquist I (1995)
Influence of nitric oxide synthase inhibition, nitric oxide
and hydroperoxide on insulin release induced by various
secretagogues. Br J Pharmacol 114: 289±296
10. Laychock RG, Modica ME, Cavanaugh CT (1991) L-Argi-
nine stimulates cyclic guanosine 3 ¢,5 ¢-monophosphate for-
mation in rat islets of Langerhans and RINm5F insulinoma
cells: evidence for l-arginine: nitric oxide synthase. Endo-
crinology 129: 3043±3052
11. Jansson L, Sandler S (1991) The nitric oxide synthase in-
hibitor NG-nitro L-arginine stimulates pancreatic islet insu-
lin release in vitro, but not in the perfused pancreas. Endo-
crinology 128: 3081±3085
12. Jones PM, Persaud SJ, Bjaaland T, Pearson JD, Howell SL
(1992) Nitric oxide is not involved in the initiation of insu-
lin secretion from rat islets of Langerhans. Diabetologia
35: 1020±1027
13. Gross R, Roye M, Manteghetti M, Hillaire-Buys D, Ribes
G (1995) Alterations of insulin response to different bcell
secretagogues and pancreatic vascular resistance induced
by Nw-nitro-L-arginine methyl ester. Br J Pharmacol 116:
1965±1972
14. kesson B, Lundquist I (1996) Modulation of the islet ni-
tric oxide system and sulphonylurea-induced insulin secre-
tion. Diabetes Res 31: 91±99
15. kesson B, MosØn H, Panagiotidis G, Lundquist I (1996)
Interaction of the islet nitric oxide system with L-arginine-
induced secretion of insulin and glucagon in mice. Br J
Pharmacol 119: 758±764
16. Bouwens L, Klöppel G (1994) Cytochemical localization of
NADPH-diaphorase in the four types of pancreatic islet
cell. Histochemistry 101: 209±214
17. Burrell MA, Montuenga LM, Carcía M, Villaro AC (1996)
Detection of nitric oxide synthase (NOS) in somatostatin-
producing cells of human and murine stomach and pancre-
as. J Histochem Cytochem 44: 339±346
18. Spinas GA, Laffranchi R, Francoys I, David I, Richter C,
Reinecke M (1998) The early phase of glucose-stimulated
insulin secretion requires nitric oxide. Diabetologia 41:
292±299
19. Marks GS, Brien JF, Nakatsu K, McLaughlin BE (1991)
Does carbon monoxide have a physiological function?
Trends Pharmacol Sci 11: 185±188
20. Verma A, Hirsch DJ, Glatt CE, Ronnett GV, Snyder SH
(1993) Carbon monoxide: a putative neural messenger. Sci-
ence 259: 381±384
21. Maines MD (1988) Heme oxygenase: function, multiplicity,
regulatory mechanisms, and clinical applications. FASEB J
2: 2557: 2568
22. Vreman HJ, Stevenson DK (1988) Heme oxygenase activi-
ty measured by carbon monoxide production. Anal Bio-
chem 168: 31±38
23. Helqvist S, Polla BS, Johannesen J, Nerup J (1991) Heat
shock protein induction in rat pancreatic islets by recombi-
nant human interleukin 1b. Diabetologia 34: 150±156
24. Strandell E, Buschard K, Saldeen J, Welsh N (1995) Inter-
leukin 1binduces expression of HSP70, heme oxygenase
and Mn-SOD in FACS-purified rat islet b-cells, but not in
a-cells. Immunol Lett 48: 145±148
25. Welsh N, Sandler S (1994) Protective action by hemin
against interleukin 1binduced inhibition of rat pancreatic
islet function. Mol Cell Endocrinol 103: 109±114
26. Henningsson R, Alm P, Lundquist I (1997) Occurrence and
putative hormone regulatory function of a constitutive
heme oxygenase in rat pancreatic islets. Am J Physiol 273:
C703±C709
27. Bruss ML, Black AL (1978) Enzymatic microdetermintion
of glycogen. Anal Biochem 84: 309±312
28. Lyons CR, Orloff GJ (1992) Molecular cloning and func-
tional expression of an inducible nitric oxide synthase
from a murine macrophage cell line. J Biol Chem 267:
6370±6374
29. Hamid Q, Springall DR, Riveros-Moreno V et al. (1993)
Induction of nitric oxide synthase in asthma. Lancet 342:
1510±1513
30. Alm P, Larsson B, Ekblad E, Sundler F, Andersson K-E
(1993) Immunohistochemical localization of peripheral ni-
tric oxide synthase-containing nerves using antibodies
raised against synthesized C- and N-terminal fragments of
a cloned enzyme from rat brain. Acta Physiol Scand 148:
421±429
31. Springall DR, Riveros-Moreno V, Buttery L et al. (1992)
Immunological detection of nitric oxide synthase(s) in hu-
man tissues using heterologous antibodies suggesting dif-
ferent isoforms. Histochemistry 98: 259±266
32. Bredt DS, Hwang PM, Glatt CE, Lowenstein C, Reed RR,
Snyder SH (1991) Cloned and expressed nitric oxide syn-
thase structurally resembles cytochrome P-450 reductase.
Nature 351: 714±718
33. Johnson GD, Araujo GM (1981) A simple method of re-
ducing the fading of immunofluorescence during microsco-
py. J Immunol Methods 43: 349±350
34. Wessendorf MW, Elde RP (1985) Characterization of an
immunofluorescence technique for the demonstration of
coexisting neurotransmitters within nerve fibers and termi-
nals. J Histochem Cytochem 33: 984±994
35. Ny L, Alm P, Larsson B, Ekström P, Andersson KE (1995)
The nitric oxide pathway in cat esophagus: localization of
nitric oxide synthase and functional aspects. Am J Physiol
268: G57±G70
36. Ekholm R, Ericson LE, Lundquist I (1971) Monoamines in
the pancreatic islets of the mouse. Subcellular localization
of 5-hydroxytryptamine by electron microscopic autorad-
iography. Diabetologia 7: 339±348
37. Lundquist I, AhrØn B, Håkanson R, Sundler F (1979) The
role of intracellular amines in the regulation of islet cell
function. In: Waldhäusl WK (ed) Proc 10th Congr Intern
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase 985
Diabetes Fed, Sept 9±14, Vienna, Austria, Exerpta Medica,
Amsterdam-Oxford-Princeton, pp 57±63
38. Umehara K, Kataoka K, Ogura T et al. (1997) Compara-
tive distribution of nitric oxide synthase (NOS) in pancreas
of the dog and rat: immunocytochemistry of neuronal type
NOS and histochemistry of NADPH-diaphorase. Brain
Res Bull 42: 469±474
39. Wessendorf MW, Brelje TC (1992) Which fluorophore is
brightest? A comparison of the staining obtained using flu-
orescein, tetramethylrhodamine, lissamine rhodamine,
Texas Red and cyanine 3.18. Histochemistry 98: 81±85
40. Vincent SR (1992) Nitric oxide and arginine-evoked insulin
secretion. Science 258: 1376
41. Shimosegawa T, Abe T, Satoh A et al. (1993) NADPH-dia-
phorase activity in neurons of the mammalian pancreas: co-
expression with vasoactive intestinal polypeptide. Gastro-
enterology 105: 999±1008
42. Wörl J, Wiesand M, Mayer B, Greskötter K-R, Neuhuber
WL (1994) Neuronal and endothelial nitric oxide synthase
immunoreactivity and NADPH-diaphorase staining in rat
and human pancreas: influence of fixation. Histochemistry
102: 353±364
43. Cunningham JM, Mabely JG, Delaney CA, Green IC
(1994) The effect of nitric oxide donors on insulin secre-
tion, cyclic GMP and cyclic AMP in rat islets of Langer-
hans and the insulin-secreting cell lines HIT-T15 and
RINm5F. Mol Cell Endocrinol 102: 23±29
44. Salehi A, Parandeh F, Lundquist I (1998) Signal transduc-
tion in islet hormone relase: interaction of nitric oxide
with basal and nutrient-induced hormone responses. Cellu-
lar Signalling 10: 645±651
45. Henningsson R, Lundquist I (1998) Arginine-induced re-
lease of insulin is decreased and glucagon increased in par-
allel with islet NO-production. Am J Physiol 275:
E500±E506
46. kesson B, Lundquist I (1998) Evidence for nitric oxide
mediated effects on islet hormone secretory phospholipase
C signal transduction mechanisms. Biosci Rep 18: 199±213
47. Sjöholm (1996) Nitric oxide donor SIN-1 inhibits insulin
release. Am J Physiol 271: C1098±C1102
48. Stamler JS, Simon DI, Osborne JA et al. (1992) S-nitrosy-
lation of proteins with nitric oxide: Synthesis and charac-
terization of biologically active compounds. Proc Natl
Acad Sci USA 89: 444±448
49. Hellman B, Idahl L-, Lernmark , Sehlin J, Täljedal I-B
(1974) Membrane sulphydryl groups and pancreatic beta
cell recognition of insulin secretagogues. Exerpt Med Int
Congr Ser 312: 65±78
50. Ammon HPT, Mark M (1985) Thiols and pancreatic b-cell
function: a review. Cell Biochem Funct 3: 157±171
P.Alm et al.: Islet nitric oxide synthase and haem oxygenase986
... Haem oxygenase (HO) is the rate-limiting enzyme resulting in degradation of haem to generate endogenous CO [4]. Consistent with the increased insulin secretion in diabetic mice, expression level of the inducible HO-1 in pancreatic islets is also up-regulated [5][6][7][8]. Furthermore, CO has been reported to increase glucose-stimulated insulin secretion (GSIS) in leptin-deficient ob/ob mice [9,10]. ...
... Our study showed that, similar to the mechanism of acetylcholine, GLP-1 and IL-6, CO also promoted βcell function by activating PLC signaling pathway but mostly acted in a compensatory manner under pathological conditions. This is likely due to lower expressions of HO-1 [5][6][7][8], PLCγ1 and PLCδ1 isoforms in normal islets and an up-regulated expressions of them in diabetic islets. However, more investigation is required to examine the changes in the expression levels of different PLC isoforms between normal islets and diabetic islets. ...
Article
In early stage of diabetes, insulin secretion from pancreatic β-cells is increased to deal with the elevated blood glucose. Previous studies have reported that islet-produced carbon monoxide (CO) is associated with increased glucose-stimulated insulin secretion from β-cells. However, this compensatory mechanism by which CO may act to enhance β-cell function remain unclear. In this study, we revealed that CO promoted intracellular calcium ([Ca²⁺]i) elevation and glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells in leptin receptor deficient db/db mice but not in C57 mice. The stimulatory effects of CO on β-cell function in db/db mice was blocked by inhibition of Phospholipase C (PLC) signaling pathway. We further demonstrated that CO triggered [Ca²⁺]i transients and enhanced GSIS in C57 islets when β-cells overexpressed with PLCγ1 and PLCδ1, but not PLCβ1. On the other hand, reducing PLCγ1 and PLCδ1 expressions in db/db islets dramatically attenuated the stimulatory effects of CO on β-cell function, whereas interfering PLCβ1 expression had no effects on CO-induced β-cell function enhancement. Our findings showing that CO elevated [Ca²⁺]i and enhanced GSIS by activating PLC signaling through PLCγ1 and PLCδ1 isoforms in db/db pancreatic β-cells may suggest an important mechanism by which CO promotes β-cell function to prevent hyperglycemia. Our study may also provide new insights into the therapy for type II diabetes and offer a potential target for therapeutic applications of CO.
... The present investigation also showed that two months of nitrate therapy was associated with increased plasma insulin levels and body weight and decreased blood glucose concentration. The increased plasma insulin levels are possibly caused by nitrate induce-enhancing the pancreatic blood flow (Alm et al. 1999). Some studies have suggested that treatment with nitrate, nitrite, constitutive NOS (cNOS), and even L-arginine, as a NO precursor, increased the serum insulin levels (Schmidt et al. 1992;Alm et al. 1999;Carlstrom et al. 2010;Nystrom et al. 2012). ...
... The increased plasma insulin levels are possibly caused by nitrate induce-enhancing the pancreatic blood flow (Alm et al. 1999). Some studies have suggested that treatment with nitrate, nitrite, constitutive NOS (cNOS), and even L-arginine, as a NO precursor, increased the serum insulin levels (Schmidt et al. 1992;Alm et al. 1999;Carlstrom et al. 2010;Nystrom et al. 2012). Jeddi et al. (2016) have also reported that dietary nitrate improved glucose tolerance and prevented an increase in blood glucose levels. ...
Article
Full-text available
Objective. Diabetes induces sensory symptoms of neuropathy as positive (hyperalgesia), negative (hypoalgesia), or both. Methods. In the present study, fifty male Wistar rats were allocated to five groups: control, control+nitrate, diabetes, diabetes+insulin, and diabetes+nitrate. Thirty days after diabetes confirmation, insulin (2–4 U/day) was injected subcutaneously in diabetes+insulin group and nitrate (100 mg/l) was added into drinking water of the control+nitrate and diabetes+nitrate groups for a period of 2 months. In order to assess the mechanical and thermal algesia, tail immersion, hot plate, and von Frey tests were performed. The serum insulin levels were determined with insulin ELISA Kit. Serum level of NOx was determined by the Griess method. Results. Both thermal and mechanical nociceptive thresholds showed a significant decrease (p<0.05) which was followed by a significant increase (p<0.01) in the thermal nociceptive threshold in the diabetes group. Chronic nitrate or insulin treatment led to a significant decrease (p<0.01) in blood glucose levels, as well as a significant (p<0.05) increase in the body weight and serum NOx. Moreover, nitrate treatment significantly increased serum insulin levels (p<0.001) compared to the other groups. Conclusion. Chronic nitrate treatment modified the thermal and mechanical sensitivities in diabetic animals.
... The present investigation also showed that two months of nitrate therapy was associated with increased plasma insulin levels and body weight and decreased blood glucose concentration. The increased plasma insulin levels are possibly caused by nitrate induce-enhancing the pancreatic blood flow (Alm et al. 1999). Some studies have suggested that treatment with nitrate, nitrite, constitutive NOS (cNOS), and even L-arginine, as a NO precursor, increased the serum insulin levels (Schmidt et al. 1992;Alm et al. 1999;Carlstrom et al. 2010;Nystrom et al. 2012). ...
... The increased plasma insulin levels are possibly caused by nitrate induce-enhancing the pancreatic blood flow (Alm et al. 1999). Some studies have suggested that treatment with nitrate, nitrite, constitutive NOS (cNOS), and even L-arginine, as a NO precursor, increased the serum insulin levels (Schmidt et al. 1992;Alm et al. 1999;Carlstrom et al. 2010;Nystrom et al. 2012). Jeddi et al. (2016) have also reported that dietary nitrate improved glucose tolerance and prevented an increase in blood glucose levels. ...
Article
Objective. Diabetes induces sensory symptoms of neuropathy as positive (hyperalgesia), negative (hypoalgesia), or both. Methods. In the present study, fifty male Wistar rats were allocated to five groups: control, control+nitrate, diabetes, diabetes+insulin, and diabetes+nitrate. Thirty days after diabetes confirmation , insulin (2-4 U/day) was injected subcutaneously in diabetes+insulin group and nitrate (100 mg/l) was added into drinking water of the control+nitrate and diabetes+nitrate groups for a period of 2 months. In order to assess the mechanical and thermal algesia, tail immersion, hot plate, and von Frey tests were performed. The serum insulin levels were determined with insulin ELISA Kit. Serum level of NOx was determined by the Griess method. Results. Both thermal and mechanical nociceptive thresholds showed a significant decrease (p<0.05) which was followed by a significant increase (p<0.01) in the thermal nociceptive threshold in the diabetes group. Chronic nitrate or insulin treatment led to a significant decrease (p<0.01) in blood glucose levels, as well as a significant (p<0.05) increase in the body weight and serum NOx. Moreover, nitrate treatment significantly increased serum insulin levels (p<0.001) compared to the other groups. Conclusion. Chronic nitrate treatment modified the thermal and mechanical sensitivities in diabetic animals.
... The presence and localization of the NO-generating enzymes NOS in various organs including pancreas are subject to controversy. By many authors a high expression of the constitutive NOS isoforms (NOS1 and NOS3) and the inducible NOS isoform (NOS2) was detected exclusively in pancreatic islets but not in the exocrine compartment or in the vasculature [5,15,25,35,45]. ...
... We have also shown that in Langerhans islets NOS1 and NOS2 were immunolabeled generally equally in all islet cells, whereas NOS3 revealed a stronger preferential immunostaining in single scattered cells that apparently might correspond to the cells that were earlier reported by other authors as somatostatin-, glucagon-or insulinimmunoreactive cells [5,15,25,45]. This might be indicative of an autocrine fashion of NO signaling in the regulation of endocrine as well as of exocrine secretion. ...
Article
Full-text available
Nitric oxide (NO) is generated by a family of enzymes termed NO synthases (NOS) that convert L-arginine to NO and citrulline. The role of NO as an important biological mediator and recognition of the pathophysiological significance of superoxides/NO interaction has led to an intensive research and development of therapies based on the interception of the NO signaling cascade in the pancreatitis course. However, the presence and localization of the NO-generating enzymes in various organs including pancreas are subject to controversy. We assumed that this controversy might reflect rather the diversity of experimental approaches and an insufficient sensitivity of the methods used. Applying tyramide signal amplification (TSA) immunohistochemical technology, we were able detect all three NOS isoforms both in exocrine and endocrine compartments and in the vasculature in the normal pancreas and in pancreatitis. This also allowed us to demonstrate that oxidative stress runs ahead of NOS up-regulation, which implies that the NO enhancement in the course of pancreatitis is likely to be an adaptive mechanism aimed at maintaining the homeostatic cellular level of the bioactive NO. The aims of this minireview are to describe normal intrapancreatic NO pathways and the role of NO in the pancreatitis course.
... Cerulein induced pancreatitis has similar pathophysiological properties compared to human edematous pancreatitis, manifesting with dysregulation of digestive enzyme production and cytoplasmic vacuolization, the death of acinar cells, edema formation, and infiltration of inflammatory cells into the pancreas [14,15]. Earlier, we reported that, in contrast to the commonly accepted view, all three NOS isoforms are expressed not only in pancreatic islets [16][17][18][19], but also in the exocrine compartments and in the vasculature. In pancreatitis, the expression of NOS was up-regulated especially within the exocrine compartment [4,5], which reflects an exceptional vulnerability of the exocrine parenchyma. ...
Article
Full-text available
The interaction between nitric oxide (NO) and superoxides is critical in the development of pancreatitis. Previously, we reported on the up-regulation of oxidative stress and NO-synthase (NOS) in the human chronic pancreatitis and in an animal model of pancreatitis induced by pancreatic duct ligation (PDL) in rats. We have shown that oxidative stress runs ahead of NOS up-regulation, which implies that the NO enhancement in the course of pancreatitis is likely to be an adaptive mechanism aimed at maintaining the homeostatic cellular level of the bioactive NO. Here, we report on the expression of NOS and oxidative stress markers (nitrotyrosine and 8-hydroxyguanosine) in the course of cerulein-induced acute pancreatitis in rats. We found that the pattern of superoxides/NO interaction in this model of acute pancreatitis is similar to that in the PDL-induced rat pancreatitis and in the human chronic pancreatitis. It means that cerulein-induced acute pancreatitis like the PDL-induced pancreatitis is a proper model for further studies of pancreatitis development in humans.
... Cerulein induced pancreatitis has similar pathophysiological properties compared to human edematous pancreatitis, manifesting with dysregulation of digestive enzyme production and cytoplasmic vacuolization, the death of acinar cells, edema formation, and infiltration of inflammatory cells into the pancreas [14,15]. Earlier, we reported that, in contrast to the commonly accepted view, all three NOS isoforms are expressed not only in pancreatic islets [16][17][18][19], but also in the exocrine compartments and in the vasculature. In pancreatitis, the expression of NOS was up-regulated especially within the exocrine compartment [4,5], which reflects an exceptional vulnerability of the exocrine parenchyma. ...
... The excitatory effects of GABAA receptor blockade in the DMV on glucose-induced insulin secretion are enhanced in the presence of the nitric oxide (NO) synthase inhibitor L-NAME. This suggests that a nitrergic inhibitory pathway is involved in pancreatic insulin secretion [54,55] . Similarly, a considerable number of studies have shown that blockade of GABA receptors within the DMV has a profound effect on gastric emptying. ...
Article
Full-text available
Recently, diabetic gastroparesis (DGP) has received much attention as its prevalence is increasing in a dramatic fashion and management of patients with DGP represents a challenge in the clinical practice due to the limited therapeutic options. DGP highlights an interrelationship between the gastric emptying and pancreatic secretory function that regulate a wide range of digestive and metabolic functions, respectively. It well documented that both gastric emptying and pancreatic secretion are under delicate control by multiple neurohormonal mechanisms including extrinsic parasympathetic pathways and gastrointestinal (GI) hormones. Interestingly, the latter released in response to various determinants that related to the rate and quality of gastric emptying. Others and we have provided strong evidence that the central autonomic nuclei send a dual output (excitatory and inhibitory) to the stomach and the pancreas in response to a variety of hormonal signals from the abdominal viscera. Most of these hormones released upon gastric emptying to provide feedback, and control this process and simultaneously regulate pancreatic secretion and postprandial glycemia. These findings emphasize an important link between gastric emptying and pancreatic secretion and its role in maintaining homeostatic processes within the GI tract. The present review deals with the neurohormonal-coupled mechanisms of gastric emptying and pancreatic secretory function that implicated in DGP and this provides new insights in our understanding of the pathophysiology of DGP. This also enhances the process of identifying potential therapeutic targets to treat DGP and limit the complications of current management practices. Keywords: Gastroparesis, Gastric emptying, Pancreatic secretion, Postprandial glycemia, Neurohormonal control.
... The excitatory effects of GABAA receptor blockade in the DMV on glucose-induced insulin secretion are enhanced in the presence of the nitric oxide (NO) synthase inhibitor L-NAME. This suggests that a nitrergic inhibitory pathway is involved in pancreatic insulin secretion [54,55] . Similarly, a considerable number of studies have shown that blockade of GABA receptors within the DMV has a profound effect on gastric emptying. ...
Article
Full-text available
Recently, diabetic gastroparesis (DGP) has received much attention as its prevalence is increasing in a dramatic fashion and management of patients with DGP represents a challenge in the clinical practice due to the limited therapeutic options. DGP highlights an interrelationship between the gastric emptying and pancreatic secretory function that regulate a wide range of digestive and metabolic functions, respectively. It well documented that both gastric emptying and pancreatic secretion are under delicate control by multiple neurohormonal mechanisms including extrinsic parasympathetic pathways and gastrointestinal (GI) hormones. Interestingly, the latter released in response to various determinants that related to the rate and quality of gastric emptying. Others and we have provided strong evidence that the central autonomic nuclei send a dual output (excitatory and inhibitory) to the stomach and the pancreas in response to a variety of hormonal signals from the abdominal viscera. Most of these hormones released upon gastric emptying to provide feedback, and control this process and simultaneously regulate pancreatic secretion and postprandial glycemia. These findings emphasize an important link between gastric emptying and pancreatic secretion and its role in maintaining homeostatic processes within the GI tract. The present review deals with the neurohormonal-coupled mechanisms of gastric emptying and pancreatic secretory function that implicated in DGP and this provides new insights in our understanding of the pathophysiology of DGP. This also enhances the process of identifying potential therapeutic targets to treat DGP and limit the complications of current management practices.
Article
Carbon monoxide (CO), a member of the multifunctional gasotransmitters family produced by heme oxygenases (i.e., HO-1 and HO-2), has received significant attention because of its involvement in carbohydrate metabolism. Experimental evidence indicates that both HO-2- and HO-1-derived CO stimulate insulin secretion, but the latter mainly acts as a compensatory response in pre-diabetes conditions. CO protects pancreatic β-cell against cytokine- and hypoxia-induced apoptosis and promotes β-cell regeneration. CO cross-talks with nitric oxide (NO) and hydrogen sulfide (H2S), other important gasotransmitters in carbohydrate metabolism, in regulating β-cell function and insulin secretion. These data speak in favor of the potential therapeutic application of CO in type 2 diabetes mellitus (T2DM) and preventing the progression of pre-diabetes to diabetes. Either CO (as both gaseous form and CO-releasing molecule) or pharmacological formulations made of natural HO inducers (i.e., bioactive components originating from plant-based foods) are potential candidates for developing CO-based therapeutics in T2DM. Future studies are needed to assess the safety/efficacy and potential therapeutic applications of CO in T2DM.
Article
The possible implication of the gasotransmitters NO and CO for the development of diabetes remains unresolved. Our previous investigations in rodents suggested NO being inhibitory, and CO stimulatory, to glucose-stimulated insulin secretion (GSIS). Here we studied the possible role of these gasotransmitters in both murine and human type 2 diabetes (T2D) by mapping the expression pattern of neural nitric oxide synthase (nNOS), inducible NOS (iNOS), constitutive heme oxygenase (HO-2), and inducible HO (HO-1) in isolated pancreatic islets. Two variants of obese murine diabetes with distinct phenotype, the db/db and the ob/ob mouse, were studied at the initiation of the diabetic condition. Plasma glucose and plasma insulin were recorded and β-cell expression levels of the different enzymes were measured with confocal microscopy and fluorescence intensity recordings. In human islets taken from nondiabetic controls (ND) and type 2 diabetes (T2D) the expression of the enzymes was analyzed by RNA-sequencing and qPCR. At the initiation of murine diabetes plasma glucose was slightly increased, whereas plasma insulin was extremely enhanced in both db/db and ob/ob mice. The β-cell expression of nNOS and iNOS was markedly increased over controls in db/db mice, known to develop severe diabetes, while it was very low in ob/ob mice, known to develop mild diabetes. HO-2 expression was unaffected in db/db and modestly decreased in ob/ob mice. HO-1 expression was slightly enhanced in ob/ob, but, in contrast, extremely enhanced in db/db mice, suggesting a counteracting, antidiabetic action by CO. Moreover, the diabetic pattern of highly increased nNOS, iNOS and HO-1 expression seen in db/db mice was also fully recognized in human T2D islets. These results suggest that increased expression of the NOS-enzymes, especially an early upregulation of nNOS, could be involved in the initial development of the severe diabetes of db/db mice as well as in human T2D. Hence, nNOS, iNOS and HO-1 might be regarded as interesting targets to take into consideration in the early treatment of a diabetic condition in different variants of T2D.
Article
Full-text available
Summary Cytokines, released in and around pancreatic islets during insulitis, have been proposed to participate in B-cell destruction associated with autoimmune diabetes. In this study we have evaluated the hypothesis that local release of the cytokine interleukin 1 (IL-1) by nonendocrine cells of the islet induce the expression of inducible nitric oxide synthase (iNOS) by 3 cells which results in the inhibition of 3 cell function. Treatment of rat islets with a combination of tumor necrosis factor (TNF) and lipopolysaccharide (LPS), conditions known to activate macrophages, stimulate the expression ofiNOS and the formation of nitrite. Although TNF + LPS induce iNOS expression and inhibit insulin secretion by intact islets, this combination does not induce the expression of iNOS by 3 or o~ cells purified by fluorescence activated cell sorting (Facs). In contrast, IL-13 induces the expression of iNOS and also inhibits insulin secretion by both intact islets and Facs- purified 3 cells, whereas TNF+LPS have no inhibitory effects on insulin secretion by purified 3 cells. Evidence suggests that TNF+LPS inhibit insulin secretion from islets by stimulating the release of IL-1 which subsequently induces the expression of iNOS by 3 cells. The IL-1 receptor antagonist protein completely prevents TNF+ LPS-induced inhibition of insulin secretion and attenuates nitrite formation from islets, and neutralization of IL-1 with antisera specific for IL- 1 oL and IL- 13 attenuates TNF + LPS-induced nitrite formation by islet s. Immunohistochemical localization of iNOS and insulin confirm that TNF+LPS induce the expression of iNOS by islet 3 cells, and that a small percentage of noninsulin-containing cells also express iNOS. Local release of IL-1 within islets appears to be required for TNF+LPS-induced inhibition of insulin secretion because TNF+LPS do not stimulate nitrite formation from islets physically separated into individual cells. These findings provide the first evidence that a limited number of nonendocrine cells can release sufficient quantities of IL-1 in islets to induce iNOS expression and inhibit the function of the 3 cell, which is selectively destroyed during the development of autoimmune diabetes.
Article
Cytokines, released in and around pancreatic islets during insulitis, have been proposed to participate in beta-cell destruction associated with autoimmune diabetes. In this study we have evaluated the hypothesis that local release of the cytokine interleukin 1 (IL-1) by nonendocrine cells of the islet induce the expression of inducible nitric oxide synthase (iNOS) by beta cells which results in the inhibition of beta cell function. Treatment of rat islets with a combination of tumor necrosis factor (TNF) and lipopolysaccharide (LPS), conditions known to activate macrophages, stimulate the expression of iNOS and the formation of nitrite. Although TNF+LPS induce iNOS expression and inhibit insulin secretion by intact islets, this combination does not induce the expression of iNOS by beta or alpha cells purified by fluorescence activated cell sorting (Facs). In contrast, IL-1 beta induces the expression of iNOS and also inhibits insulin secretion by both intact islets and Facs-purified beta cells, whereas TNF+LPS have no inhibitory effects on insulin secretion by purified beta cells. Evidence suggests that TNF+LPS inhibit insulin secretion from islets by stimulating the release of IL-1 which subsequently induces the expression of iNOS by beta cells. The IL-1 receptor antagonist protein completely prevents TNF+LPS-induced inhibition of insulin secretion and attenuates nitrite formation from islets, and neutralization of IL-1 with antisera specific for IL-1 alpha and IL-1 beta attenuates TNF+LPS-induced nitrite formation by islets. Immunohistochemical localization of iNOS and insulin confirm that TNF+LPS induce the expression of iNOS by islet beta cells, and that a small percentage of noninsulin-containing cells also express iNOS. Local release of IL-1 within islets appears to be required for TNF+LPS-induced inhibition of insulin secretion because TNF+LPS do not stimulate nitrite formation from islets physically separated into individual cells. These findings provide the first evidence that a limited number of nonendocrine cells can release sufficient quantities of IL-1 in islets to induce iNOS expression and inhibit the function of the beta cell, which is selectively destroyed during the development of autoimmune diabetes.
Article
Endothelium-derived relaxing factor (EDRF) activity has been attributed to the highly labile nitric oxide radical (NO). In view of the fact that the plasma and cellular milieux contain reactive species that can rapidly inactivate NO, it has been postulated that NO is stabilized by a carrier molecule that preserves its biological activity. Reduced thiol species are candidates for this role, reacting readily in the presence of NO to yield biologically active S-nitrosothiols that are more stable than NO itself. Because sulfhydryl groups in proteins represent an abundant source of reduced thiol in biologic systems, we examined the reaction of several sulfhydryl-containing proteins of diverse nature and function upon exposure to authentic NO and EDRF. We demonstrate that S-nitroso proteins form readily under physiologic conditions and possess EDRF-like effects of vasodilation and platelet inhibition. These observations suggest that S-nitrosothiol groups in proteins may serve as intermediates in the cellular metabolism of NO and raise the possibility of an additional type of cellular regulatory mechanism.
Article
We examined the relation between the islet NO system and islet hormone secretion induced by either the non-glucose nutrient α-ketoisocaproic acid (KIC) or, in some experiments, glucose. KIC dose dependently stimulated insulin but inhibited glucagon secretion. In a medium devoid of any nutrient, the NO synthase (NOS)-inhibitor NG-nitro-l-arginine methyl ester (l-NAME) induced an increase in basal insulin release but a decrease in glucagon release. These effects were evident also in K+-depolarised islets. KIC-induced insulin release was increased by l-NAME. This increase was abolished in K+-depolarised islets. In contrast, glucose- induced insulin release was potentiated by l-NAME after K+ depolarisation. The intracellular NO donor hydroxylamine dose dependently inhibited KIC-stimulated insulin release and reversed KIC-induced suppression of glucagon release. Our data suggest that islet hormone secretion in a medium devoid of nutrients is greatly affected by the islet NO system, whereas KIC-induced secretion is little affected. Glucose-induced insulin release, however, is accompanied by increased NOS activity, the NOS-activating signal being derived from the glycolytic–pentose shunt part of glucose metabolism. The observed NO effects on islet hormone release can proceed independently of membrane-depolarisation events.
Article
The cytokine IL-1β has previously been demonstrated to induce the expression of the stress genes iNOS, hsp70, heme oxygenase and Mn-SOD in rat pancreatic islets in vitro. The aim of this study was to determine whether the IL-1β-induced effects are specific for the insulin producing β-cell, or whether other islet cells, such as the glucagon-producing α-cell, respond to IL-1β addition. Purified rat α- and β-cell suspensions were obtained by fluorescence-activated cell sorting and incubated with or without IL-1β (25 U/ml) for 24 h. The α- and β-cell contents of hsp70, heme oxygenase and Mn-SOD and medium nitrite levels were determined. It was found that IL-1β exposure induced the production of nitric oxide in β-cells, but not in α-cells. Moreover, the expression of hsp70, heme oxygenase and Mn-SOD was also induced in β-cells, but not in α-cells. There were no detectable levels of hsp70 in α-cells. It is concluded that the stress gene response following IL-1β exposure in markedly different in α- and β-cells. This finding may be of importance for the understanding of the autoimmune destruction of β-cells in insulin-dependent diabetes mellitus.