1
|
Aluko EO, Nna VU, Fasanmade AA. Angiotensin converting enzyme inhibitor potentiates the hypoglycaemic effect of NG-nitro-L-arginine methyl ester (L-NAME) in rats. Arch Physiol Biochem 2022; 128:1524-1532. [PMID: 32584611 DOI: 10.1080/13813455.2020.1780263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The inhibition of renin angiotensin system pathway has been largely documented to be effective in the control of cardiovascular events. The present study investigated the effect of angiotensin converting enzyme (ACE) inhibitor on fasting blood glucose level in hypertension induced by the inhibition of nitric oxide synthase (NOS) in male Wistar rats. Hypertension was induced by the inhibition of NOS using a non-selective NOS inhibitor, NG-nitro-L-arginine methyl ester (L-NAME). The blockade of NOS resulted in an increase in blood pressure, ACE, angiotensin II and endothelin-1 levels, and a decrease in fasting blood glucose and nitric oxide (NO) levels. The hypertensive rats treated with ACE inhibitor (ramipril) recorded a decrease in blood pressure, ACE, angiotensin II, endothelin-1, NO and fasting blood glucose levels, and an increase in prostacyclin level. In conclusion, ACE inhibitor potentiated the hypoglycaemic effect of NOS inhibitor and this effect is independent of NO and pancreatic insulin release.
Collapse
Affiliation(s)
- Esther Oluwasola Aluko
- Department of Physiology, Faculty of Basic Medical Sciences, University of Uyo, Uyo, Akwa Ibom State, Nigeria
| | - Victor Udo Nna
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | | |
Collapse
|
2
|
Andreone L, Fuertes F, Sétula C, Barcala Tabarrozzi AE, Orellano MS, Dewey RA, Bottino R, De Bosscher K, Perone MJ. Compound A attenuates proinflammatory cytokine-induced endoplasmic reticulum stress in beta cells and displays beneficial therapeutic effects in a mouse model of autoimmune diabetes. Cell Mol Life Sci 2022; 79:587. [PMID: 36370223 DOI: 10.1007/s00018-022-04615-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/15/2022]
Abstract
Type 1 diabetes (T1D) is characterized by an immune-mediated progressive destruction of the insulin-producing β-cells. Proinflammatory cytokines trigger endoplasmic reticulum (ER) stress and subsequent insulin secretory deficiency in cultured β-cells, mimicking the islet microenvironment in T1D. β-cells undergo physiologic ER stress due to the high rate of insulin production and secretion under stimulated conditions. Severe and uncompensated ER stress in β-cells is induced by several pathological mechanisms before onset and during T1D. We previously described that the small drug Compound A (CpdA), a selective glucocorticoid receptor (GR/NR3C1, nuclear receptor subfamily 3, group C, member 1) ligand with demonstrated inflammation-suppressive activity in vivo, is an effective modulator of effector T and dendritic cells and of macrophages, yet, in a GR-independent manner. Here, we focus on CpdA's therapeutic potential in T1D cellular and animal models. We demonstrate that CpdA improves the unfolded protein response (UPR) by attenuating ER stress and favoring the survival and function of β-cells exposed to an environment of proinflammatory cytokines. CpdA administration to NODscid mice adoptively transferred with diabetogenic splenocytes (from diabetic NOD mice) led to a delay of disease onset and reduction of diabetes incidence. Histological analysis of the pancreas showed a reduction in islet leukocyte infiltration (insulitis) and preservation of insulin expression in CpdA-treated normoglycemic mice in comparison with control group. These new findings together with our previous reports justify further studies on the administration of this small molecule as a novel therapeutic strategy with dual targets (effector immune and β-cells) during autoimmune diabetes.
Collapse
Affiliation(s)
- Luz Andreone
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Florencia Fuertes
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Carolina Sétula
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Andres E Barcala Tabarrozzi
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Miranda S Orellano
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Ricardo A Dewey
- Laboratorio de Terapia Génica Y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Buenos Aires, Argentina
| | - Rita Bottino
- Imagine Pharma, Pittsburgh, Pennsylvania, PA and Allegheny Health Network, Pittsburgh, PA, USA
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-Department of Medical Protein Research, VIB, Ghent University, Ghent, Belgium
| | - Marcelo J Perone
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina.
| |
Collapse
|
3
|
Yokoyama A, Suzuki S, Okamoto K, Sugawara A. The physiological and pathophysiological roles of carbohydrate response element binding protein in the kidney. Endocr J 2022; 69:605-612. [PMID: 35474028 DOI: 10.1507/endocrj.ej22-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Glucose is not only the energy fuel for most cells, but also the signaling molecule which affects gene expression via carbohydrate response element binding protein (ChREBP), a Mondo family transcription factor. In response to high glucose conditions, ChREBP regulates glycolytic and lipogenic genes by binding to carbohydrate response elements (ChoRE) in the regulatory region of its target genes, thus elucidating the role of ChREBP for converting excessively ingested carbohydrates to fatty acids as an energy storage in lipogenic tissues such as the liver and adipose tissue. While the pathophysiological roles of ChREBP for fatty liver and obesity in these tissues are well known, much of the physiological and pathophysiological roles of ChREBP in other tissues such as the kidney remains unclear despite its high levels of expression in them. This review will thus highlight the roles of ChREBP in the kidney and briefly introduce the latest research results that have been reported so far.
Collapse
Affiliation(s)
- Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Susumu Suzuki
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Koji Okamoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| |
Collapse
|
4
|
OKAMOTO H, TAKASAWA S. Okamoto model for necrosis and its expansions, CD38-cyclic ADP-ribose signal system for intracellular Ca 2+ mobilization and Reg (Regenerating gene protein)-Reg receptor system for cell regeneration. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:423-461. [PMID: 34629354 PMCID: PMC8553518 DOI: 10.2183/pjab.97.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/22/2021] [Indexed: 05/03/2023]
Abstract
In pancreatic islet cell culture models and animal models, we studied the molecular mechanisms involved in the development of insulin-dependent diabetes. The diabetogenic agents, alloxan and streptozotocin, caused DNA strand breaks, which in turn activated poly(ADP-ribose) polymerase/synthetase (PARP) to deplete NAD+, thereby inhibiting islet β-cell functions such as proinsulin synthesis and ultimately leading to β-cell necrosis. Radical scavengers protected against the formation of DNA strand breaks and inhibition of proinsulin synthesis. Inhibitors of PARP prevented the NAD+ depletion, inhibition of proinsulin synthesis and β-cell death. These findings led to the proposed unifying concept for β-cell damage and its prevention (the Okamoto model). The model met one proof with PARP knockout animals and was further extended by the discovery of cyclic ADP-ribose as the second messenger for Ca2+ mobilization in glucose-induced insulin secretion and by the identification of Reg (Regenerating gene) for β-cell regeneration. Physiological and pathological events found in pancreatic β-cells have been observed in other cells and tissues.
Collapse
Affiliation(s)
- Hiroshi OKAMOTO
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Shin TAKASAWA
- Department of Biochemistry, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
5
|
Suzuki S, Yokoyama A, Noro E, Aoki S, Shimizu K, Shimada H, Sugawara A. Expression and pathophysiological significance of carbohydrate response element binding protein (ChREBP) in the renal tubules of diabetic kidney. Endocr J 2020; 67:335-345. [PMID: 31813922 DOI: 10.1507/endocrj.ej19-0133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Carbohydrate response element binding protein (ChREBP), a glucose responsive transcription factor, mainly regulates expression of genes involved in glucose metabolism and lipogenesis. Recently, ChREBP is speculated to be involved in the onset and progression of diabetic nephropathy (DN). However, there exists no report regarding the localization and function of ChREBP in the kidney. Therefore, we analyzed the localization of Chrebp mRNA expression in the wild type (WT) mice kidney using laser microdissection method, and observed its dominant expression in the proximal tubules. In diabetic mice, mRNA expression of Chrebp target genes in the proximal tubules, including Chrebpβ and thioredoxin-interacting protein (Txnip), significantly increased comparing with that of WT mice. Co-overexpression of ChREBP and its partner Mlx, in the absence of glucose, also increased TXNIP mRNA expression as well as high glucose in human proximal tubular epithelial cell line HK-2. Since TXNIP is well known to be involved in the production of reactive oxygen species (ROS), we next examined the effect of ChREBP/Mlx co-overexpression, in the absence of glucose, on ROS production in HK-2 cells. Interestingly, ChREBP/Mlx co-overexpression also induced ROS production significantly as well as high glucose. Moreover, both high glucose-induced increase of TXNIP mRNA expression and ROS production were abrogated by ChREBP small interfering RNA transfection. Taken together, high glucose-activated ChREBP in the renal proximal tubules induce the expression of TXNIP mRNA, resulting in the production of ROS which may cause renal tubular damage. It is therefore speculated that ChREBP is involved in the onset and progression of DN.
Collapse
Affiliation(s)
- Susumu Suzuki
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Erika Noro
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Satoshi Aoki
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Kyoko Shimizu
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiroki Shimada
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| |
Collapse
|
6
|
Oleson BJ, Corbett JA. Dual Role of Nitric Oxide in Regulating the Response of β Cells to DNA Damage. Antioxid Redox Signal 2018; 29:1432-1445. [PMID: 28978225 PMCID: PMC6166691 DOI: 10.1089/ars.2017.7351] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/09/2017] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Cytokines released in and around pancreatic islets during islet inflammation are believed to contribute to impaired β cell function and β cell death during the development of diabetes. Nitric oxide, produced by β cells in response to cytokine exposure, controls many of the responses of β cells during islet inflammation. Recent Advances: Although nitric oxide has been shown to inhibit insulin secretion and oxidative metabolism and induce DNA damage in β cells, it also activates protective pathways that promote recovery of insulin secretion and oxidative metabolism and repair of damaged DNA. Recent studies have identified a novel role for nitric oxide in selectively regulating the DNA damage response in β cells. CRITICAL ISSUES Does nitric oxide mediate cytokine-induced β cell damage, or is nitric oxide produced by β cells in response to cytokines to protect β cells from damage? FUTURE DIRECTIONS β cells appear to be the only islet endocrine cell type capable of responding to proinflammatory cytokines with the production of nitric oxide, and these terminally differentiated cells have a limited capacity to regenerate. It is likely that there is a physiological purpose for this response, and understanding this could open new areas of study regarding the loss of functional β cell mass during diabetes development.
Collapse
Affiliation(s)
- Bryndon J. Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John A. Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
7
|
Jiang X, Zhou Y, Wu KKL, Chen Z, Xu A, Cheng KKY. APPL1 prevents pancreatic beta cell death and inflammation by dampening NFκB activation in a mouse model of type 1 diabetes. Diabetologia 2017; 60:464-474. [PMID: 28011992 DOI: 10.1007/s00125-016-4185-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 11/17/2016] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS Beta cell inflammation and demise is a feature of type 1 diabetes. The insulin-sensitising molecule 'adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1' (APPL1), which contains an NH2-terminal Bin/Amphiphysin/Rvs domain, a central pleckstrin homology domain and a COOH-terminal phosphotyrosine-binding domain, has been shown to modulate inflammatory response in various cell types but its role in regulating beta cell mass and inflammation in type 1 diabetes remains unknown. Thus, we investigated whether APPL1 prevents beta cell apoptosis and inflammation in diabetes. METHODS Appl1-knockout mice and their wild-type littermates, as well as C57BL/6N mice injected with adeno-associated virus encoding APPL1 or green fluorescent protein, were treated with multiple-low-dose streptozotocin (MLDS) to induce experimental type 1 diabetes. Their glucose metabolism and beta cell function were assessed. The effect of APPL1 deficiency on beta cell function upon exposure to a diabetogenic cytokine cocktail (CKS; consisting of TNF-α, IL-1β and IFN-γ) was assessed ex vivo. RESULTS Expression of APPL1 was significantly reduced in pancreatic islets from mouse models of type 1 diabetes or islets treated with CKS. Hyperglycaemia, beta cell loss and insulitis induced by MLDS were exacerbated by genetic deletion of Appl1 but were alleviated by beta cell-specific overexpression of APPL1. APPL1 preserved beta cell mass by reducing beta cell apoptosis upon treatment with MLDS. Mechanistically, APPL1 deficiency potentiate CKS-induced phosphorylation of NFκB inhibitor, α (IκBα) and subsequent phosphorylation and transcriptional activation of p65, leading to a dramatic induction of NFκB-regulated apoptotic and proinflammatory programs in beta cells. Pharmacological inhibition of NFκB or inducible NO synthase (iNOS) largely abrogate the detrimental effects of APPL1 deficiency on beta cell functions. CONCLUSIONS/INTERPRETATION APPL1 negatively regulates inflammation and apoptosis in pancreatic beta cells by dampening the NFκB-iNOS-NO axis, representing a promising target for treating type 1 diabetes.
Collapse
Affiliation(s)
- Xue Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, L8, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Yawen Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, L8, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kelvin K L Wu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, L8, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Zhanrui Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, L8, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, L8, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
| | - Kenneth K Y Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, L8, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
| |
Collapse
|
8
|
Lundquist I, Mohammed Al-Amily I, Meidute Abaraviciene S, Salehi A. Metformin Ameliorates Dysfunctional Traits of Glibenclamide- and Glucose-Induced Insulin Secretion by Suppression of Imposed Overactivity of the Islet Nitric Oxide Synthase-NO System. PLoS One 2016; 11:e0165668. [PMID: 27820841 PMCID: PMC5098820 DOI: 10.1371/journal.pone.0165668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/14/2016] [Indexed: 01/15/2023] Open
Abstract
Metformin lowers diabetic blood glucose primarily by reducing hepatic gluconeogenesis and increasing peripheral glucose uptake. However, possible effects by metformin on beta-cell function are incompletely understood. We speculated that metformin might positively influence insulin secretion through impacting the beta-cell nitric oxide synthase (NOS)-NO system, a negative modulator of glucose-stimulated insulin release. In short-time incubations with isolated murine islets either glibenclamide or high glucose augmented insulin release associated with increased NO production from both neural and inducible NOS. Metformin addition suppressed the augmented NO generation coinciding with amplified insulin release. Islet culturing with glibenclamide or high glucose revealed pronounced fluorescence of inducible NOS in the beta-cells being abolished by metformin co-culturing. These findings were reflected in medium nitrite-nitrate levels. A glucose challenge following islet culturing with glibenclamide or high glucose revealed markedly impaired insulin response. Metformin co-culturing restored this response. Culturing murine islets and human islets from controls and type 2 diabetics with high glucose or high glucose + glibenclamide induced a pronounced decrease of cell viability being remarkably restored by metformin co-culturing. We show here, that imposed overactivity of the beta-cell NOS-NO system by glibenclamide or high glucose leads to insulin secretory dysfunction and reduced cell viability and also, importantly, that these effects are relieved by metformin inhibiting beta-cell NO overproduction from both neural and inducible NOS thus ameliorating a concealed negative influence by NO induced by sulfonylurea treatment and/or high glucose levels. This double-edged effect of glibenclamide on the beta-cellsuggests sulfonylurea monotherapy in type 2 diabetes being avoided.
Collapse
Affiliation(s)
- Ingmar Lundquist
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
- Department of Experimental Medical Science, University of Lund, Lund, Sweden
| | - Israa Mohammed Al-Amily
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
| | - Sandra Meidute Abaraviciene
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
- Dept. of Physiology, Biochemistry, Microbiology and Laboratory Medicine Vilnius University, and Dep. of Regenerative Medicine, State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | - Albert Salehi
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
- Department of Neuroscience and Physiology, Metabolic Research Unit, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Assmann TS, Brondani LA, Bouças AP, Rheinheimer J, de Souza BM, Canani LH, Bauer AC, Crispim D. Nitric oxide levels in patients with diabetes mellitus: A systematic review and meta-analysis. Nitric Oxide 2016; 61:1-9. [PMID: 27677584 DOI: 10.1016/j.niox.2016.09.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/30/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Altered serum nitric oxide (NO) levels in patients with diabetes mellitus (DM) have been reported by different studies; however, results are still controversial. Until this date, no meta-analysis evaluated the association of NO levels with DM. Thus, this paper describes a meta-analysis conducted to evaluate if there is a relationship between NO levels and type 1 DM (T1DM) or type 2 DM (T2DM). METHODS A literature search was done to identify all studies that investigated NO levels between T1DM or T2DM patients (cases) and non-diabetic subjects (controls). Measurement of nitrate and nitrite (NOx - the stable NO products) were used to estimate NO concentrations because they closely reflect NO bioavailability. Weighted mean differences (WMD) of NOx levels between case and control samples were calculated for T1DM and T2DM groups. RESULTS Thirty studies were eligible for inclusion in the meta-analysis (8 in T1DM samples and 22 in T2DM samples). NOx levels were increased in European T1DM patients compared with controls [random effect model (REM) WMD = 8.55, 95% CI 2.88 - 14.21]. No other ethnicity was evaluated in T1DM studies. NOx levels were also increased in both European (REM WMD = 18.76, 95% CI 1.67 - 35.85) and Asian (REM WMD = 18.41, 95% CI 8.01 - 28.81) T2DM patients, but not in Latin American patients compared with controls. CONCLUSIONS This meta-analysis detected a significant increase in NOx levels in European T1DM patients as well as European and Asian T2DM patients. Further studies in other ethnicities are necessary to confirm these data.
Collapse
Affiliation(s)
- Taís S Assmann
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Letícia A Brondani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Ana P Bouças
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Jakeline Rheinheimer
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bianca M de Souza
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luís H Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andrea C Bauer
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
10
|
Abouzed TK, Munesue S, Harashima A, Masuo Y, Kato Y, Khailo K, Yamamoto H, Yamamoto Y. Preventive Effect of Salicylate and Pyridoxamine on Diabetic Nephropathy. J Diabetes Res 2016; 2016:1786789. [PMID: 28042580 PMCID: PMC5155113 DOI: 10.1155/2016/1786789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/31/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023] Open
Abstract
Objective. Diabetic nephropathy is a life-threatening complication in patients with long-standing diabetes. Hemodynamic, inflammatory, and metabolic factors are considered as developmental factors for diabetic nephropathy. In this study, we evaluated whether pharmacological interventions with salicylate, compared to pyridoxamine, could prevent diabetic nephropathy in mice. Methods. Male mice overexpressing inducible nitric oxide synthase in pancreatic β-cells were employed as a diabetic model. Salicylate (3 g/kg diet) or pyridoxamine (1 g/L drinking water; ~200 mg/kg/day) was given for 16 weeks to assess the development of diabetic nephropathy. Treatment with long-acting insulin (Levemir 2 units/kg twice a day) was used as a control. Results. Although higher blood glucose levels were not significantly affected by pyridoxamine, early to late stage indices of nephropathy were attenuated, including kidney enlargement, albuminuria, and increased serum creatinine, glomerulosclerosis, and inflammatory and profibrotic gene expressions. Salicylate showed beneficial effects on diabetic nephropathy similar to those of pyridoxamine, which include lowering blood glucose levels and inhibiting macrophage infiltration into the kidneys. Attenuation of macrophage infiltration into the kidneys and upregulation of antiglycating enzyme glyoxalase 1 gene expression were found only in the salicylate treatment group. Conclusions. Treatment with salicylate and pyridoxamine could prevent the development of diabetic nephropathy in mice and, therefore, would be a potentially useful therapeutic strategy against kidney problems in patients with diabetes.
Collapse
Affiliation(s)
- Tarek Kamal Abouzed
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8640, Japan
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El Sheikh 33516, Egypt
| | - Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8640, Japan
| | - Ai Harashima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8640, Japan
| | - Yusuke Masuo
- Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yukio Kato
- Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Khaled Khailo
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El Sheikh 33516, Egypt
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8640, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa 920-8640, Japan
- *Yasuhiko Yamamoto:
| |
Collapse
|
11
|
Wojcik M, Zieleniak A, Zurawska-Klis M, Cypryk K, Wozniak LA. Increased expression of immune-related genes in leukocytes of patients with diagnosed gestational diabetes mellitus (GDM). Exp Biol Med (Maywood) 2015; 241:457-65. [PMID: 26568332 DOI: 10.1177/1535370215615699] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/14/2015] [Indexed: 12/16/2022] Open
Abstract
Compelling evidence indicates that the immune system is linked to metabolism in gestational diabetes mellitus (GDM), but factors participating in these processes still are awaiting identification. Inducible nitric oxide synthase, encoded by the NOS2 gene, and surfactant protein D, encoded by the SFTPD gene, have been implicated in diabetes. We investigated NOS2 and SFTPD mRNA levels in leukocytes obtained from 125 pregnant women with (n = 87) or without (control group; n = 38) GDM, and, in turn, correlated their expression with clinical parameters of subjects. Leukocytes were isolated from the blood of pregnant women and NOS2 and SFTPD expression in these cells was determined by quantitative real time PCR (qRT-PCR). Univariate correlation analyses were performed to assess an association between leukocyte NOS2 and SFTPD expression and clinical characteristics of patients. qRT-PCR experiments disclosed significantly increased leukocyte NOS2 and SFTPD mRNA levels in hyperglycemic GDM patients (P < 0.05). In the entire study group, there were significant positive associations of leukocyte NOS2 and SFTPD mRNAs with C-reactive protein. Additionally, transcript level of SFTPD also correlated positively with fasting glycemia and insulin resistance. This study demonstrates that an impaired glucose metabolism in GDM may be predominant predictor of leukocyte NOS2 and SFTPD overexpression in diabetic patients. Furthermore, alterations in the expression of these genes are associated with glucose metabolism dysfunction and/or inflammation during pregnancy. In addition, these findings support the utilization of leukocytes as good experimental model to study a relationship between immune-related genes and metabolic changes in women with GDM, as well as to assess the potential mechanisms underlying these alterations.
Collapse
Affiliation(s)
- Marzena Wojcik
- Department of Structural Biology, Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, 90-752 Lodz, Poland
| | - Andrzej Zieleniak
- Department of Structural Biology, Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, 90-752 Lodz, Poland
| | - Monika Zurawska-Klis
- Diabetology and Metabolic Diseases Department, Medical University of Lodz, 92-213 Lodz, Poland Diabetological Medical Center "OmniMed", 93-338 Lodz, Poland
| | - Katarzyna Cypryk
- Diabetology and Metabolic Diseases Department, Medical University of Lodz, 92-213 Lodz, Poland Diabetological Medical Center "OmniMed", 93-338 Lodz, Poland
| | - Lucyna Alicja Wozniak
- Department of Structural Biology, Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, 90-752 Lodz, Poland
| |
Collapse
|
12
|
El-Refaei MF, Abduljawad SH, Alghamdi AH. Alternative Medicine in Diabetes - Role of Angiogenesis, Oxidative Stress, and Chronic Inflammation. Rev Diabet Stud 2015; 11:231-44. [PMID: 26177484 PMCID: PMC5397289 DOI: 10.1900/rds.2014.11.231] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/12/2015] [Accepted: 02/06/2015] [Indexed: 12/11/2022] Open
Abstract
Diabetes is a chronic metabolic disorder that is characterized by hyperglycemia due to lack of or resistance to insulin. Patients with diabetes are frequently afflicted with ischemic vascular disease and impaired wound healing. Type 2 diabetes is known to accelerate atherosclerotic processes, endothelial cell dysfunction, glycosylation of extracellular matrix proteins, and vascular denervation. Herbal medicines and naturally occurring substances may positively affect diabetes management, and could thus be utilized as cost-effective means of supporting treatment in developing countries. Natural treatments have been used in these countries for a long time to treat diabetes. The present review analyses the features of aberrant angiogenesis, abnormalities in growth factors, oxidative stress, and metabolic derangements relevant to diabetes, and how herbal substances and their active chemical constituents may counteract these events. Evidence for possible biochemical effectiveness and limitations of herbal medicines are given, as well as details regarding the role of cytokines and nitric oxide.
Collapse
|
13
|
Kaur M, Bedi O, Sachdeva S, Reddy BVKK, Kumar P. Rodent animal models: from mild to advanced stages of diabetic nephropathy. Inflammopharmacology 2014; 22:279-93. [PMID: 25149089 PMCID: PMC7101706 DOI: 10.1007/s10787-014-0215-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 08/08/2014] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy (DN) is a secondary complication of both type 1 and type 2 diabetes, resulting from uncontrolled high blood sugar. 30-40% of diabetic patients develop DN associated with a poor life expectancy and end-stage renal disease, causing serious socioeconomic problems. Although an exact pathogenesis of DN is still unknown, several factors such as hyperglycemia, hyperlipidemia, hypertension and proteinuria may contribute to the progression of renal damage in diabetic nephropathy. DN is confirmed by measuring blood urea nitrogen, serum creatinine, creatinine clearance and proteinuria. Clinical studies show that intensive control of hyperglycemia and blood pressure could successfully reduce proteinuria, which is the main sign of glomerular lesions in DN, and improve the renal prognosis in patients with DN. Diabetic rodent models have traditionally been used for doing research on pathogenesis and developing novel therapeutic strategies, but have limitations for translational research. Diabetes in animal models such as rodents are induced either spontaneously or by using chemical, surgical, genetic, or other techniques and depicts many clinical features or related phenotypes of the disease. This review discusses the merits and demerits of the models, which are used for many reasons in the research of diabetes and diabetic complications.
Collapse
Affiliation(s)
- Manpreet Kaur
- Pharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001 Punjab India
| | - Onkar Bedi
- Pharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001 Punjab India
| | - Shilpi Sachdeva
- Pharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001 Punjab India
| | - B. V. K. Krishna Reddy
- Pharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001 Punjab India
| | - Puneet Kumar
- Pharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001 Punjab India
| |
Collapse
|
14
|
Kim EJ, Kim J, Lee MY, Sudhanva MS, Devakumar S, Jeon YJ. Silymarin Inhibits Cytokine-Stimulated Pancreatic Beta Cells by Blocking the ERK1/2 Pathway. Biomol Ther (Seoul) 2014; 22:282-7. [PMID: 25143805 PMCID: PMC4131525 DOI: 10.4062/biomolther.2014.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 12/11/2022] Open
Abstract
We show that silymarin, a polyphenolic flavonoid isolated from milk thistle (Silybum marianum), inhibits cytokine mixture (CM: TNF-α, IFN-γ, and IL-1β)-induced production of nitric oxide (NO) in the pancreatic beta cell line MIN6N8a. Immunostaining and Western blot analysis showed that silymarin inhibits iNOS gene expression. RT-PCR showed that silymarin inhibits iNOS gene expression in a dose-dependent manner. We also showed that silymarin inhibits extracellular signal-regulated protein kinase-1 and 2 (ERK1/2) phosphorylation. A MEK1 inhibitor abrogated CM-induced nitrite production, similar to silymarin. Treatment of MIN6N8a cells with silymarin also inhibited CM-stimulated activation of NF-κB, which is important for iNOS transcription. Collectively, we demonstrate that silymarin inhibits NO production in pancreatic beta cells, and silymarin may represent a useful anti-diabetic agent.
Collapse
Affiliation(s)
- Eun Jeong Kim
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | - Jeeho Kim
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | - Min Young Lee
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | | | | | - Young Jin Jeon
- Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| |
Collapse
|
15
|
Sansbury BE, Hill BG. Regulation of obesity and insulin resistance by nitric oxide. Free Radic Biol Med 2014; 73:383-99. [PMID: 24878261 PMCID: PMC4112002 DOI: 10.1016/j.freeradbiomed.2014.05.016] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/16/2014] [Accepted: 05/17/2014] [Indexed: 02/07/2023]
Abstract
Obesity is a risk factor for developing type 2 diabetes and cardiovascular disease and has quickly become a worldwide pandemic with few tangible and safe treatment options. Although it is generally accepted that the primary cause of obesity is energy imbalance, i.e., the calories consumed are greater than are utilized, understanding how caloric balance is regulated has proven a challenge. Many "distal" causes of obesity, such as the structural environment, occupation, and social influences, are exceedingly difficult to change or manipulate. Hence, molecular processes and pathways more proximal to the origins of obesity-those that directly regulate energy metabolism or caloric intake-seem to be more feasible targets for therapy. In particular, nitric oxide (NO) is emerging as a central regulator of energy metabolism and body composition. NO bioavailability is decreased in animal models of diet-induced obesity and in obese and insulin-resistant patients, and increasing NO output has remarkable effects on obesity and insulin resistance. This review discusses the role of NO in regulating adiposity and insulin sensitivity and places its modes of action into context with the known causes and consequences of metabolic disease.
Collapse
Affiliation(s)
- Brian E Sansbury
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bradford G Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
16
|
Yamamoto Y, Yamamoto H. Controlling the receptor for advanced glycation end-products to conquer diabetic vascular complications. J Diabetes Investig 2014; 3:107-14. [PMID: 24843553 PMCID: PMC4020727 DOI: 10.1111/j.2040-1124.2011.00191.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Diabetic vascular complications, such as cardiovascular disease, stroke and microangiopathy, lead to high rates of morbidity and mortality in patients with long‐term diabetes. Extensive intracellular and extracellular formation of advanced glycation end‐products (AGE) is considered a causative factor in vascular injuries in diabetes. Receptor‐dependent mechanisms are involved in AGE‐induced cellular dysfunction and tissue damage. The receptor for AGE (RAGE), originally an AGE‐binding receptor, is now recognized as a member of pattern‐recognition receptors and a pro‐inflammatory molecular device that mediates danger signals to the body. Previous animal studies have shown RAGE dependent of diabetic vascular injuries. Prophylactic and therapeutic strategies focusing on RAGE and its ligand axis will be of great importance in conquering diabetic vascular complications. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2011.00191.x, 2012)
Collapse
Affiliation(s)
- Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
17
|
Wang T, Sun P, Chen L, Huang Q, Chen K, Jia Q, Li Y, Wang H. Cinnamtannin D-1 protects pancreatic β-cells from palmitic acid-induced apoptosis by attenuating oxidative stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:5038-5045. [PMID: 24815044 DOI: 10.1021/jf500387d] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
In previous studies, A-type procyanidin oligomers isolated from Cinnamomum tamala were proved to possess antidiabetic effect and protect pancreatic β-cells in vivo. The aim of this study was to unveil the mechanisms of protecting pancreatic β-cells from palmitic acid-induced apoptosis by cinnamtannin D-1 (CD1), one of the main A-type procyanidin oligomers in C. tamala. CD1 was discovered to dose-dependently reduce palmitic acid- or H2O2-induced apoptosis and oxidative stress in INS-1 cells, MIN6 cells, and primary cultured murine islets. Moreover, CD1 could reverse palmitic acid-induced dysfunction of glucose-stimulated insulin secretion in primary cultured islets. These results indicate that reduction of apoptosis and oxidative stress might account for the protection effect of CD1, which provided a better understanding of the mechanisms of the antidiabetic effects of procyanidin oligomers.
Collapse
Affiliation(s)
- Ting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Yagishita Y, Fukutomi T, Sugawara A, Kawamura H, Takahashi T, Pi J, Uruno A, Yamamoto M. Nrf2 protects pancreatic β-cells from oxidative and nitrosative stress in diabetic model mice. Diabetes 2014; 63:605-18. [PMID: 24186865 DOI: 10.2337/db13-0909] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transcription factor Nrf2 (NF-E2-related factor 2) regulates wide-ranging cytoprotective genes in response to environmental stress. Keap1 (Kelch-like ECH-associated protein 1) is an adaptor protein for Cullin3-based ubiquitin E3 ligase and negatively regulates Nrf2. The Keap1-Nrf2 system plays important roles in the oxidative stress response and metabolism. However, the roles Nrf2 plays in prevention of pancreatic β-cell damage remain elusive. To demonstrate the roles of Nrf2 in pancreatic β-cells, we used four genetically engineered mouse models: 1) β-cell-specific Keap1-conditional knockout mice, 2) β-cell-specific Nos2 transgenic mice, 3) conventional Nrf2-heterozygous knockout mice, and 4) β-cell-specific Nrf2-conditional knockout mice. We found that Nrf2 induction suppressed the oxidative DNA-adduct formation in pancreatic islets of iNOS-Tg mice and strongly restored insulin secretion from pancreatic β-cells in the context of reactive species (RS) damage. Consistently, Nrf2 suppressed accumulation of intracellular RS in isolated islets and pancreatic β-cell lines and also decreased nitrotyrosine levels. Nrf2 induced glutathione-related genes and reduced pancreatic β-cell apoptosis mediated by nitric oxide. In contrast, Nrf2 depletion in Nrf2-heterozygous knockout and β-cell-specific Nrf2-conditional knockout mice strongly aggravated pancreatic β-cell damage. These results demonstrate that Nrf2 induction prevents RS damage in pancreatic β-cells and that the Keap1-Nrf2 system is the crucial defense pathway for the physiological and pathological protection of pancreatic β-cells.
Collapse
Affiliation(s)
- Yoko Yagishita
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Autoimmune diabetes is characterized by the selective destruction of insulin-secreting β-cells that occurs during an inflammatory reaction in and around pancreatic islets of Langerhans. Cytokines such as interleukin-1, released by activated immune cells, have been shown to inhibit insulin secretion from pancreatic β-cells and cause islet destruction. In response to cytokines, β-cells express inducible nitric oxide synthase and produce micromolar levels of the free radical nitric oxide. Nitric oxide inhibits the mitochondrial oxidation of glucose resulting in an impairment of insulin secretion. Nitric oxide is also responsible for cytokine-mediated DNA damage in β-cells. While nitric oxide mediates the inhibitory and toxic effects of cytokines, it also activates protective pathways that allow β-cells to recover from this damage. This review will focus on the dual role of nitric oxide as a mediator of cytokine-induced damage and the activator of repair mechanisms that protect β-cells from cytokine-mediated injury.
Collapse
Affiliation(s)
| | - Bryndon J Oleson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
21
|
Broniowska KA, Mathews CE, Corbett JA. Do β-cells generate peroxynitrite in response to cytokine treatment? J Biol Chem 2013; 288:36567-78. [PMID: 24194521 DOI: 10.1074/jbc.m113.522243] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The purpose of this study was to determine the reactive species that is responsible for cytokine-mediated β-cell death. Inhibitors of inducible nitric oxide synthase prevent this death, and addition of exogenous nitric oxide using donors induces β-cell death. The reaction of nitric oxide with superoxide results in the generation of peroxynitrite, and this powerful oxidant has been suggested to be the mediator of β-cell death in response to cytokine treatment. Recently, coumarin-7-boronate has been developed as a probe for the selective detection of peroxynitrite. Using this reagent, we show that addition of the NADPH oxidase activator phorbol 12-myristate 13-acetate to nitric oxide-producing macrophages results in peroxynitrite generation. Using a similar approach, we demonstrate that cytokines fail to stimulate peroxynitrite generation by rat islets and insulinoma cells, either with or without phorbol 12-myristate 13-acetate treatment. When forced to produce superoxide using redox cyclers, this generation is associated with protection from nitric oxide toxicity. These findings indicate that: (i) nitric oxide is the likely mediator of the toxic effects of cytokines, (ii) β-cells do not produce peroxynitrite in response to cytokines, and (iii) when forced to produce superoxide, the scavenging of nitric oxide by superoxide is associated with protection of β-cells from nitric oxide-mediated toxicity.
Collapse
Affiliation(s)
- Katarzyna A Broniowska
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226 and
| | | | | |
Collapse
|
22
|
Youn CK, Park SJ, Li MH, Lee MY, Lee KY, Cha MJ, Kim OH, You HJ, Chang IY, Yoon SP, Jeon YJ. Radicicol Inhibits iNOS Expression in Cytokine-Stimulated Pancreatic Beta Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:315-20. [PMID: 23946691 PMCID: PMC3741488 DOI: 10.4196/kjpp.2013.17.4.315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/22/2013] [Accepted: 07/05/2013] [Indexed: 11/17/2022]
Abstract
Here, we show that radicicol, a fungal antibiotic, resulted in marked inhibition of inducible nitric oxide synthase (iNOS) transcription by the pancreatic beta cell line MIN6N8a in response to cytokine mixture (CM: TNF-α, IFN-γ, and IL-1β). Treatment of MIN6N8a cells with radicicol inhibited CM-stimulated activation of NF-κB/Rel, which plays a critical role in iNOS transcription, in a dose-related manner. Nitrite production in the presence of PD98059, a specific inhibitor of the extracellular signal-regulated protein kinase-1 and 2 (ERK1/2) pathway, was dramatically diminished, suggesting that the ERK1/2 pathway is involved in CM-induced iNOS expression. In contrast, SB203580, a specific inhibitor of p38, had no effect on nitrite generation. Collectively, this series of experiments indicates that radicicol inhibits iNOS gene expression by blocking ERK1/2 signaling. Due to the critical role that NO release plays in mediating destruction of pancreatic beta cells, the inhibitory effects of radicicol on iNOS expression suggest that radicicol may represent a useful anti-diabetic activity.
Collapse
Affiliation(s)
- Cha Kyung Youn
- DNA Damage Response Network Center, Chosun University, Kwangju 501-709, Korea. ; Department of Pharmacology, School of Medicine, Chosun University, Gwangju 501-759, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Thoonen R, Sips PY, Bloch KD, Buys ES. Pathophysiology of hypertension in the absence of nitric oxide/cyclic GMP signaling. Curr Hypertens Rep 2013; 15:47-58. [PMID: 23233080 DOI: 10.1007/s11906-012-0320-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signaling system is a well-characterized modulator of cardiovascular function, in general, and blood pressure, in particular. The availability of mice mutant for key enzymes in the NO-cGMP signaling system facilitated the identification of interactions with other blood pressure modifying pathways (e.g. the renin-angiotensin-aldosterone system) and of gender-specific effects of impaired NO-cGMP signaling. In addition, recent genome-wide association studies identified blood pressure-modifying genetic variants in genes that modulate NO and cGMP levels. Together, these findings have advanced our understanding of how NO-cGMP signaling regulates blood pressure. In this review, we will summarize the results obtained in mice with disrupted NO-cGMP signaling and highlight the relevance of this pathway as a potential therapeutic target for the treatment of hypertension.
Collapse
Affiliation(s)
- Robrecht Thoonen
- Molecular Cardiology Research Institute, Molecular Cardiology Research Center, Tufts Medical Center, Boston, MA 02111, USA.
| | | | | | | |
Collapse
|
24
|
Shibata K, Shimokawa H, Yanagihara N, Otsuji Y, Tsutsui M. Nitric oxide synthases and heart failure - lessons from genetically manipulated mice. J UOEH 2013; 35:147-158. [PMID: 23774658 DOI: 10.7888/juoeh.35.147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Nitric oxide (NO) is synthesized by three distinct NO synthase (NOS) isoforms (neuronal, inducible, and endothelial NOS), all of which are expressed in the human heart. The roles of NOSs in the pathogenesis of heart failure have been described in pharmacological studies with NOS inhibitors. Recently, genetically engineered animals have been used. We have generated mice in which all 3 NOS isoforms are completely disrupted (triple n/i/eNOS(-/-) mice). Morphological, echocardiographic, and hemodynamic analysis were performed in wild-type, singly nNOS(-/-), iNOS(-/-), eNOS(-/-), and triple n/i/eNOS(-/-) mice. Importantly, significant left ventricular (LV) hypertrophy and diastolic dysfunction was noted only in n/i/eNOS(-/-) mice, and those pathology was similar to diastolic heart failure in humans. Finally, treatment with an angiotensin II type 1 (AT1) receptor blocker, significantly prevented those abnormalities. These results provide the evidence that AT1 receptor pathway plays a center role in the pathogenesis of cardiac disorders in the n/i/eNOS(-/-) mice. Our studies with triple n/i/eNOS(-/-) mice provide pivotal insights into an understanding of the pathophysiology of NOSs in human heart failure.
Collapse
Affiliation(s)
- Kiyoko Shibata
- Department of Second Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan
| | | | | | | | | |
Collapse
|
25
|
Leiter EH, Schile A. Genetic and Pharmacologic Models for Type 1 Diabetes. CURRENT PROTOCOLS IN MOUSE BIOLOGY 2013; 3:9-19. [PMID: 24592352 PMCID: PMC3936677 DOI: 10.1002/9780470942390.mo120154] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes (T1D) is characterized by a partial or total insufficiency of insulin. The premiere animal model of autoimmune T cell-mediated T1D is the NOD mouse. A dominant negative mutation in the mouse insulin 2 gene (Ins2Akita ) produces a severe insulin deficiency syndrome without autoimmune involvement, as do a variety of transgenes overexpressed in beta cells. Pharmacologically-induced T1D (without autoimmunity) elicted by alloxan or streptozotocin at high doses can generate hyperglycemia in almost any strain of mouse by direct toxicity. Multiple low doses of streptozotocin combine direct beta cell toxicity with local inflammation to elicit T1D in a male sex-specific fashion. A summary of protocols relevant to the management of these different mouse models will be covered in this overview.
Collapse
Affiliation(s)
- Edward H. Leiter
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, Tel: 207-288-6370, FAX: 207-288-6077
| | - Andrew Schile
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, Tel: 207-288-6370, FAX: 207-288-6077
| |
Collapse
|
26
|
Padgett LE, Broniowska KA, Hansen PA, Corbett JA, Tse HM. The role of reactive oxygen species and proinflammatory cytokines in type 1 diabetes pathogenesis. Ann N Y Acad Sci 2013; 1281:16-35. [PMID: 23323860 PMCID: PMC3715103 DOI: 10.1111/j.1749-6632.2012.06826.x] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Type 1 diabetes (T1D) is a T cell–mediated autoimmune disease characterized by the destruction of insulin-secreting pancreatic β cells. In humans with T1D and in nonobese diabetic (NOD) mice (a murine model for human T1D), autoreactive T cells cause β-cell destruction, as transfer or deletion of these cells induces or prevents disease, respectively. CD4+ and CD8+ T cells use distinct effector mechanisms and act at different stages throughout T1D to fuel pancreatic β-cell destruction and disease pathogenesis. While these adaptive immune cells employ distinct mechanisms for β-cell destruction, one central means for enhancing their autoreactivity is by the secretion of proinflammatory cytokines, such as IFN-γ, TNF-α, and IL-1. In addition to their production by diabetogenic T cells, proinflammatory cytokines are induced by reactive oxygen species (ROS) via redox-dependent signaling pathways. Highly reactive molecules, proinflammatory cytokines are produced upon lymphocyte infiltration into pancreatic islets and induce disease pathogenicity by directly killing β cells, which characteristically possess low levels of antioxidant defense enzymes. In addition to β-cell destruction, proinflammatory cytokines are necessary for efficient adaptive immune maturation, and in the context of T1D they exacerbate autoimmunity by intensifying adaptive immune responses. The first half of this review discusses the mechanisms by which autoreactive T cells induce T1D pathogenesis and the importance of ROS for efficient adaptive immune activation, which, in the context of T1D, exacerbates autoimmunity. The second half provides a comprehensive and detailed analysis of (1) the mechanisms by which cytokines such as IL-1 and IFN-γ influence islet insulin secretion and apoptosis and (2) the key free radicals and transcription factors that control these processes.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
27
|
Muhammed SJ, Lundquist I, Salehi A. Pancreatic β-cell dysfunction, expression of iNOS and the effect of phosphodiesterase inhibitors in human pancreatic islets of type 2 diabetes. Diabetes Obes Metab 2012; 14:1010-9. [PMID: 22687049 DOI: 10.1111/j.1463-1326.2012.01632.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 03/16/2012] [Accepted: 06/04/2012] [Indexed: 11/30/2022]
Abstract
AIMS Induction of inducible nitric oxide synthase (iNOS) in pancreatic islets leads to exaggerated nitric oxide (NO) production associated with dysfunctional β-cells. We examined insulin secretion, iNOS expression and its relationship to the cAMP system in islets from human type 2 diabetes. METHODS Insulin, glucagon and cAMP were analysed by RIA; iNOS or phosphodiesterase (PDE) expression by quantitative PCR (qPCR), Western blot and confocal microscopy; cell viability by MTS. RESULTS Diabetic islets displayed impaired insulin and glucagon responses to glucose, disturbed cAMP generation and high inducible nitric oxide synthase (iNOS) mRNA and protein expression. Confocal microscopy showed iNOS protein expression in diabetic islets being confined to insulin, glucagon and somatostatin cells. Culture of diabetic islets at 5.5 mmol/l glucose with dibutyryl-cAMP (Bt(2) -cAMP) for 24 h was accompanied by marked suppression of iNOS mRNA, reduced nitrite production and increased insulin secretion. Diabetic islets displayed marked increase in PDE3A and PDE3B mRNA expression. Short-time incubation of diabetic islets showed, among the PDE inhibitors tested, cilostazol being most favourable to increase insulin secretion. Diabetic islets were most susceptible to long-term (72 h) culture at high glucose (20 mmol/l) reacting with increased apoptosis. Bt(2) -cAMP and the PDE inhibitors cilostazol, milrinone and IBMX efficiently increased cell viability at high glucose during culture. Defective glucose-stimulated insulin release upon induction of iNOS was restored by iNOS inhibitor aminoguanidine. CONCLUSION Our results suggest that in islets from type 2 diabetes, stimulatory effects in certain cAMP-compartments induced by PDE inhibitors might play a central role in the suppression of iNOS, resulting in increased β-cell viability and improved secretory response to glucose.
Collapse
Affiliation(s)
- S J Muhammed
- Department of Clinical Science, Division of Endocrine Pharmacology, SUS, Malmö, University of Lund, Malmö, Sweden.
| | | | | |
Collapse
|
28
|
Novotny GW, Lundh M, Backe MB, Christensen DP, Hansen JB, Dahllöf MS, Pallesen EMH, Mandrup-Poulsen T. Transcriptional and translational regulation of cytokine signaling in inflammatory β-cell dysfunction and apoptosis. Arch Biochem Biophys 2012; 528:171-84. [PMID: 23063755 DOI: 10.1016/j.abb.2012.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/20/2012] [Accepted: 09/22/2012] [Indexed: 12/19/2022]
Abstract
Disease is conventionally viewed as the chaotic inappropriate outcome of deranged tissue function resulting from aberrancies in cellular processes. Yet the patho-biology of cellular dysfunction and death encompasses a coordinated network no less sophisticated and regulated than maintenance of homeostatic balance. Cellular demise is far from passive subordination to stress but requires controlled coordination of energy-requiring activities including gene transcription and protein translation that determine the graded transition between defensive mechanisms, cell cycle regulation, dedifferentiation and ultimately to the activation of death programmes. In fact, most stressors stimulate both homeostasis and regeneration on one hand and impairment and destruction on the other, depending on the ambient circumstances. Here we illustrate this bimodal ambiguity in cell response by reviewing recent progress in our understanding of how the pancreatic β cell copes with inflammatory stress by changing gene transcription and protein translation by the differential and interconnected action of reactive oxygen and nitric oxide species, microRNAs and posttranslational protein modifications.
Collapse
Affiliation(s)
- Guy W Novotny
- Section of Endocrinological Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Gilbert ER, Liu D. Epigenetics: the missing link to understanding β-cell dysfunction in the pathogenesis of type 2 diabetes. Epigenetics 2012; 7:841-52. [PMID: 22810088 PMCID: PMC3427279 DOI: 10.4161/epi.21238] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes (T2D) is a growing health problem worldwide. While peripheral insulin resistance is common during obesity and aging in both animals and people, progression to T2D is largely due to insulin secretory dysfunction and significant apoptosis of functional β-cells, leading to an inability to compensate for insulin resistance. It is recognized that environmental factors and nutrition play an important role in the pathogenesis of diabetes. However, our knowledge surrounding molecular mechanisms by which these factors trigger β-cell dysfunction and diabetes is still limited. Recent discoveries raise the possibility that epigenetic changes in response to environmental stimuli may play an important role in the development of diabetes. In this paper, we review emerging knowledge regarding epigenetic mechanisms that may be involved in β-cell dysfunction and pathogenesis of diabetes, including the role of nutrition, oxidative stress and inflammation. We will mainly focus on the role of DNA methylation and histone modifications but will also briefly review data on miRNA effects on the pancreatic islets. Further studies aimed at better understanding how epigenetic regulation of gene expression controls β-cell function may reveal potential therapeutic targets for prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Elizabeth R. Gilbert
- Department of Animal and Poultry Sciences; College of Agriculture and Life Sciences; Virginia Tech; Blacksburg, VA USA
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise; College of Agriculture and Life Sciences; Virginia Tech; Blacksburg, VA USA
| |
Collapse
|
30
|
Abstract
Autoimmune Type 1 A Diabetes (T1D) is characterized by dependence on exogenous insulin consequential to the autoimmune attack and destruction of insulin-producing islet beta cells. Pancreatic islet cell inflammation, or insulitis, precedes beta cell death and T1D onset. In the insulitic lesion, innate immune cells produce chemokines and cytokines that recruit and activate adaptive immune cells (Eizirik D et al., Nat Rev Endocrinol 5:219-226, 2009). Locally produced cytokines not only increase immune surveillance of beta cells (Hanafusa T and Imagawa A, Ann NY Acad Sci 1150:297-299, 2008), but also cause beta cell dysfunction and decreased insulin secretion due to the generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) by the beta cells. This, coupled to the high levels of ROS and RNS secreted by activated macrophages and the low antioxidant capacities of beta cells (Huurman VA, PLoS One 3:e2435, 2008; Schatz D, Pediatr Diabetes 5:72-79, 2004; Verge CF, Diabetes 44:1176-1179, 1995), implicates free radicals as important effectors in T1D pathogenesis (Eizirik D et al., Nat Rev Endocrinol 5:219-226, 2009; Hanafusa T and Imagawa A, Ann NY Acad Sci 1150:297-299, 2008; Eisenbarth GS and Jeffrey J, Arq Bras Endocrinol Metabol 52:146-155, 2008; Pietropaolo M et al., Pediatr Diabetes 6:184-192, 2005).
Collapse
Affiliation(s)
- Yaíma L Lightfoot
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | | | | |
Collapse
|
31
|
Jimenez-Feltstrom J, Salehi A, Meidute Abaraviciene S, Henningsson R, Lundquist I. Abnormally decreased NO and augmented CO production in islets of the leptin-deficient ob/ob mouse might contribute to explain hyperinsulinemia and islet survival in leptin-resistant type 2 obese diabetes. ACTA ACUST UNITED AC 2011; 170:43-51. [DOI: 10.1016/j.regpep.2011.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 03/24/2011] [Accepted: 04/16/2011] [Indexed: 01/14/2023]
|
32
|
Kishi S, Abe H, Akiyama H, Tominaga T, Murakami T, Mima A, Nagai K, Kishi F, Matsuura M, Matsubara T, Iehara N, Ueda O, Fukushima N, Jishage KI, Doi T. SOX9 protein induces a chondrogenic phenotype of mesangial cells and contributes to advanced diabetic nephropathy. J Biol Chem 2011; 286:32162-9. [PMID: 21795715 DOI: 10.1074/jbc.m111.244541] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diabetic nephropathy (DN) is the most important chronic kidney disease. We previously reported that Smad1 transcriptionally regulates the expression of extracellular matrix in DN. Phenotypic change in mesangial cells (MCs) is a key pathologic event in the progression of DN. The aim of this study is to investigate a novel mechanism underlying chondrogenic phenotypic change in MCs that results in the development of DN. MCs showed chondrogenic potential in a micromass culture, and BMP4 induced the expression of chondrocyte markers (SRY-related HMG Box 9 (SOX9) and type II collagen (COL2)). Advanced glycation end products induced the expression of chondrocyte marker proteins downstream from the BMP4-Smad1 signaling pathway in MCs. In addition, hypoxia also induced the expression of BMP4, hypoxia-inducible factor-1α (HIF-1α), and chondrocyte markers. Overexpression of SOX9 caused ectopic expression of proteoglycans and COL2 in MCs. Furthermore, forced expression of Smad1 induced chondrocyte markers as well. Dorsomorphin inhibited these inductions. Glomerular expressions of HIF-1α, BMP4, and chondrocyte markers were observed in diabetic nephropathy mice. These positive stainings were observed in mesangial sclerotic lesions. SOX9 was partially colocalized with HIF-1α and BMP4 in diabetic glomeruli. BMP4 knock-in transgenic mice showed not only similar pathological lesions to DN, but also the induction of chondrocyte markers in the sclerotic lesions. Here we demonstrate that HIF-1α and BMP4 induce SOX9 expression and subsequent chondrogenic phenotype change in DN. The results suggested that the transdifferentiation of MCs into chondrocyte-like cells in chronic hypoxic stress may result in irreversible structural change in DN.
Collapse
Affiliation(s)
- Seiji Kishi
- Department of Nephrology, Graduate School of Medicine, Health-Bioscience Institute, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tanioka T, Tamura Y, Fukaya M, Shinozaki S, Mao J, Kim M, Shimizu N, Kitamura T, Kaneki M. Inducible nitric-oxide synthase and nitric oxide donor decrease insulin receptor substrate-2 protein expression by promoting proteasome-dependent degradation in pancreatic beta-cells: involvement of glycogen synthase kinase-3beta. J Biol Chem 2011; 286:29388-29396. [PMID: 21700708 DOI: 10.1074/jbc.m110.192732] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Insulin receptor substrate-2 (IRS-2) plays a critical role in the survival and function of pancreatic β-cells. Gene disruption of IRS-2 results in failure of the β-cell compensatory mechanism and diabetes. Nonetheless, the regulation of IRS-2 protein expression in β-cells remains largely unknown. Inducible nitric-oxide synthase (iNOS), a major mediator of inflammation, has been implicated in β-cell damage in type 1 and type 2 diabetes. The effects of iNOS on IRS-2 expression have not yet been investigated in β-cells. Here, we show that iNOS and NO donor decreased IRS-2 protein expression in INS-1/832 insulinoma cells and mouse islets, whereas IRS-2 mRNA levels were not altered. Interleukin-1β (IL-1β), alone or in combination with interferon-γ (IFN-γ), reduced IRS-2 protein expression in an iNOS-dependent manner without altering IRS-2 mRNA levels. Proteasome inhibitors, MG132 and lactacystin, blocked the NO donor-induced reduction in IRS-2 protein expression. Treatment with NO donor led to activation of glycogen synthase kinase-3β (GSK-3β) and c-Jun N-terminal kinase (JNK/SAPK) in β-cells. Inhibition of GSK-3β by pharmacological inhibitors or siRNA-mediated knockdown significantly prevented NO donor-induced reduction in IRS-2 expression in β-cells. In contrast, a JNK inhibitor, SP600125, did not effectively block reduced IRS-2 expression in NO donor-treated β-cells. These data indicate that iNOS-derived NO reduces IRS-2 expression by promoting protein degradation, at least in part, through a GSK-3β-dependent mechanism. Our findings suggest that iNOS-mediated decreased IRS-2 expression may contribute to the progression and/or exacerbation of β-cell failure in diabetes.
Collapse
Affiliation(s)
- Toshihiro Tanioka
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Yoshiaki Tamura
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Makiko Fukaya
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Shohei Shinozaki
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Ji Mao
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Minhye Kim
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Nobuyuki Shimizu
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| | - Masao Kaneki
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, Massachusetts 02129 and.
| |
Collapse
|
34
|
Liu GY, Zhang YX, Chai TY. Phytochelatin synthase of Thlaspi caerulescens enhanced tolerance and accumulation of heavy metals when expressed in yeast and tobacco. PLANT CELL REPORTS 2011; 30:1067-76. [PMID: 21327392 DOI: 10.1007/s00299-011-1013-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 01/01/2011] [Accepted: 01/11/2011] [Indexed: 05/22/2023]
Abstract
Phytochelatin synthase (PCS) is key enzyme for heavy metal detoxification and accumulation in plant. In this study, we isolated the PCS gene TcPCS1 from the hyperaccumulator Thlaspi caerulescens. Overexpression of TcPCS1 enhanced PC production in tobacco. Cd accumulation in the roots and shoots of TcPCS1 transgenic seedlings was increased compared to the wild type (WT), while Cd translocation from roots to shoots was not affected under Cd treatment. The root length of the TcPCS1 transgenic tobacco seedlings was significantly longer than that of the WT under Cd stress. These data indicate that TcPCS1 expression might increase Cd accumulation and tolerance in transgenic tobacco. In addition, the malondialdehyde content in TcPCS1 plants was below that of the wild type. However, the antioxidant enzyme activities of superoxide dismutase, peroxidase and catalase were found to be significantly higher than those of the WT when the transgenic plant was exposed to Cd stress. This suggests that the increase in PC production might enhance the Cd accumulation and thus increase the oxidative stress induced by the cadmium. The production of PCs could cause a transient decrease in the cytosolic glutathione (GSH) pool, and Cd and lower GSH concentration caused an increase in the oxidative response. We also determined TcPCS1 in Thlaspi caerulescens was regulated after exposure to various concentrations of CdCl(2) over different treatment times. Expression of TcPCS1 leading to increased Cd accumulation and enhanced metal tolerance, but the Cd contents were restrained by adding zinc in Saccharomyces cerevisiae transformants.
Collapse
MESH Headings
- Adaptation, Physiological/drug effects
- Adaptation, Physiological/genetics
- Aminoacyltransferases/genetics
- Aminoacyltransferases/metabolism
- Cadmium/metabolism
- Cadmium/toxicity
- Gene Expression Profiling
- Gene Expression Regulation, Plant/drug effects
- Glutathione/metabolism
- Malondialdehyde/metabolism
- Metals, Heavy/metabolism
- Metals, Heavy/toxicity
- Organ Specificity/drug effects
- Organ Specificity/genetics
- Phytochelatins/metabolism
- Plant Roots/anatomy & histology
- Plant Roots/drug effects
- Plant Roots/metabolism
- Plant Shoots/drug effects
- Plant Shoots/metabolism
- Plants, Genetically Modified
- RNA, Plant/genetics
- RNA, Plant/metabolism
- Reactive Oxygen Species/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Saccharomyces cerevisiae/metabolism
- Stress, Physiological/drug effects
- Stress, Physiological/genetics
- Thlaspi/drug effects
- Thlaspi/enzymology
- Thlaspi/genetics
- Nicotiana/drug effects
- Nicotiana/genetics
- Nicotiana/metabolism
- Transformation, Genetic/drug effects
Collapse
Affiliation(s)
- Ge-Yu Liu
- College of Life Science, Graduate University of Chinese Academy of Sciences, Yuquan Rd 19, Beijing 100049, China
| | | | | |
Collapse
|
35
|
Epigallocatechin gallate delays the onset of type 1 diabetes in spontaneous non-obese diabetic mice. Br J Nutr 2010; 105:1218-25. [PMID: 21144096 DOI: 10.1017/s0007114510004824] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Type 1 diabetes (T1D) results from the autoimmune-mediated destruction of pancreatic β-cells, leading to deficiency of insulin production. Successful islet transplantation can normalise hyperglycaemia in T1D patients; however, the limited availability of the islets, loss of islet cell mass through apoptosis after islet isolation and potential autoimmune destruction of the transplanted islets prevent the widespread use of this procedure. Therefore, the search for novel and cost-effective agents that can prevent or treat T1D is extremely important to decrease the burden of morbidity from this disease. In the present study, we discovered that ( - )-epigallocatechin gallate (EGCG, 0·05 % in drinking-water), the primary polyphenolic component in green tea, effectively delayed the onset of T1D in non-obese diabetic (NOD) mice. At 32 weeks of age, eight (66·7 %) out of twelve mice in the control group developed diabetes, whereas only three (25 %) out of twelve mice in the EGCG-treated group became diabetic (P < 0·05). Consistently, mice supplemented with EGCG had significantly higher plasma insulin levels and survival rate but lower glycosylated Hb concentrations compared with the control animals. EGCG had no significant effects on food or water intake and body weight in mice, suggesting that the glucose-lowering effect was not due to an alteration in these parameters. While EGCG did not modulate insulitis, it elevated the circulating anti-inflammatory cytokine IL-10 level in NOD mice. These findings demonstrate that EGCG may be a novel, plant-derived compound capable of reducing the risk of T1D.
Collapse
|
36
|
Tsutsui M, Shimokawa H, Otsuji Y, Yanagihara N. Pathophysiological relevance of NO signaling in the cardiovascular system: Novel insight from mice lacking all NO synthases. Pharmacol Ther 2010; 128:499-508. [DOI: 10.1016/j.pharmthera.2010.08.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Araoka T, Abe H, Tominaga T, Mima A, Matsubara T, Murakami T, Kishi S, Nagai K, Doi T. Transcription factor 7-like 2 (TCF7L2) regulates activin receptor-like kinase 1 (ALK1)/Smad1 pathway for development of diabetic nephropathy. Mol Cells 2010; 30:209-18. [PMID: 20803090 DOI: 10.1007/s10059-010-0109-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 05/17/2010] [Accepted: 05/28/2010] [Indexed: 11/29/2022] Open
Abstract
Smad1 has previously been shown to play a key role in the development of diabetic nephropathy (DN), by increasing synthesis of extracellular matrix. However, the regulatory mechanism of Smad1 in DN is still unclear. This study aims to elucidate molecular interactions between activin receptor-like kinase 1 (ALK1)/Smad1 signaling pathway and transcription factor 7-like 2 (TCF7L2) in the progression of DN in vitro and in vivo. The expressions of TCF7L2 and ALK1 were induced by advanced glycation end products (AGEs) in parallel with Smad1, phosphorylated Smad1 (pSmad1), and alpha-smooth muscle actin (α-SMA) through TGF-β1 in cultured mesangial cells. Constitutively active ALK1 increased pSmad1 and α-SMA expressions. The binding of TCF7L2 to ALK1 promoter was confirmed by chromatin immunoprecipitation assay. Furthermore, TCF7L2 induced promoter activity of ALK1. AGEs and TGF-β1 induced a marked increase in TCF7L2 expression in parallel with ALK1. Overexpression of TCF7L2 increased the expressions of ALK1 and Smad1. Inversely, TCF7L2 knockdown by siRNA suppressed α-SMA expression as well as ALK1 and Smad1. The iNOS transgenic mice (iNOS-Tgm), which developed diabetic glomerulosclerosis resembling human diabetic nephropathy, exhibited markedly increased expressions of ALK1, TCF7L2, Smad1, pSmad1, and α-SMA in glomeruli in association with mesangial matrix expansion. These results provide a new evidence that the TCF7L2/ALK1/Smad1 pathway plays a key role in the development of DN.
Collapse
Affiliation(s)
- Toshikazu Araoka
- Deapartment of Nephrology, Graduate School of Medicine, Institute of Health-Bio-Science, University of Tokushima, Tokushima, 770-8503, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Shimokawa H, Tsutsui M. Nitric oxide synthases in the pathogenesis of cardiovascular disease: lessons from genetically modified mice. Pflugers Arch 2010; 459:959-67. [PMID: 20179961 DOI: 10.1007/s00424-010-0796-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 01/27/2010] [Accepted: 01/28/2010] [Indexed: 02/06/2023]
Abstract
Nitric oxide (NO) is produced in almost all tissues and organs, exerting a variety of biological actions under both physiological and pathological conditions. NO is synthesized by three distinct NO synthase (NOS) isoforms (neuronal, inducible, and endothelial NOS), all of which are expressed in the human cardiovascular system. Although the regulatory roles of NOSs in cardiovascular diseases have been described in pharmacological studies with selective and non-selective NOS inhibitors, the specificity of the NOS inhibitors continues to be an issue of debate. To overcome this issue, genetically engineered animals have been used. All types of NOS gene-deficient animals, including singly, doubly, and triply NOS-deficient mice, and various types of NOS gene-transgenic (TG) animals, including conditional and non-conditional TG mice bearing endothelium-specific or cardiomyocyte-specific overexpression of each NOS gene, have thus been developed. The roles of individual NOS isoforms as well as the entire NOS system in the cardiovascular system have been extensively investigated in those mice, providing pivotal insights into an understanding of the pathophysiology of NOSs in human cardiovascular diseases. Based on studies with the murine NOS genetic models, this review briefly summarizes the latest knowledge of NOSs and cardiovascular diseases.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | | |
Collapse
|
39
|
Mi QS, Yan SL, Wang ZZ, Ding KH, Li C, Wang L, Zhou L, Yamamoto Y, Yamamoto H, Okamoto H, Isales C. Spontaneous bone loss in RIP-iNOS transgenic mouse: a mouse model for diabetes-mediated osteopenia/osteoporosis. Cell Cycle 2009; 8:4179-81. [PMID: 19946218 DOI: 10.4161/cc.8.24.10152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
40
|
Okamoto H, Takasawa S. Recent advances in physiological and pathological significance of NAD+ metabolites: roles of poly(ADP-ribose) and cyclic ADP-ribose in insulin secretion and diabetogenesis. Nutr Res Rev 2009; 16:253-66. [PMID: 19087393 DOI: 10.1079/nrr200362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Poly(ADP-ribose) synthetase/polymerase (PARP) activation causes NAD+ depletion in pancreatic beta-cells, which results in necrotic cell death. On the other hand, ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase (CD38) synthesizes cyclic ADP-ribose from NAD+, which acts as a second messenger, mobilizing intracellular Ca2+ for insulin secretion in response to glucose in beta-cells. PARP also acts as a regenerating gene (Reg) transcription factor to induce beta-cell regeneration. This provides the new concept that NAD+ metabolism can control the cellular function through gene expression. Clinically, PARP could be one of the most important therapeutic targets; PARP inhibitors prevent cell death, maintain the formation of a second messenger, cyclic ADP-ribose, to achieve cell function, and keep PARP functional as a transcription factor for cell regeneration.
Collapse
Affiliation(s)
- Hiroshi Okamoto
- Department of Biochemistry and Advanced Biological Sciences for Regeneration (Kotobiken Medical Laboratories) Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| | | |
Collapse
|
41
|
Role of nitric oxide in the pathogenesis of encephalomyocarditis virus-induced diabetes in mice. J Virol 2009; 83:8004-11. [PMID: 19535454 DOI: 10.1128/jvi.00205-09] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The D variant of encephalomyocarditis virus (EMC-D virus) causes diabetes in mice by destroying pancreatic beta cells. In mice infected with a low dose of EMC-D virus, macrophages play an important role in beta-cell destruction by producing soluble mediators such as interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNF-alpha), and nitric oxide (NO). To investigate the role of NO and inducible NO synthase (iNOS) in the development of diabetes in EMC-D virus-infected mice, we infected iNOS-deficient DBA/2 mice with EMC-D virus (2 x 10(2) PFU/mouse). Mean blood glucose levels in EMC-D virus-infected iNOS-deficient mice and wild-type mice were 205.5 and 466.7 mg/dl, respectively. Insulitis and macrophage infiltration were reduced in islets of iNOS-deficient mice compared with wild-type mice at 3 days after EMC-D virus infection. Apoptosis of beta cells was decreased in iNOS-deficient mice, as evidenced by reduced numbers of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cells. There were no differences in mRNA expression of antiapoptotic molecules Bcl-2, Bcl-xL, Bcl-w, Mcl-1, cIAP-1, and cIAP-2 between wild-type and iNOS-deficient mice, whereas expression of proapoptotic Bax and Bak mRNAs was significantly decreased in iNOS-deficient mice. Expression of IL-1beta and TNF-alpha mRNAs was significantly decreased in both islets and macrophages of iNOS-deficient mice compared with wild-type mice after EMC-D virus infection. Nuclear factor kappaB was less activated in macrophages of iNOS-deficient mice after virus infection. We conclude that NO plays an important role in the activation of macrophages and apoptosis of pancreatic beta cells in EMC-D virus-infected mice and that deficient iNOS gene expression inhibits macrophage activation and beta-cell apoptosis, contributing to prevention of EMC-D virus-induced diabetes.
Collapse
|
42
|
Tsutsui M, Shimokawa H, Otsuji Y, Ueta Y, Sasaguri Y, Yanagihara N. Nitric oxide synthases and cardiovascular diseases: insights from genetically modified mice. Circ J 2009; 73:986-93. [PMID: 19430166 DOI: 10.1253/circj.cj-09-0208] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide (NO) is produced in almost all tissues and organs, exerting a variety of biological actions under both physiological and pathological conditions. NO is synthesized by 3 distinct NO synthase (NOS) isoforms (neuronal, inducible, and endothelial NOS), all of which are expressed in the human cardiovascular system. The regulatory roles of NOSs in cardiovascular diseases have been described in pharmacological studies with selective and non-selective NOS inhibitors. However, the specificity of the NOS inhibitors continues to be an issue of debate. To overcome this issue, genetically engineered animals have been used. All types of NOS gene-deficient (knockout: KO) animals, including singly, doubly, and triply NOS-KO mice, and various types of NOS gene-transgenic (TG) animals, including conditional and non-conditional TG mice bearing endothelium-specific or cardiomyocyte-specific overexpression of each NOS gene, have thus far been developed. The roles of individual NOS isoforms, as well as the entire NOS system, in the cardiovascular system have been extensively investigated in those mice, and the results provide pivotal insights into the pathophysiology of NOSs in human cardiovascular diseases. Based on studies with murine NOS genetic models, this review summarizes the latest knowledge of NOSs and cardiovascular diseases.
Collapse
Affiliation(s)
- Masato Tsutsui
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Yasuda H, Jin Z, Nakayama M, Yamada K, Kishi M, Okumachi Y, Arai T, Moriyama H, Yokono K, Nagata M. NO-mediated cytotoxicity contributes to multiple low-dose streptozotocin-induced diabetes but not to NOD diabetes. Diabetes Res Clin Pract 2009; 83:200-7. [PMID: 19117633 DOI: 10.1016/j.diabres.2008.11.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 11/01/2008] [Accepted: 11/10/2008] [Indexed: 11/27/2022]
Abstract
Type 1 diabetes (T1D) is caused mostly by autoimmune destruction of pancreatic beta-cells, the precise mechanism of which remains unclear. Two major effector mechanisms have been proposed: direct cell-mediated and indirect cytokine-mediated cytotoxicity. Cytokine-mediated beta-cell destruction is presumed mainly caused by NO production. To evaluate the role of iNOS expression in T1D, this study used a novel iNOS inhibitor ONO-1714. ONO-1714 significantly reduced cytokine-mediated cytotoxicity and NO production in both MIN6N9a cells and C57BL/6 islets in the presence of IL-1beta, TNF-alpha, and IFN-gamma. To evaluate whether NO contributes to diabetes progression in vivo, ONO-1714 was administered to four different mouse models of autoimmune diabetes: multiple low-dose STZ (MLDS)-induced C57BL/6, CY-induced, adoptive transfer and spontaneous NOD diabetes. Exposure to STZ in vitro induced NO production in MIN6N9a cells and C57BL/6 islets, and in vivo injection of ONO-1714 to MLDS-treated mice significantly reduced hyperglycemia and interestingly, led to complete suppression of cellular infiltration of pancreatic islets. In contrast, when ONO-1714 was injected into spontaneous NOD mice and CY-induced and adoptive transfer models of NOD diabetes, overt diabetes could not be inhibited in these models. These findings suggest that NO-mediated cytotoxicity significantly contributes to MLDS-induced diabetes but not to NOD diabetes.
Collapse
Affiliation(s)
- Hisafumi Yasuda
- Department of Internal and Geriatric Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Van Dyke K, Ghareeb E, Van Dyke M, Sosa A, Hoeldtke RD, Van Thiel DH. Luminescence experiments involved in the mechanism of streptozotocin diabetes and cataract formation. LUMINESCENCE 2009; 23:386-91. [PMID: 18651583 DOI: 10.1002/bio.1050] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Streptozotocin (STZ)-induced diabetes is linked to excessive nitric oxide (NO), and possibly peroxynitrite (OONO(-)) and/or other nitrogen oxides, e.g. nitrogen trioxide (N(2)O(3)), which damages DNA of pancreatic beta cells, causing death and loss of insulin. Simultaneous injection of carboxy-PTIO (CPTIO) and STZ prevents diabetes and cataract formation in rats, whereas 4-hydroxy-Tempo (4HT) does not. CPTIO oxidizes nitric oxide to nitrite, which prevents production of the diabetogenic toxin. Peroxynitrite may not be involved, since 4HT (converts O(2)(-) to H(2)O(2)) injected with STZ produces diabetes. All six of the control rats injected with STZ became diabetic and developed cataracts after 3 months. Eight rats injected with STZ and CPTIO were non-diabetic with no cataracts up to a year. This work establishes the idea that excessive nitric oxide is a primary initiator in STZ diabetes. Luminescence experiments using OONO(-) generation from SIN-1 with L-012 indicates that 4HT is an effective inhibitor, while CPTIO is ineffective. Experiments with dilute solutions of nitrogen trioxide added to ladder or plasmid DNA reveal extensive nicking of DNA, thereby raising the possibility that other oxides of nitrogen could be involved with the damage to DNA. It can be concluded that diabetes can be prevented by oxidizing excessive NO from STZ.
Collapse
Affiliation(s)
- Knox Van Dyke
- Department of Biochemistry and Molecular Pharmacology, Robert C. Byrd Medical Center, West Virginia University, Morgantown, WV 26506, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Kato I, Oya T, Suzuki H, Takasawa K, Ichsan AM, Nakada S, Ishii Y, Shimada Y, Sasahara M, Tobe K, Takasawa S, Okamoto H, Hiraga K. A novel model of insulin-dependent diabetes with renal and retinal lesions by transgenic expression of CaMKIIalpha (Thr286Asp) in pancreatic beta-cells. Diabetes Metab Res Rev 2008; 24:486-97. [PMID: 18551708 DOI: 10.1002/dmrr.864] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND The activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) in pancreatic beta-cells has been thought to play a central role in Ca2+-mediated insulin secretion. However, the physiological and pathological significance of CaMKII activation in pancreatic beta-cells has never been investigated in vivo. METHODS We generated transgenic (TG) mice overexpressing the constitutively active-type CaMKIIalpha (Thr286Asp) in beta-cells. The mice were extensively examined histologically and biochemically. Time-course changes of blood glucose, haemoglobin A1C and insulin were also determined. RESULTS Western blot and immunohistochemical analyses showed overexpression of CaMKIIalpha proteins in pancreatic beta-cells of TG mice. All TG mice developed severe hypoinsulinaemic diabetes by P28. In vivo BrdU labelling analysis revealed that cell proliferation in TG islets is severely impaired. Immunohistochemical examination revealed accumulations of NF-kappaB in nuclei of TG beta-cells at P21, which are associated with DNA laddering, a hallmark of apoptosis. At P28, pancreatic and serum insulin levels were both significantly (p < 0.05) lower in TG mice (0.037 +/- 0.005 ng/microg and 0.50 +/- 0.01 ng/mL) than in wild-type mice (0.997 +/- 0.093 ng/microg and 2.50 +/- 0.22 ng/mL). TG mice at P140 showed enlargement of kidney, mesangial expansion and glomerulosclerosis, which are associated with urinary albumin excretion. TG mice at P140-P168 developed severe retinal lesions such as disrupted ganglion cells and showed a flat pattern in electroretinography. CONCLUSIONS The TG mice established herein will be valuable as a novel model of severe insulin-dependent diabetes accompanied by an early progression of diabetic micro-vascular complications.
Collapse
Affiliation(s)
- Ichiro Kato
- Department of Biochemistry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Impaired glucose-stimulated insulin secretion in the GK rat is associated with abnormalities in islet nitric oxide production. ACTA ACUST UNITED AC 2008; 151:139-46. [PMID: 18662725 DOI: 10.1016/j.regpep.2008.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2008] [Revised: 05/23/2008] [Accepted: 07/02/2008] [Indexed: 11/24/2022]
Abstract
We investigated implications of nitric oxide (NO) derived from islet neuronal constitutive NO synthase (ncNOS) and inducible NOS (iNOS) on insulin secretory mechanisms in the mildly diabetic GK rat. Islets from GK rats and Wistar controls were analysed for ncNOS and iNOS by HPLC, immunoblotting and immunocytochemistry in relation to insulin secretion stimulated by glucose or l-arginine in vitro and in vivo. No obvious difference in ncNOS fluorescence in GK vs control islets was seen but freshly isolated GK islets displayed a marked iNOS expression and activity. After incubation at low glucose GK islets showed an abnormal increase in both iNOS and ncNOS activities. At high glucose the impaired glucose-stimulated insulin release was associated with an increased iNOS expression and activity and NOS inhibition dose-dependently amplified insulin secretion in both GK and control islets. This effect by NOS inhibition was also evident in depolarized islets at low glucose, where forskolin had a further amplifying effect in GK but not in control islets. NOS inhibition increased basal insulin release in perfused GK pancreata and amplified insulin release after glucose stimulation in both GK and control pancreata, almost abrogating the nadir separating first and second phase in controls. A defective insulin response to l-arginine was seen in GK rats in vitro and in vivo, being partially restored by NOS inhibition. The results suggest that increased islet NOS activities might contribute to the defective insulin response to glucose and l-arginine in the GK rat. Excessive iNOS expression and activity might be deleterious for the beta-cells over time.
Collapse
|
47
|
Salehi A, Meidute Abaraviciene S, Jimenez-Feltstrom J, Ostenson CG, Efendic S, Lundquist I. Excessive islet NO generation in type 2 diabetic GK rats coincides with abnormal hormone secretion and is counteracted by GLP-1. PLoS One 2008; 3:e2165. [PMID: 18478125 PMCID: PMC2367446 DOI: 10.1371/journal.pone.0002165] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Accepted: 03/25/2008] [Indexed: 11/22/2022] Open
Abstract
Background A distinctive feature of type 2 diabetes is inability of insulin-secreting β-cells to properly respond to elevated glucose eventually leading to β-cell failure. We have hypothesized that an abnormally increased NO production in the pancreatic islets might be an important factor in the pathogenesis of β-cell dysfunction. Principal Findings We show now that islets of type 2 spontaneous diabetes in GK rats display excessive NO generation associated with abnormal iNOS expression in insulin and glucagon cells, increased ncNOS activity, impaired glucose-stimulated insulin release, glucagon hypersecretion, and impaired glucose-induced glucagon suppression. Pharmacological blockade of islet NO production by the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) greatly improved hormone secretion from GK islets suggesting islet NOS activity being an important target to inactivate for amelioration of islet cell function. The incretin hormone GLP-1, which is used in clinical practice suppressed iNOS and ncNOS expression and activity with almost full restoration of insulin release and partial restoration of glucagon release. GLP-1 suppression of iNOS expression was reversed by PKA inhibition but unaffected by the proteasome inhibitor MG132. Injection of glucose plus GLP-1 in the diabetic rats showed that GLP-1 amplified the insulin response but induced a transient increase and then a poor depression of glucagon. Conclusion The results suggest that abnormally increased NO production within islet cells is a significant player in the pathogenesis of type 2 diabetes being counteracted by GLP-1 through PKA-dependent, nonproteasomal mechanisms.
Collapse
Affiliation(s)
- Albert Salehi
- Department of Clinical Science, Universitetssjukhuset Malmö Allmäna Sjukhus, Division of Endocrine Pharmacology, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
48
|
Palmitate-induced beta-cell dysfunction is associated with excessive NO production and is reversed by thiazolidinedione-mediated inhibition of GPR40 transduction mechanisms. PLoS One 2008; 3:e2182. [PMID: 18478115 PMCID: PMC2366067 DOI: 10.1371/journal.pone.0002182] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Accepted: 03/13/2008] [Indexed: 11/19/2022] Open
Abstract
Background Type 2 diabetes often displays hyperlipidemia. We examined palmitate effects on pancreatic islet function in relation to FFA receptor GPR40, NO generation, insulin release, and the PPARγ agonistic thiazolidinedione, rosiglitazone. Principal Findings Rosiglitazone suppressed acute palmitate-stimulated GPR40-transduced PI hydrolysis in HEK293 cells and insulin release from MIN6c cells and mouse islets. Culturing islets 24 h with palmitate at 5 mmol/l glucose induced β-cell iNOS expression as revealed by confocal microscopy and increased the activities of ncNOS and iNOS associated with suppression of glucose-stimulated insulin response. Rosiglitazone reversed these effects. The expression of iNOS after high-glucose culturing was unaffected by rosiglitazone. Downregulation of GPR40 by antisense treatment abrogated GPR40 expression and suppressed palmitate-induced iNOS activity and insulin release. Conclusion We conclude that, in addition to mediating acute FFA-stimulated insulin release, GPR40 is an important regulator of iNOS expression and dysfunctional insulin release during long-term exposure to FFA. The adverse effects of palmitate were counteracted by rosiglitazone at GPR40, suggesting that thiazolidinediones are beneficial for β-cell function in hyperlipidemic type 2 diabetes.
Collapse
|
49
|
Stosic-Grujicic S, Stojanovic I, Maksimovic-Ivanic D, Momcilovic M, Popadic D, Harhaji L, Miljkovic D, Metz C, Mangano K, Papaccio G, Al-Abed Y, Nicoletti F. Macrophage migration inhibitory factor (MIF) is necessary for progression of autoimmune diabetes mellitus. J Cell Physiol 2008; 215:665-75. [PMID: 18064633 DOI: 10.1002/jcp.21346] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine of the innate immune system that plays a major role in the induction of immunoinflammatory responses. To examine the role of endogenous MIF in the pathogenesis of type 1 diabetes (TID) we evaluated the effects of administration of neutralizing anti-MIF antibodies to NOD mice with accelerated forms of diabetes induced by injection of cyclophosphamide or by transfer of diabetogenic spleen cells. Both accelerated forms of diabetes were markedly reduced by anti-MIF antibody. Furthermore, MIF-deficient (MIF(-/-)) mice were less susceptible to the induction of immunoinflammatory diabetes, insulitis and apoptosis within the endocrine pancreas by multiple low doses of streptozotocin (MLD-STZ) than genetically matched wild type (WT) mice. MIF deficiency resulted in lower proliferation and lymphocyte adhesion, as well as reduced production from the spleens and peritoneal cells of a variety of inflammatory mediators typically associated with development of the disease including IL-12, IL-23, TNF-alpha, and IL-1beta. Furthermore, MIF deletion affected the production of IL-18, TNF-alpha, IL-1beta, and iNOS in the islets of Langerhans. These data, along with the higher expression of IL-4 and TGF-beta observed in the periphery and in the pancreas of MLD-STZ-challenged MIF(-/-) mice as compared to WT controls suggest that MIF deficiency has induced an immune deviation towards protective type 2/3 response. These results suggest that MIF participates in T1D by controlling the functional activity of monocytes/macrophages and T cells and modulating their secretory capacity of pro- and anti-inflammatory molecules.
Collapse
|
50
|
Wasniewska M, Arrigo T, Crisafulli G, Aversa T, Messina MF, Salzano G, De Luca F. In the Italian population sexual dimorphism affects pre-natal thyroid migration but not biochemical severity of gland ectopia and pre-natal bone maturation. J Endocrinol Invest 2008; 31:341-5. [PMID: 18475053 DOI: 10.1007/bf03346368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
UNLABELLED The aim of the present study was to retrospectively re-evaluate a population of selected infants with congenital hypothyroidism (CH), in order to investigate whether sexual dimorphism affects: a) CH etiology; b) its biochemical severity at the time of screening and recall; c) patients' biochemical response to replacement treatment during the 1st yr of life; d) their bone maturation (BM) at birth; e) their psychomotor status at 1 yr. This retrospective study covers 192 infants (116 females) with persistent CH who were selected from a larger population of CH patients identified during a 10-yr period (1990-1999) by the screening programs of 5 northern, central, and southern regions of Italy. Thirty boys (39.5%) and 66 girls (56.9%) were found to have ectopia, whereas the remaining 46 boys and 50 girls exhibited the other causes of CH. When compared with the prevalence of the remaining causes that of ectopia was significantly higher in girls than in boys (66/116 vs 30/76; chi2=5.57, p<0.025), and sex ratio in ectopia was significantly different also compared with the orthotopic gland group only (66/84 vs 30/51; chi2=6.02, p<0.025). No differences between males and females were detected in the groups with either athyreosis or orthotopic gland. In no groups were there differences between sexes for gestational age, birth auxological data, percentage of newborns with bone retardation or developmental quotient at 1 yr. Thyroid tests at birth, age at TSH normalization and average thyroid tests under L-T4 treatment during the 1st yr did not differ between sexes in any of the groups. CONCLUSIONS a) in the Italian population sexual dimorphism affects pre-natal thyroid migration but neither biochemical severity of ectopia, nor pre-natal bone maturation and psychomotor development; b) girls with CH do not require higher doses of initial therapy in order to achieve TSH normalization; c) future developmental and molecular studies on ectopia etiology in CH need to take into account the effect of sexual dimorphism.
Collapse
Affiliation(s)
- M Wasniewska
- Department of Pediatrics, University of Messina, 98123 Messina, Italy.
| | | | | | | | | | | | | |
Collapse
|