1
|
Dakroub A, Dbouk A, Asfour A, Nasser SA, El-Yazbi AF, Sahebkar A, Eid AA, Iratni R, Eid AH. C-peptide in diabetes: A player in a dual hormone disorder? J Cell Physiol 2024; 239:e31212. [PMID: 38308646 DOI: 10.1002/jcp.31212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
C-peptide, a byproduct of insulin synthesis believed to be biologically inert, is emerging as a multifunctional molecule. C-peptide serves an anti-inflammatory and anti-atherogenic role in type 1 diabetes mellitus (T1DM) and early T2DM. C-peptide protects endothelial cells by activating AMP-activated protein kinase α, thus suppressing the activity of NAD(P)H oxidase activity and reducing reactive oxygen species (ROS) generation. It also prevents apoptosis by regulating hyperglycemia-induced p53 upregulation and mitochondrial adaptor p66shc overactivation, as well as reducing caspase-3 activity and promoting expression of B-cell lymphoma-2. Additionally, C-peptide suppresses platelet-derived growth factor (PDGF)-beta receptor and p44/p42 mitogen-activated protein (MAP) kinase phosphorylation to inhibit vascular smooth muscle cells (VSMC) proliferation. It also diminishes leukocyte adhesion by virtue of its capacity to abolish nuclear factor kappa B (NF-kB) signaling, a major pro-inflammatory cascade. Consequently, it is envisaged that supplementation of C-peptide in T1DM might ameliorate or even prevent end-organ damage. In marked contrast, C-peptide increases monocyte recruitment and migration through phosphoinositide 3-kinase (PI-3 kinase)-mediated pathways, induces lipid accumulation via peroxisome proliferator-activated receptor γ upregulation, and stimulates VSMC proliferation and CD4+ lymphocyte migration through Src-kinase and PI-3K dependent pathways. Thus, it promotes atherosclerosis and microvascular damage in late T2DM. Indeed, C-peptide is now contemplated as a potential biomarker for insulin resistance in T2DM and linked to increased coronary artery disease risk. This shift in the understanding of the pathophysiology of diabetes from being a single hormone deficiency to a dual hormone disorder warrants a careful consideration of the role of C-peptide as a unique molecule with promising diagnostic, prognostic, and therapeutic applications.
Collapse
Affiliation(s)
- Ali Dakroub
- St. Francis Hospital and Heart Center, Roslyn, New York, USA
| | - Ali Dbouk
- Department of Medicine, Saint-Joseph University Medical School, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Aref Asfour
- Leeds Teaching Hospitals NHS Trust, West Yorkshire, United Kingdom
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, UAE
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Jiang H, Zheng S, Qian Y, Zhou Y, Dai H, Liang Y, He Y, Gao R, Lv H, Zhang J, Xia Z, Bian W, Yang T, Fu Q. Restored UBE2C expression in islets promotes β-cell regeneration in mice by ubiquitinating PER1. Cell Mol Life Sci 2023; 80:226. [PMID: 37486389 PMCID: PMC11072275 DOI: 10.1007/s00018-023-04868-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
Insulin deficiency may be due to the reduced proliferation capacity of islet β-cell, contributing to the onset of diabetes. It is therefore imperative to investigate the mechanism of the β-cell regeneration in the islets. NKX6.1, one of the critical β-cell transcription factors, is a pivotal element in β-cell proliferation. The ubiquitin-binding enzyme 2C (UBE2C) was previously reported as one of the downstream molecules of NKX6.1 though the exact function and mechanism of UBE2C in β-cell remain to be elucidated. Here, we determined a subpopulation of islet β-cells highly expressing UBE2C, which proliferate actively. We also discovered that β-cell compensatory proliferation was induced by UBE2C via the cell cycle renewal pathway in weaning and high-fat diet (HFD)-fed mice. Moreover, the reduction of β-cell proliferation led to insulin deficiency in βUbe2cKO mice and, therefore, developed type 2 diabetes. UBE2C was found to regulate PER1 degradation through the ubiquitin-proteasome pathway via its association with a ubiquitin ligase, CUL1. PER1 inhibition rescues UBE2C knockout-induced β-cell growth inhibition both in vivo and in vitro. Notably, overexpression of UBE2C via lentiviral transduction in pancreatic islets was able to relaunch β-cell proliferation in STZ-induced diabetic mice and therefore partially alleviated hyperglycaemia and glucose intolerance. This study indicates that UBE2C positively regulates β-cell proliferation by promoting ubiquitination and degradation of the biological clock suppressor PER1. The beneficial effect of UBE2C on islet β-cell regeneration suggests a promising application in treating diabetic patients with β-cell deficiency.
Collapse
Affiliation(s)
- Hemin Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuai Zheng
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Qian
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuncai Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Dai
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yucheng Liang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunqiang He
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Gao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Lv
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Zhang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiqing Xia
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenxuan Bian
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Qi Fu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Yang Y, Zhang D, Song M, Wang C, Lv J, Zhou J, Chen M, Ma L, Mei C. Macrophages promote heat stress nephropathy in mice via the C3a-C3aR-TNF pathway. Immunobiology 2023; 228:152337. [PMID: 36689826 DOI: 10.1016/j.imbio.2023.152337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/14/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
Heat-stress nephropathy (HSN) is associated with recurrent dehydration. However, the mechanisms underlying HSN remain largely unknown. In this study, we evaluated the role of dehydration in HSN and kidney injury in mice. Firstly, we found that complement was strongly activated in the mice that were exposed to dehydration; and among complement components, the interaction between C3a and its receptor, C3aR, was more closely associated with kidney injury. Then two-month-old mice were intraperitoneally injected with 2% dimethyl sulfoxide (DMSO) or the C3aR inhibitor SB290157 during dehydration. DMSO-treated mice exhibited excessive macrophage infiltration, renal cell apoptosis, and kidney fibrosis. In contrast, SB290157-treated mice had no apparent kidney injury. By fluorescence-activated cell sorting (FACS), we found that SB290157 treatment in mice remarkably inhibited macrophage infiltration and suppressed CCR2 expression in macrophages. In addition, C3a binding to C3aR promoted macrophage polarization toward the M1 phenotype and increased the production of TNF-α, which induced renal tubular epithelial cell (RTEC) apoptosis in vivo and in vitro. Interestingly, C3a treatment failed to directly induce TNF-α production and apoptosis in RTECs. However, TNF-α production in response to C3a treatment was significantly elevated when RTECs were cocultured with macrophages, suggesting that macrophages rather than RTECs are the target of C3a-C3aR interaction. At last, we proved that infusion of macrophages which highly expressed TNF-α would significantly deteriorate HSN in TNF-KO mice when they were exposed to recurrent dehydration. This study uncovers a novel mechanism underlying the pathogenesis of HSN, and a potential pathway to prevent kidney injury during dehydration.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nephrology, The 981(th) Hospital of Joint Logistic Support Force, Chengde, China; Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, The Navy Military Medical University, Shanghai, China.
| | - Dongjuan Zhang
- Department of Nephrology, The 981(th) Hospital of Joint Logistic Support Force, Chengde, China
| | - Minghui Song
- Clinical Laboratory, Hainan Hospital of General Hospital of Chinese People's Liberation Army, Sanya, China
| | - Chao Wang
- Kidney Diagnostic and Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Jiayi Lv
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, The Navy Military Medical University, Shanghai, China
| | - Jie Zhou
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, The Navy Military Medical University, Shanghai, China; Department of Nephrology, Affiliated ShuGuang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meihan Chen
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, The Navy Military Medical University, Shanghai, China; Department of Nephrology, Shanghai Tenth People's Hospital, TongJi University, Shanghai, China
| | - Lu Ma
- Kidney Diagnostic and Therapeutic Center of the Chinese People's Liberation Army, Beidaihe Rehabilitation and Recuperation Center of the Chinese People's Liberation Army, Qinhuangdao, China
| | - Changlin Mei
- Kidney Institution of the Chinese People's Liberation Army, Chang Zheng Hospital, The Navy Military Medical University, Shanghai, China.
| |
Collapse
|
4
|
Zhou X, Xu C, Dong J, Liao L. Role of renal tubular programed cell death in diabetic kidney disease. Diabetes Metab Res Rev 2023; 39:e3596. [PMID: 36401596 PMCID: PMC10078574 DOI: 10.1002/dmrr.3596] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/22/2022] [Accepted: 10/10/2022] [Indexed: 11/21/2022]
Abstract
The pathogenic mechanism of diabetic kidney disease (DKD) is involved in various functions; however, its inadequate characterisation limits the availability of effective treatments. Tubular damage is closely correlated with renal function and is thought to be the main contributor to the injury observed in early DKD. Programed cell death (PCD) occurs during the biological development of the living body. Accumulating evidence has clarified the fundamental role of abnormalities in tubular PCD during DKD pathogenesis. Among PCD types, classical apoptosis, autophagic cell death, and pyroptosis are the most studied and will be the focus of this review. Our review aims to elucidate the current knowledge of the mechanism of DKD and the potential therapeutic potential of drugs targeting tubular PCD pathways in DKD.
Collapse
Affiliation(s)
- Xiaojun Zhou
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Chunmei Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Jianjun Dong
- Division of Endocrinology, Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
5
|
Luppi P, Drain N, To R, Stolz D, Wallace C, Watkins S, Drain P. Autocrine C-peptide protects INS1 β cells against palmitic acid-induced oxidative stress in peroxisomes by inducing catalase. Endocrinol Diabetes Metab 2020; 3:e00147. [PMID: 32704568 PMCID: PMC7375117 DOI: 10.1002/edm2.147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/26/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS C-peptide, produced by pancreatic β cells and co-secreted in the bloodstream with insulin, has antioxidant properties in glucose- and hydrogen peroxide (H2O2)-exposed INS1 β cells. Palmitic acid, the most physiologically abundant long-chain free fatty acid in humans, is metabolized in peroxisomes of β cells accumulating H2O2 that can lead to oxidative stress. Here, we tested the hypothesis that C-peptide protects β cells from palmitic acid-induced stress by lowering peroxisomal H2O2. MATERIALS AND METHODS We exposed INS1 β cells to palmitic acid and C-peptide in the setting of increasing glucose concentration and tested for changes in parameters of stress and death. To study the ability of C-peptide to lower peroxisomal H2O2, we engineered an INS1 β cell line stably expressing the peroxisomal-targeted H2O2 sensor HyPer, whose fluorescence increases with cellular H2O2. An INS1 β cell line stably expressing a live-cell fluorescent catalase reporter was used to detect changes in catalase gene expression. RESULTS C-peptide protects INS1 β cells from the combined effect of palmitic acid and glucose by reducing peroxisomal H2O2 to baseline levels and increasing expression of catalase. CONCLUSIONS In conditions of glucolipotoxicity, C-peptide increases catalase expression and reduces peroxisomal oxidative stress and death of INS1 β cells. Maintenance of C-peptide secretion is a pro-survival requisite for β cells in adverse conditions. Loss of C-peptide secretion would render β cells more vulnerable to stress and death leading to secretory dysfunction and diabetes.
Collapse
Affiliation(s)
- Patrizia Luppi
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Nicholas Drain
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ramsey To
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Donna Stolz
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Callen Wallace
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Simon Watkins
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Peter Drain
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| |
Collapse
|
6
|
Poteryaeva ON, Usynin IF. [Molecular mechanisms of action and physiological effects of the proinsulin C-peptide (a systematic review)]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:196-207. [PMID: 32588825 DOI: 10.18097/pbmc20206603196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The C-peptide is a fragment of proinsulin, the cleavage of which forms active insulin. In recent years, new information has appeared on the physiological effects of the C-peptide, indicating its positive effect on many organs and tissues, including the kidneys, nervous system, heart, vascular endothelium and blood microcirculation. Studies on experimental models of diabetes mellitus in animals, as well as clinical trials in patients with diabetes, have shown that the C-peptide has an important regulatory effect on the early stages of functional and structural disorders caused by this disease. The C-peptide exhibits its effects through binding to a specific receptor on the cell membrane and activation of downstream signaling pathways. Intracellular signaling involves G-proteins and Ca2+-dependent pathways, resulting in activation and increased expression of endothelial nitric oxide synthase, Na+/K+-ATPase and important transcription factors involved in apoptosis, anti-inflammatory and other intracellular defense mechanisms. This review gives an idea of the C-peptide as a bioactive endogenous peptide that has its own biological activity and therapeutic potential.
Collapse
Affiliation(s)
- O N Poteryaeva
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| | - I F Usynin
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| |
Collapse
|
7
|
Bulboacă AE, Boarescu PM, Bolboacă SD, Blidaru M, Feștilă D, Dogaru G, Nicula CA. Comparative Effect Of Curcumin Versus Liposomal Curcumin On Systemic Pro-Inflammatory Cytokines Profile, MCP-1 And RANTES In Experimental Diabetes Mellitus. Int J Nanomedicine 2019; 14:8961-8972. [PMID: 31819412 PMCID: PMC6873975 DOI: 10.2147/ijn.s226790] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 10/25/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose Anti-inflammatory proprieties of curcumin were proved to be useful in various diseases, including diabetes mellitus. The aim of this study was to assess the anti-inflammatory comparative effect of curcumin solution with liposomal curcumin formula, regarding the improvement of serum levels of TNF-α (tumor necrosis factor-alpha), IL-6 (interleukin), IL-1α, IL-1β, MCP-1 (monocyte chemoattractant protein-1) and RANTES in experimental diabetes, induced by streptozotocin (STZ), in rats. Materials and methods Six groups of 7 rats were investigated regarding the effect of i.p. (intraperitoneal) administration of two concentrations of curcumin solution (CC1 and CC2) and two concentrations of liposomal curcumin (LCC1 and LCC2): group 1 – control group with i.p. administration of 1 mL saline solution, group 2 – i.p. STZ administration (60mg/kg bw, bw=body weight), group 3 – STZ+CC1 administration, group 4 – STZ+CC2 administration, group 5 – STZ+ LCC1 administration and group 6 – STZ+ LCC2 administration. The concentrations of curcumin formulas were 1 mg/0.1 kg bw for CC1 and LCC1 and 2 mg/0.1 kg bw for CC2 and LCC2, respectively. Serum levels of C-peptide (as an indicator of pancreatic function) and TNF-α, IL-6, IL-1α, IL-1β, MCP-1, and RANTES (as biomarkers for systemic inflammation) were assessed for each group. Results The plasma level of C-peptide showed significant improvements when LCC was administrated, with better results for LCC2 when compared to LCC1 (P<0.003). LCC2 pretreatment proved to be more efficient in reducing the level of TNF-α (P<0.003) and RANTES (P<0.003) than CC2 pretreatment. Upon comparing LCC2 with LCC1 formulas, the differences were significant for TNF-α (P=0.004), IL-1β (P=0.022), and RANTES (P=0.003) levels. Conclusion Liposomal curcumin in a dose of 2 mg/0.1 kg bw proved to have an optimum therapeutic effect as a pretreatment in DM induced by STZ. This result can constitute a base for clinical studies for curcumin efficiency as adjuvant therapy in type 1 DM.
Collapse
Affiliation(s)
- Adriana Elena Bulboacă
- Pathophysiology Department, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Paul Mihai Boarescu
- Pathophysiology Department, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Sorana D Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine And Pharmacy, Cluj-Napoca, Romania
| | - Mihai Blidaru
- Pathophysiology Department, Iuliu Hațieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| | - Dana Feștilă
- Department of Maxillofacial Surgery and Radiology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriela Dogaru
- Department of Physical Medicine and Rehabilitation, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Ariadna Nicula
- Department of Ophthalmology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
8
|
Yaribeygi H, Maleki M, Sathyapalan T, Sahebkar A. The effect of C-peptide on diabetic nephropathy: A review of molecular mechanisms. Life Sci 2019; 237:116950. [PMID: 31605709 DOI: 10.1016/j.lfs.2019.116950] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023]
Abstract
C-peptide is a small peptide connecting two chains of proinsulin molecule and is dissociated before the release of insulin. It is secreted in an equimolar amount to insulin from the pancreatic beta-cells into the circulation. Recent evidence demonstrates that it has other physiologic activities beyond its structural function. C-peptide modulates intracellular signaling pathways in various pathophysiologic states and, could potentially be a new therapeutic target for different disorders including diabetic complications. There is growing evidence that c-peptide has modulatory effects on the molecular mechanisms involved in the development of diabetic nephropathy. Although we have little direct evidence, pharmacological properties of c-peptide suggest that it can provide potent renoprotective effects especially, in a c-peptide deficient milieu as in type 1 diabetes mellitus. In this review, we describe possible molecular mechanisms by which c-peptide may improve renal efficiency in a diabetic milieu.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mina Maleki
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull HU3 2JZ, UK
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Yu G, Wang W, Wang X, Xu M, Zhang L, Ding L, Guo R, Shi Y. Network pharmacology-based strategy to investigate pharmacological mechanisms of Zuojinwan for treatment of gastritis. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:292. [PMID: 30382864 PMCID: PMC6211468 DOI: 10.1186/s12906-018-2356-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/18/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Zuojinwan (ZJW), a classic herbal formula, has been extensively used to treat gastric symptoms in clinical practice in China for centuries. However, the pharmacological mechanisms of ZJW still remain vague to date. METHODS In the present work, a network pharmacology-based strategy was proposed to elucidate its underlying multi-component, multi-target, and multi-pathway mode of action against gastritis. First we collected putative targets of ZJW based on TCMSP and STITCH databases, and a network containing the interactions between the putative targets of ZJW and known therapeutic targets of gastritis was built. Then four topological parameters, "degree", "betweenness", "closeness", and "coreness" were calculated to identify the major targets in the network. Furthermore, the major hubs were imported to the Metacore database to perform a pathway enrichment analysis. RESULTS A total of 118 nodes including 59 putative targets of ZJW were picked out as major hubs in terms of their topological importance. The results of pathway enrichment analysis indicated that putative targets of ZJW mostly participated in various pathways associated with anti-inflammation response, growth and development promotion and G-protein-coupled receptor signaling. More importantly, five putative targets of ZJW (EGFR, IL-6, IL-1β, TNF-α and MCP-1) and two known therapeutic targets of gastritis (CCKBR and IL-12β) and a link target NF-κB were recognized as active factors involved in the main biological functions of treatment, implying the underlying mechanisms of ZJW acting on gastritis. CONCLUSION ZJW could alleviate gastritis through the molecular mechanisms predicted by network pharmacology, and this research demonstrates that the network pharmacology approach can be an effective tool to reveal the mechanisms of traditional Chinese medicine (TCM) from a holistic perspective.
Collapse
Affiliation(s)
- Guohua Yu
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Wubin Wang
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Xu Wang
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Meng Xu
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Lili Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Lei Ding
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Rui Guo
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029 China
- Shenzhen Hospital, Beijing University of Chinese Medicine, No. 1 Dayun road, Sports New City Road, Shenzhen, 518172 China
| |
Collapse
|
10
|
Affiliation(s)
- J Wahren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Abstract
Kidney disease is a serious development in diabetes mellitus and poses an increasing clinical problem. Despite increasing incidence and prevalence of diabetic kidney disease, there have been no new therapies for this condition in the last 20 years. Mounting evidence supports a biological role for C-peptide, and findings from multiple studies now suggest that C-peptide may beneficially affect the disturbed metabolic and pathophysiological pathways leading to the development of diabetic nephropathy. Studies of C-peptide in animal models and in humans with type 1 diabetes all suggest a renoprotective effect for this peptide. In diabetic rodents, C-peptide reduces glomerular hyperfiltration and albuminuria. Cohort studies of diabetic patients with combined islet and kidney transplants suggest that maintained C-peptide secretion is protective of renal graft function. Further, in short-term studies of patients with type 1 diabetes, administration of C-peptide is also associated with a lowered hyperfiltration rate and reduced microalbuminuria. Thus, the available information suggests that type 1 diabetes should be regarded as a dual hormone deficiency disease and that clinical trials of C-peptide in diabetic nephropathy are both justified and urgently required.
Collapse
Affiliation(s)
- N J Brunskill
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| |
Collapse
|
12
|
Abstract
In this review, we present findings that support autocrine cell protection by C-peptide in the context of clinical studies of type 1 diabetes (T1D), which universally measure C-peptide serum levels as a surrogate for β cell functional mass. Over the last decade, evidence has accumulated that supports models in which C-peptide, cosecreted with insulin by pancreatic β cells, acts on peripheral targets including the vascular endothelium to reduce oxidative stress and apoptosis subsequent to exposure to diabetic insults. In parallel, as assays have become more sensitive, C-peptide has been detected in the circulation of most subjects with T1D where higher C-peptide levels are associated with fewer and slower development of diabetic microvascular complications, consistent with antioxidant protection by C-peptide. Clinical trials investigating C-peptide-replacement therapy effects have demonstrated amelioration of T1D nephropathy and neuropathy. Recently, the antioxidant action of C-peptide was extended to the β cells secreting it, that is an autocrine mechanism. Autocrine protection has major implications for the treatment of diabetes because the more C-peptide secreted, the more protection provided to the same β cells resulting in a slower decay in β cell functional mass over the time course of disease. Why β cells evolved to cosecrete an antioxidant C-peptide hormone together with the glycaemia-lowering insulin hormone is explored in the context of proposed evolutionary advantages of physiologically transient oxidative stress and insulin resistance as an adaptation for survival through times of fuel scarcity. The importance of recognizing autocrine C-peptide protection of functional β cell mass in observational clinical studies, and its therapeutic implications in interventional C-peptide-replacement studies, will be discussed.
Collapse
Affiliation(s)
- P Luppi
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - P Drain
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Wang J, Li Y, Xu M, Li D, Wang Y, Qi J, He K. C-peptide exhibits a late induction effect on matrix metallopeptidase-9 in high glucose-stimulated rat mesangial cells. Exp Ther Med 2016; 12:4142-4146. [PMID: 28101192 DOI: 10.3892/etm.2016.3873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/06/2016] [Indexed: 12/12/2022] Open
Abstract
Insufficient matrix metalloproteinase (MMP)-9 and MMP-2 is considered to be a contributor of extracellular matrix (ECM) accumulation in diabetic nephropathy (DN). C-peptide can reverse fibrosis, thus exerting a beneficial effect on DN. Whether C-peptide induces MMP-9 and MMP-2 to reverse ECM accumulation is not clear. In the present study, in order to determine ECM metabolism, rat mesangial cells were treated with high glucose (HG) and C-peptide intervention, then the early and late effects of C-peptide on HG-affected MMP-9 and MMP-2 were evaluated. Firstly, it was confirmed that HG mainly suppressed MMP-9 expression levels. Furthermore, C-peptide treatment induced MMP-9 expression at 6 h and suppressed it at 24 h, revealing the early dual effects of C-peptide on MMP-9 expression. Subsequently, significant increase in MMP-9 expression at 72, 96 and 120 h C-peptide treatment was observed. These changes in MMP-9 protein content confirmed its expression changes following late C-peptide treatment. Furthermore, at 96 and 120 h C-peptide treatment reversed the HG-inhibited MMP-9 secretion, further indicating the late induction effect of C-peptide on MMP-9. The present results demonstrated that C-peptide exerted a late induction effect on MMP-9 in HG-stimulated rat mesangial cells, which may be associated with the underlying mechanism of C-peptide's reversal effects on DN.
Collapse
Affiliation(s)
- Junxia Wang
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yanning Li
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Mingzhi Xu
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Dandan Li
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yu Wang
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Jinsheng Qi
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Kunyu He
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
14
|
Wahren J, Larsson C. C-peptide: new findings and therapeutic possibilities. Diabetes Res Clin Pract 2015; 107:309-19. [PMID: 25648391 DOI: 10.1016/j.diabres.2015.01.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/15/2015] [Indexed: 12/18/2022]
Abstract
Much new information on C-peptide physiology has appeared during the past 20 years. It has been shown that C-peptide binds specifically to cell membranes, elicits intracellular signaling via G-protein and Ca2+ -dependent pathways, resulting in activation and increased expression of endothelial nitric oxide synthase, Na+, K+ -ATPase and several transcription factors of importance for anti-inflammatory, anti-oxidant and cell protective mechanisms. Studies in animal models of diabetes and early clinical trials in patients with type 1 diabetes demonstrate that C-peptide in replacement doses elicits beneficial effects on early stages of diabetes-induced functional and structural abnormalities of the peripheral nerves, the kidneys and the retina. Much remains to be learned about C-peptide's mechanism of action and long-term clinical trials in type 1 diabetes subjects will be required to determine C-peptide's clinical utility. Nevertheless, even a cautious evaluation of the available evidence presents the picture of a bioactive endogenous peptide with therapeutic potential.
Collapse
Affiliation(s)
- John Wahren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Cebix AB, Stockholm, Sweden.
| | | |
Collapse
|
15
|
Richards JP, Bowles EA, Gordon WR, Ellsworth ML, Stephenson AH, Sprague RS. Mechanisms of C-peptide-mediated rescue of low O2-induced ATP release from erythrocytes of humans with type 2 diabetes. Am J Physiol Regul Integr Comp Physiol 2014; 308:R411-8. [PMID: 25552662 DOI: 10.1152/ajpregu.00420.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The circulating erythrocyte, by virtue of the regulated release of ATP in response to reduced oxygen (O2) tension, plays a key role in maintaining appropriate perfusion distribution to meet tissue needs. Erythrocytes from individuals with Type 2 diabetes (DM2) fail to release ATP in response to this stimulus. However, the administration of C-peptide and insulin at a 1:1 ratio was shown to restore this important physiological response in humans with DM2. To begin to investigate the mechanisms by which C-peptide influences low O2-induced ATP release, erythrocytes from healthy humans and humans with DM2 were exposed to reduced O2 in a thin-film tonometer, and ATP release under these conditions was compared with release during normoxia. We determined that 1) low O2-induced ATP release from DM2 erythrocytes is rescued by C-peptide in the presence and absence of insulin, 2) the signaling pathway activated by C-peptide in human erythrocytes involves PKC, as well as soluble guanylyl cyclase (sGC) and 3) inhibitors of cGMP degradation rescue low O2-induced ATP release from DM2 erythrocytes. These results provide support for the hypothesis that both PKC and sGC are components of a signaling pathway activated by C-peptide in human erythrocytes. In addition, since both C-peptide and phosphodiesterase 5 inhibitors rescue low O2-induced ATP release from erythrocytes of humans with DM2, their administration to humans with DM2 could aid in the treatment and/or prevention of the vascular disease associated with this condition.
Collapse
Affiliation(s)
- Jennifer P Richards
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Elizabeth A Bowles
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Weston R Gordon
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Mary L Ellsworth
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Alan H Stephenson
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Randy S Sprague
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
16
|
Yosten GLC, Maric-Bilkan C, Luppi P, Wahren J. Physiological effects and therapeutic potential of proinsulin C-peptide. Am J Physiol Endocrinol Metab 2014; 307:E955-68. [PMID: 25249503 PMCID: PMC4254984 DOI: 10.1152/ajpendo.00130.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Connecting Peptide, or C-peptide, is a product of the insulin prohormone, and is released with and in amounts equimolar to those of insulin. While it was once thought that C-peptide was biologically inert and had little biological significance beyond its role in the proper folding of insulin, it is now known that C-peptide binds specifically to the cell membranes of a variety of tissues and initiates specific intracellular signaling cascades that are pertussis toxin sensitive. Although it is now clear that C-peptide is a biologically active molecule, controversy still remains as to the physiological significance of the peptide. Interestingly, C-peptide appears to reverse the deleterious effects of high glucose in some tissues, including the kidney, the peripheral nerves, and the vasculature. C-peptide is thus a potential therapeutic agent for the treatment of diabetes-associated long-term complications. This review addresses the possible physiologically relevant roles of C-peptide in both normal and disease states and discusses the effects of the peptide on sensory nerve, renal, and vascular function. Furthermore, we highlight the intracellular effects of the peptide and present novel strategies for the determination of the C-peptide receptor(s). Finally, a hypothesis is offered concerning the relationship between C-peptide and the development of microvascular complications of diabetes.
Collapse
Affiliation(s)
- Gina L C Yosten
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri;
| | - Christine Maric-Bilkan
- Division of Cardiovascular Sciences, Vascular Biology and Hypertension Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Patrizia Luppi
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - John Wahren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; and Cebix Inc., Karolinska Institutet Science Park, Solna, Sweden
| |
Collapse
|
17
|
Luppi P, Drain P. Autocrine C-peptide mechanism underlying INS1 beta cell adaptation to oxidative stress. Diabetes Metab Res Rev 2014; 30:599-609. [PMID: 24459093 DOI: 10.1002/dmrr.2528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/20/2013] [Accepted: 01/04/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Excessive generation of reactive oxygen species (ROS) causing oxidative stress plays a major role in the pathogenesis of diabetes by inducing beta cell secretory dysfunction and apoptosis. Recent evidence has shown that C-peptide, produced by beta cells and co-secreted with insulin in the circulation of healthy individuals, decreases ROS and prevents apoptosis in dysfunctional vascular endothelial cells. In this study, we tested the hypothesis that an autocrine activity of C-peptide similarly decreases ROS when INS1 beta cells are exposed to stressful conditions of diabetes. METHODS Reactive oxygen species and apoptosis were induced in INS1 beta cells pretreated with C-peptide by either 22 mM glucose or 100 μM hydrogen peroxide (H2 O2 ). To test C-peptide's autocrine activity, endogenous C-peptide secretion was inhibited by the KATP channel opener diazoxide and H2 O2 -induced ROS assayed after addition of either exogenous C-peptide or the secretagogue glibenclamide. In similar experiments, extracellular potassium, which depolarizes the membrane otherwise hyperpolarized by diazoxide, was used to induce endogenous C-peptide secretion. ROS was measured using the cell-permeant dye chloromethyl-2',7'-dichlorodihydrofluorescein diacetate (CM-H2 -DCFDA). Insulin secretion and apoptosis were assayed by enzyme-linked immunosorbent assay. RESULTS C-peptide significantly decreased high glucose-induced and H2 O2 -induced ROS and prevented apoptosis of INS1 beta cells. Diazoxide significantly increased H2 O2 -induced ROS, which was reversed by exogenous C-peptide or glibenclamide or potassium chloride. CONCLUSIONS These findings demonstrate an autocrine C-peptide mechanism in which C-peptide is bioactive on INS1 beta cells exposed to stressful conditions and might function as a natural antioxidant to limit beta cell dysfunction and loss contributing to diabetes.
Collapse
Affiliation(s)
- Patrizia Luppi
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | | |
Collapse
|
18
|
Bhatt MP, Lim YC, Ha KS. C-peptide replacement therapy as an emerging strategy for preventing diabetic vasculopathy. Cardiovasc Res 2014; 104:234-44. [PMID: 25239825 DOI: 10.1093/cvr/cvu211] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Lack of C-peptide, along with insulin, is the main feature of Type 1 diabetes mellitus (DM) and is also observed in progressive β-cell loss in later stage of Type 2 DM. Therapeutic approaches to hyperglycaemic control have been ineffective in preventing diabetic vasculopathy, and alternative therapeutic strategies are necessary to target both hyperglycaemia and diabetic complications. End-stage organ failure in DM seems to develop primarily due to vascular dysfunction and damage, leading to two types of organ-specific diseases, such as micro- and macrovascular complications. Numerous studies in diabetic patients and animals demonstrate that C-peptide treatment alone or in combination with insulin has physiological functions and might be beneficial in preventing diabetic complications. Current evidence suggests that C-peptide replacement therapy might prevent and ameliorate diabetic vasculopathy and organ-specific complications through conservation of vascular function, as well as prevention of endothelial cell death, microvascular permeability, vascular inflammation, and neointima formation. In this review, we describe recent advances on the beneficial role of C-peptide replacement therapy for preventing diabetic complications, such as retinopathy, nephropathy, neuropathy, impaired wound healing, and inflammation, and further discuss potential beneficial effects of combined C-peptide and insulin supplement therapy to control hyperglycaemia and to prevent organ-specific complications.
Collapse
Affiliation(s)
- Mahendra Prasad Bhatt
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Kangwondaehak-gil 1, Chuncheon, Kangwon-do 200-701, Republic of Korea
| | - Young-Cheol Lim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Kangwondaehak-gil 1, Chuncheon, Kangwon-do 200-701, Republic of Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Kangwondaehak-gil 1, Chuncheon, Kangwon-do 200-701, Republic of Korea
| |
Collapse
|
19
|
Safar MM, Abdelsalam RM. H2S donors attenuate diabetic nephropathy in rats: Modulation of oxidant status and polyol pathway. Pharmacol Rep 2014; 67:17-23. [PMID: 25560570 DOI: 10.1016/j.pharep.2014.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 07/26/2014] [Accepted: 08/01/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND Sulfurous mineral water and its main active ingredient sodium hydrosulfide (NaHS) are major sources of H2S. The present study aimed to explore their protective effect on one of the serious long-term complications of diabetes; diabetic nephropathy. METHODS Sulfurous mineral water (as drinking water), NaHS (14 μmol/kg/day; ip), and gliclazide (10mg/kg; po) were administered daily for 6 weeks to streptozotocin (STZ)-diabetic rats. RESULTS STZ-induced diabetes was associated with body weight reduction, hyperglycemia, overproduction of glycated hemoglobin, as well as decline in serum insulin, C-peptide, and insulin like growth factor-I. Besides, diabetes impaired kidney functions and imposed oxidative and nitrosative stress as manifested by elevated contents of renal thiobarbituric acid reactive substances and nitric oxide, parallel to reduced glutathione content. These deleterious effects were antagonized by sulfurous water and to a better extent by NaHS. Activities of myeloperoxidase and sorbitol dehydrogenase were not altered by STZ or any of the treatments. However, STZ-induced diabetes was accompanied by an increment of aldose reductase which was only mitigated by gliclazide and NaHS. Histopathological examination of kidney sections corroborated the biochemical findings. CONCLUSION This study suggests a novel therapeutic approach for diabetic nephropathy using H2S donors.
Collapse
Affiliation(s)
- Marwa M Safar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Rania M Abdelsalam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
20
|
Richards JP, Yosten GLC, Kolar GR, Jones CW, Stephenson AH, Ellsworth ML, Sprague RS. Low O2-induced ATP release from erythrocytes of humans with type 2 diabetes is restored by physiological ratios of C-peptide and insulin. Am J Physiol Regul Integr Comp Physiol 2014; 307:R862-8. [PMID: 25080497 DOI: 10.1152/ajpregu.00206.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ATP release from erythrocytes in response to reduced oxygen (O2) tension stimulates local vasodilation, enabling these cells to direct perfusion to areas in skeletal muscle in need of O2. Erythrocytes of humans with type 2 diabetes do not release ATP in response to low O2. Both C-peptide and insulin individually inhibit low O2-induced ATP release from healthy human erythrocytes, yet when coadministered at physiological concentrations and ratios, no inhibition is seen. Here, we determined: that 1) erythrocytes of healthy humans and humans with type 2 diabetes possess a C-peptide receptor (GPR146), 2) the combination of C-peptide and insulin at physiological ratios rescues low O2-induced ATP release from erythrocytes of humans with type 2 diabetes, 3) residual C-peptide levels reported in humans with type 2 diabetes are not adequate to rescue low O2-induced ATP release in the presence of 1 nM insulin, and 4) the effects of C-peptide and insulin are neither altered by increased glucose levels nor explained by changes in erythrocyte deformability. These results suggest that the addition of C-peptide to the treatment regimen for type 2 diabetes could have beneficial effects on tissue oxygenation, which would help to ameliorate the concomitant peripheral vascular disease.
Collapse
Affiliation(s)
- Jennifer P Richards
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Gina L C Yosten
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Grant R Kolar
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Cory W Jones
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Alan H Stephenson
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Mary L Ellsworth
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| | - Randy S Sprague
- Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
21
|
Usarek M, Jagielski AK, Krempa P, Dylewska A, Kiersztan A, Drozak J, Girstun A, Derlacz RA, Bryla J. Proinsulin C-peptide potentiates the inhibitory action of insulin on glucose synthesis in primary cultured rabbit kidney-cortex tubules: Metabolic studies. Biochem Cell Biol 2014; 92:1-8. [DOI: 10.1139/bcb-2013-0074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Effects of equimolar concentrations of proinsulin C-peptide and insulin on glucose synthesis were studied in primary cultures of rabbit kidney-cortex tubules grown in the presence of alanine, glycerol, and octanoate. The rhodamine-labeled C-peptide entered renal tubular cells and localized in nuclei, both in the presence and absence of insulin; preincubations with the unlabeled compound inhibited internalization. C-peptide did not affect glucose formation when added alone but potentiated the inhibitory action of insulin by about 20% due to a decrease in flux through glucose-6-phosphate isomerase (GPI) and (or) glucose-6-phosphatase (G6Pase). GPI inhibition was caused by: (i) increased intracellular contents of fructose-1,6-bisphosphate and fructose-1-phosphate, inhibitors of the enzyme and (ii) reduced level of the phosphorylated GPI, which exhibits higher enzymatic activity in the presence of casein kinase 2. A decrease in flux through G6Pase, due to diminished import of G6P by G6P-transporter from the cytoplasm into endoplasmic reticulum lumen, is also suggested. The data show for the first time that in the presence of insulin and C-peptide, both GPI and G6P-ase may act as regulatory enzymes of renal gluconeogenic pathway.
Collapse
Affiliation(s)
- Michal Usarek
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Adam Konrad Jagielski
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Paulina Krempa
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Anna Dylewska
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Anna Kiersztan
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Jakub Drozak
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Agnieszka Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Rafal Andrzej Derlacz
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Jadwiga Bryla
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| |
Collapse
|
22
|
Li Y, Zhao M, Li B, Qi J. Dynamic localization and functional implications of C-peptide might for suppression of iNOS in high glucose-stimulated rat mesangial cells. Mol Cell Endocrinol 2013; 381:255-60. [PMID: 23973463 DOI: 10.1016/j.mce.2013.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 02/08/2023]
Abstract
Although C-peptide has unique beneficial effects on diabetic nephropathy (DN), its functional localization and molecular mechanism have not been fully clarified. Whether C-peptide exhibits its protective role through the regulation of inducible nitric oxide synthase (iNOS), a key enzyme in oxidative stress, is not clear. In this study, it was revealed that C-peptide could enter the nucleus of high glucose-stimulated mesangial cells, especially in a time-dependent manner by high glucose pretreatment, while no C-peptide was detected in low glucose-cultured mesangial cells. The dynamic functional localization of C-peptide might be the intrinsic cause of its unique beneficial effects for DN, which may provide a foundation for further clarification of its underlying mechanism. Our preliminarily data also shown C-peptide suppressed the iNOS expression. Taking together, these results revealed the dynamic functional localization of C-peptide by high glucose stimulation in rat mesangial cells, which might suppress expression of iNOS to exhibit its protective effects.
Collapse
Affiliation(s)
- Yanning Li
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal, Hebei Medical University, Shijiazhuang, China
| | | | | | | |
Collapse
|
23
|
Wen Q, Liang T, Qin F, Wei J, He Q, Luo X, Chen X, Zheng N, Huang R. Lyoniresinol 3α-O-β-D-glucopyranoside-mediated hypoglycaemia and its influence on apoptosis-regulatory protein expression in the injured kidneys of streptozotocin-induced mice. PLoS One 2013; 8:e81772. [PMID: 24312585 PMCID: PMC3849267 DOI: 10.1371/journal.pone.0081772] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/16/2013] [Indexed: 12/26/2022] Open
Abstract
Averrhoa carambola L. (Oxalidaceae) root (ACLR) has a long history of use in traditional Chinese medicine for treating diabetes and diabetic nephropathy (DN). (±)-Lyoniresinol 3α-O-β-D-glucopyranoside (LGP1, LGP2) were two chiral lignan glucosides that were isolated from the ACLR. The purpose of this study was to investigate the effect of LGP1 and LGP2-mediated hypoglycaemia on renal injury in streptozotocin (STZ)-induced diabetic mice. STZ-induced diabetic mice were administrated LGP1 and LGP2 orally (20, 40, 80 mg/kg body weight/d) for 14 days. Hyperglycaemia and the expression of related proteins such as nuclear factor-κB (NF-κB), caspase-3, -8, -9, and Bcl-associated X protein (Bax) were markedly decreased by LGP1 treatment. However, LGP2 treatment had no hypoglycaemic activity. Diabetes-dependent alterations in the kidney such as glomerular hypertrophy, excessive extracellular matrix amassing, and glomerular and tubular basement membrane thickening were improved after 14 days of LGP1 treatment. B cell lymphoma Leukaemia-2 (Bcl-2) expression was reduced in the STZ-induced diabetic mouse kidneys but was enhanced by LGP1 treatment. These findings suggest that LGP1 treatment may inhibit diabetic nephropathy progression and may regulate several pharmacological targets for treating or preventing diabetic nephropathy.
Collapse
Affiliation(s)
- Qingwei Wen
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Tao Liang
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Feizhang Qin
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinbin Wei
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiaoling He
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiu Luo
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoyu Chen
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Ni Zheng
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Renbin Huang
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
- * E-mail:
| |
Collapse
|
24
|
Luppi P, Kallas Å, Wahren J. Can C-peptide mediated anti-inflammatory effects retard the development of microvascular complications of type 1 diabetes? Diabetes Metab Res Rev 2013; 29:357-62. [PMID: 23463541 DOI: 10.1002/dmrr.2409] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 01/30/2013] [Accepted: 02/28/2013] [Indexed: 02/07/2023]
Abstract
Hyperglycemia is considered to be the major cause of microvascular complications of diabetes. Growing evidence highlights the importance of hyperglycemia-mediated inflammation in the initiation and progression of microvascular complications in type 1 diabetes. We hypothesize that lack of proinsulin C-peptide and lack of its anti-inflammatory properties contribute to the development of microvascular complications. Evidence gathered over the past 20 years shows that C-peptide is a biologically active peptide in its own right. It has been shown to reduce formation of reactive oxygen species and nuclear factor-κB activation induced by hyperglycemia, resulting in inhibition of cytokine, chemokine and cell adhesion molecule formation as well as reduced apoptotic activity. In addition, C-peptide stimulates and induces the expression of both Na⁺, K⁺-ATPase and endothelial nitric oxide synthase. Animal studies and small-scale clinical trials in type 1 diabetes patients suggest that C-peptide replacement combined with regular insulin therapy exerts beneficial effects on kidney and nerve dysfunction. Further clinical trials in patients with microvascular complications including measurements of inflammatory markers are warranted to explore the clinical significance of the aforementioned, previously unrecognized, C-peptide effects.
Collapse
Affiliation(s)
- Patrizia Luppi
- Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Rangos Research Center, Pittsburgh, PA 15201, USA
| | | | | |
Collapse
|
25
|
Affiliation(s)
- John Wahren
- Department of Molecular Medicine and Surgery, Karolinska Institutet,Stockholm, Sweden.
| | | | | |
Collapse
|
26
|
Sadik NAH, El-Seweidy MM, Shaker OG. The antiapoptotic effects of sulphurous mineral water and sodium hydrosulphide on diabetic rat testes. Cell Physiol Biochem 2011; 28:887-898. [PMID: 22178941 DOI: 10.1159/000335803] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2011] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND/AIMS It is well known that diabetes mellitus is associated with the impairment of testicular function. In the present study, we aimed to study the effects of sulphurous mineral water or sodium hydrosulphide (NaHS) on apoptotic testicular damage in rats with streptozotocin (STZ)-induced diabetes. METHODS Sulphurous mineral water (as drinking water) or NaHS (14 μmol/kg body weight/day, I.P.) was administered for 7 wks to rats with STZ-induced diabetes. RESULTS Hyperglycaemia, an overproduction of glycated haemoglobin (HbA1C) and a decline in serum insulin, C-peptide and insulin-like growth factor-I (IGF-I) were observed in diabetic rats. A decline in the serum testosterone level and an impairment of spermatogenesis, as indicated by a histopathological examination of diabetic rats, demonstrated significant testicular damage. Sulphurous mineral water and NaHS treatment may have improved the level of testicular GSH by blocking the overexpression of some apoptosis-related regulatory proteins such as Bax/Bcl-2, cytochrome c, caspase-9 and -3, and p53. This anti-apoptotic potential was associated with an increase in serum testosterone level and the amelioration of hyperglycaemia-related biochemical parameters. The histopathological examination was in harmony with the biochemical and molecular findings. CONCLUSION Our study provides the first indication that sulphurous mineral water and NaHS may have a novel anti-apoptotic potential that could be a useful treatment in preventing diabetes-induced testicular dysfunction.
Collapse
Affiliation(s)
- Nermin A H Sadik
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | | | | |
Collapse
|
27
|
Lindahl E, Nordquist L, Müller P, El Agha E, Friederich M, Dahlman-Wright K, Palm F, Jörnvall H. Early transcriptional regulation by C-peptide in freshly isolated rat proximal tubular cells. Diabetes Metab Res Rev 2011; 27:697-704. [PMID: 21618400 DOI: 10.1002/dmrr.1220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 03/06/2011] [Accepted: 05/17/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND Clinical studies have shown that proinsulin C-peptide exerts renoprotective effects in type 1 diabetes, although the underlying mechanisms are poorly understood. As C-peptide has been shown to induce several intracellular events and to localize to nuclei, we aimed to determine whether gene transcription is affected in proximal tubular kidney cells, and if so, whether the genes with altered transcription include those related to protective mechanisms. METHODS The effect of C-peptide incubation (2 h) on gene expression was investigated in freshly isolated proximal tubular cells from streptozotocin-diabetic Sprague-Dawley rats using global gene expression profiling and real-time quantitative polymerase chain reaction. Protein expression was assayed using western blotting. Different bioinformatic strategies were employed. RESULTS Gene transcription profiling demonstrated differential transcription of 492 genes (p < 0.01) after 2 h of C-peptide exposure, with the majority of these genes repressed (83%). Real-time quantitative polymerase chain reaction validation supported a trend of several G protein-coupled receptors being activated, and certain transcription factors being repressed. Also, C-peptide repressed the transcription of genes associated with the pathways of circulatory and inflammatory diseases. CONCLUSION This study shows that C-peptide exerts early effects on gene transcription in proximal tubular cells. The findings also bring further knowledge to the renoprotective mechanisms of C-peptide in type 1 diabetes, and support a transcriptional activity for C-peptide. It is suggested that C-peptide may play a regulatory role in the gene expression of proximal tubular cells.
Collapse
Affiliation(s)
- Emma Lindahl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Cifarelli V, Geng X, Styche A, Lakomy R, Trucco M, Luppi P. C-peptide reduces high-glucose-induced apoptosis of endothelial cells and decreases NAD(P)H-oxidase reactive oxygen species generation in human aortic endothelial cells. Diabetologia 2011; 54:2702-12. [PMID: 21773684 DOI: 10.1007/s00125-011-2251-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 06/20/2011] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS Reactive oxygen species (ROS) generated during hyperglycaemia are implicated in the development of diabetic vascular complications. High glucose increases oxidative stress in endothelial cells and induces apoptosis. A major source of ROS in endothelial cells exposed to glucose is the NAD(P)H oxidase enzyme. Several studies demonstrated that C-peptide, the product of proinsulin cleavage within the pancreatic beta cells, displays anti-inflammatory effects in certain models of vascular dysfunction. However, the molecular mechanism underlying this effect is unclear. We hypothesised that C-peptide reduces glucose-induced ROS generation by decreasing NAD(P)H oxidase activation and prevents apoptosis METHODS Human aortic endothelial cells (HAEC) were exposed to 25 mmol/l glucose in the presence or absence of C-peptide and tested for protein quantity and activity of caspase-3 and other apoptosis markers by ELISA, TUNEL and immunoblotting. Intracellular ROS were measured by flow cytometry using the ROS sensitive dye chloromethyl-2',7'-dichlorodihydrofluorescein diacetate (CM-H(2)-DCDFA). NAD(P)H oxidase activation was assayed by lucigenin. Membrane and cytoplasmic levels of the NAD(P)H subunit ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1) (RAC-1) and its GTPase activity were studied by immunoblotting and ELISA. RAC-1 (also known as RAC1) gene expression was investigated by quantitative real-time PCR. RESULTS C-peptide significantly decreased caspase-3 levels and activity and upregulated production of the anti-apoptotic factor B cell CLL/lymphoma 2 (BCL-2). Glucose-induced ROS production was quenched by C-peptide and this was associated with a decreased NAD(P)H oxidase activity and reduced RAC-1 membrane production and GTPase activity. CONCLUSIONS/INTERPRETATION In glucose-exposed endothelial cells, C-peptide acts as an endogenous antioxidant molecule by reducing RAC-1 translocation to membrane and NAD(P)H oxidase activation. By preventing oxidative stress, C-peptide protects endothelial cells from glucose-induced apoptosis.
Collapse
Affiliation(s)
- V Cifarelli
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, Children's Hospital of Pittsburgh, 530 45th Street, Pittsburgh, PA 15201, USA
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Reperfusion injury following hemorrhagic shock is accompanied by the development of a systemic inflammatory state that may lead to organ failure. Insulin connecting peptide (C-peptide) has been shown to exert anti-inflammatory effects in sepsis and myocardial ischemia-reperfusion injury and to ameliorate renal dysfunction in diabetic animals. Hence, we investigated the effect of C-peptide on kidney injury after hemorrhagic shock. We hypothesized that C-peptide would exert renoprotective effects by blunting inflammation. Hemorrhagic shock was induced in male rats (3-4 months old) by withdrawing blood from the femoral artery to a mean arterial pressure of 50 mmHg. Animals were kept in shock for 3 h, at which time they were rapidly resuscitated by returning their shed blood. At the time of resuscitation and every hour thereafter, one group of animals received C-peptide (280 nmol/kg), whereas another group received vehicle. Hemorrhagic shock resulted in significant rise in plasma levels of creatinine and elevated kidney neutrophil infiltration as evaluated by myeloperoxidase activity in vehicle-treated rats in comparison with sham rats, thus suggesting kidney injury. Treatment with C-peptide significantly attenuated the rise in creatinine and kidney myeloperoxidase activity when compared with vehicle group. At a molecular level, these effects of C-peptide were associated with reduced expression of the c-Fos subunit and reduced activation of the proinflammatory kinases, extracellular signal-regulated kinase 1/2 (ERK 1/2), and c-Jun N-terminal kinase and subsequently reduced DNA binding of activator protein 1 in the kidney. Thus, our data suggest that C-peptide may exert renoprotective effects after hemorrhagic shock by modulating activator protein 1 signaling.
Collapse
|
30
|
Nordquist L, Palm F, Andresen BT. Renal and vascular benefits of C-peptide: Molecular mechanisms of C-peptide action. Biologics 2011; 2:441-52. [PMID: 19707375 PMCID: PMC2721399 DOI: 10.2147/btt.s3221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
C-peptide has long been thought to be an inert byproduct of insulin production, but it has become apparent, and accepted, that C-peptide has important biological properties. C-peptide displays beneficial effects in many tissues affected by diabetic complications, such as increased peripheral blood flow and protection from renal damage. However, the mechanisms mediating these effects remain unclear. C-peptide interacts with cellular membranes at unidentified sites distinctive of the insulin family of receptors, and signals to multiple targets known to play a role in diabetes and diabetic complications, such as Na+/K+-ATPase and NOS. In general, the physiological and molecular effects of C-peptide resemble insulin, but C-peptide also possesses traits separate from those of insulin. These basic studies have been confirmed in human studies, suggesting that C-peptide may lend itself to clinical applications. However, the molecular and physiological properties of C-peptide are not completely elucidated, and large clinical studies have not begun. In order to further these goals, we critically summarize the current state of knowledge regarding C-peptide’s renal and vascular effects and the molecular signaling of C-peptide.
Collapse
Affiliation(s)
- Lina Nordquist
- Department of Medical Cell Biology, Division of Integrative Physiology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
31
|
Horita S, Seki G, Yamada H, Suzuki M, Koike K, Fujita T. Insulin resistance, obesity, hypertension, and renal sodium transport. Int J Hypertens 2011; 2011:391762. [PMID: 21629870 PMCID: PMC3095959 DOI: 10.4061/2011/391762] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 02/16/2011] [Indexed: 12/12/2022] Open
Abstract
Sodium transport through various nephron segments is quite important in regulating sodium reabsorption and blood pressure. Among several regulators of this process, insulin acts on almost all the nephron segments and is a strong enhancer of sodium reabsorption. Sodium-proton exchanger type 3 (NHE3) is a main regulator of sodium reabsorption in the luminal side of proximal tubule. In the basolateral side of the proximal tubule, sodium-bicarbonate cotransporter (NBCe1) mediates sodium and bicarbonate exit from tubular cells. In the distal nephron and the connecting tubule, epithelial sodium channel (ENaC) is of great importance to sodium reabsorption. NHE3, NBCe1, and ENaC are all regulated by insulin. Recently with-no-lysine (WNK) kinases, responsible for familial hypertension, stimulating sodium reabsorption in the distal nephron, have been found to be also regulated by insulin. We will discuss the regulation of renal sodium transport by insulin and its roles in the pathogenesis of hypertension in insulin resistance.
Collapse
Affiliation(s)
- Shoko Horita
- Department of Internal Medicine, Faculty of Medicine, The University of Tokyo 7-3-1 Hongo, Bunkyo, Tokyo 113-8655, Japan
| | | | | | | | | | | |
Collapse
|
32
|
El-Seweidy MM, Sadik NAH, Shaker OG. Role of sulfurous mineral water and sodium hydrosulfide as potent inhibitors of fibrosis in the heart of diabetic rats. Arch Biochem Biophys 2011; 506:48-57. [PMID: 20965145 DOI: 10.1016/j.abb.2010.10.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 10/10/2010] [Accepted: 10/14/2010] [Indexed: 02/05/2023]
Abstract
This study examined the downstream signaling whereby hyperglycemia may lead to myocardial fibrosis and apoptosis in the left ventricle of diabetic rats. The effects of sulfurous mineral water or sodium hydrosulfide (NaHS) as possible modulators were also examined. Sulfurous mineral water (as drinking water) and NaHS (14μmol/kg/day, IP) were administered for 7 week to rats with streptozotocin (STZ)-induced diabetes. Hyperglycemia, overproduction of glycated hemoglobin (HbA1C) and serum decline in insulin, C-peptide and insulin like growth factor-I (IGF-I) were observed in diabetic rats. Up-regulation of gene expressions of nuclear factor (NF-κB), profibrogenic growth factor such as transforming growth factor-β1 (TGF-β1), matrix metalloproteniase-2 (MMP-2), procollagen-1 and Fas ligand (Fas-L) were observed in the left ventricle of diabetic rats. A linear positive correlation between TGF-β1 and MMP-2 was also detected in diabetic group. An increase in hydroxyproline level and a disturbance in oxidative balance were detected in heart of diabetic rats. Sulfurous mineral water and NaHS treatment possibly, by improving cardiac GSH level, counteracted the enhanced expression of NF-κB, the profibrogenic and apoptotic parameters. Histopathological examination was in accordance with the biochemical and molecular findings of this study. We suggest a novel therapeutic approach of sulfurous mineral water and exogenous supplementation of H(2)S in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Mohamed M El-Seweidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | | | | |
Collapse
|
33
|
Jaya A, Shanthi P, Sachdanand P. Hypoglycemic Effect of Semecarpus anacardium in Streptozotocin Induced Diabetic Rats. INT J PHARMACOL 2010. [DOI: 10.3923/ijp.2010.435.443] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
34
|
Ibeas E, Fuentes L, Martín R, Hernández M, Nieto ML. Inflammatory protein sPLA2-IIA abrogates TNFα-induced apoptosis in human astroglioma cells: Crucial role of ERK. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1837-47. [DOI: 10.1016/j.bbamcr.2009.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 10/07/2009] [Accepted: 10/13/2009] [Indexed: 02/08/2023]
|
35
|
Abstract
Although long believed to be inert, C-peptide has now been shown to have definite biological effects both in vitro and in vivo in diabetic animals and in patients with type 1 diabetes. These effects point to a protective action of C-peptide against the development of diabetic microvascular complications. Underpinning these observations is undisputed evidence of C-peptide binding to a variety of cell types at physiologically relevant concentrations, and the downstream stimulation of multiple cell signaling pathways and gene transcription via the activation of numerous transcription factors. These pathways affect such fundamental cellular processes as re-absorptive and/or secretory phenotype, migration, growth, and survival. Whilst the receptor remains to be identified, experimental data points strongly to the existence of a specific G-protein-coupled receptor for C-peptide. Of the cell types studied so far, kidney tubular cells express the highest number of C-peptide binding sites. Accordingly, C-peptide exerts major effects on the function of these cells, and in the context of diabetic nephropathy appears to antagonise the pathophysiological effects of major disease mediators such as TGFbeta1 and TNFalpha. Therefore, based on its cellular activity profile C-peptide appears well positioned for development as a therapeutic tool to treat microvascular complications in type 1 diabetes.
Collapse
Affiliation(s)
- Claire E Hills
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | | |
Collapse
|
36
|
Abstract
Proinsulin C-peptide has been found to exert beneficial effects in many tissues affected by diabetic microvascular complications, including the kidneys. Glomerular hyperfiltration and microalbuminuria are early markers of diabetic nephropathy. C-peptide at physiological concentrations effectively reduces diabetes-induced glomerular hyperfiltration via constriction of the afferent arteriole, dilation of the efferent arteriole, and inhibition of tubular reabsorption in experimental models of type 1 diabetes. The glomerular hypertrophy and mesangial matrix expansion seen in early diabetes can be reduced or prevented by C-peptide administration, possibly via interference with TGF-beta1 and TNFalpha signaling. Several of C-peptide's reno-protective effects have been confirmed in human studies; reduced glomerular hyperfiltration and diminished urinary albumin excretion have been documented in type 1 diabetes patients receiving replacement doses of C-peptide for periods of up to 3 months. In this review, we critically summarize the current state of knowledge regarding C-peptide's renal effects, and discuss possible mechanisms of its beneficial effects in diabetic nephropathy.
Collapse
Affiliation(s)
- Lina Nordquist
- Department of Medical Cell Biology, Division of Integrative Physiology, Uppsala University, 75123 Uppsala, Sweden
| | | |
Collapse
|
37
|
|
38
|
Yanamadala V, Negoro H, Denker BM. Heterotrimeric G proteins and apoptosis: intersecting signaling pathways leading to context dependent phenotypes. Curr Mol Med 2009; 9:527-45. [PMID: 19601805 PMCID: PMC2822437 DOI: 10.2174/156652409788488784] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Apoptosis, a programmed cell death mechanism, is a fundamental process during the normal development and somatic maintenance of all multicellular organisms and thus is highly conserved and tightly regulated through numerous signaling pathways. Apoptosis is of particular clinical importance as its dysregulation contributes significantly to numerous human diseases, primarily through changes in the expression and activation of key apoptotic regulators. Each of the four families of heterotrimeric G proteins (G(s), G(i/o), G(q/11) and G(12/13)) has been implicated in numerous cellular signaling processes, including proliferation, transformation, migration, differentiation, and apoptosis. Heterotrimeric G protein signaling is an important but not widely studied mechanism regulating apoptosis. G protein Signaling and Apoptosis broadly cover two large bodies of literature and share numerous signaling pathways. Examination of the intersection between these two areas is the focus of this review. Several studies have implicated signaling through each of the four heterotrimeric G protein families to regulate apoptosis within numerous disease contexts, but the mechanism(s) are not well defined. Each G protein family has been shown to stimulate and/or inhibit apoptosis in a context-dependent fashion through regulating numerous downstream effectors including the Bcl-2 family, NF-kappaB, PI3 Kinase, MAP Kinases, and small GTPases. These cell-type specific and G protein coupled receptor dependent effects have led to a complex body of literature of G protein regulation of apoptosis. Here, we review the literature and summarize apoptotic signaling through each of the four heterotrimeric G protein families (and the relevant G protein coupled receptors), and discuss limitations and future directions for research on regulating apoptosis through G protein coupled mechanisms. Continued investigation in this field is essential for the identification of important targets for pharmacological intervention in numerous diseases.
Collapse
Affiliation(s)
- Vijay Yanamadala
- Renal Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Hideyuki Negoro
- Renal Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Bradley M. Denker
- Renal Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Nordquist L, Johansson M. Proinsulin C-peptide: friend or foe in the development of diabetes-associated complications? Vasc Health Risk Manag 2009; 4:1283-8. [PMID: 19337542 PMCID: PMC2663462 DOI: 10.2147/vhrm.s3955] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The proinsulin connecting peptide, C-peptide, is a cleavage product of insulin synthesis that is co-secreted with insulin by pancreatic β-cells following glucose stimulation. Recombinant insulin, used in the treatment of diabetes, lacks C-peptide and preclinical and clinical studies suggest that lack of C-peptide may exacerbate diabetes-associated complications. In accordance with this, several studies suggest that C-peptide has beneficial effects in a number of diabetes-associated complications. C-peptide has been shown to prevent diabetic neuropathy by improving endoneural blood flow, preventing neuronal apoptosis and by preventing axonal swelling. In the vascular system, C-peptide has been shown to prevent vascular dysfunction in diabetic rats, and to possess anti-proliferative effects on vascular smooth muscle cells, which may prevent atherosclerosis. However, C-peptide depositions have been found in arteriosclerotic lesions of patients with hyperinsulinemic diabetes and C-peptide has been shown to induce pro-inflammatory mediators, such as nuclear factor kappa B, inducible nitric oxide synthase, and cyclooxygenase-2, indicating that C-peptide treatment could be associated with side-effects that may accelerate the development of diabetes-associated complications. This review provides a brief summary of recent research in the field and discusses potential beneficial and detrimental effects of C-peptide supplementation.
Collapse
Affiliation(s)
- Lina Nordquist
- Department of Medical Cell Biology, Division of Integrative Physiology, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
40
|
Abstract
In recent years, accumulating evidence indicates a biological function for proinsulin C-peptide. These results challenge the traditional view that C-peptide is essentially inert and only useful as a surrogate marker of insulin release. Accordingly, it is now clear that C-peptide binds with high affinity to cell membranes, probably to a pertussis-toxin-sensitive G-protein-coupled receptor. Subsequently, multiple signalling pathways are potently and dose-dependently activated in multiple cell types by C-peptide with the resulting activation of gene transcription and altered cell phenotype. In diabetic animals and Type 1 diabetic patients, short-term studies indicate that C-peptide also enhances glucose disposal and metabolic control. Furthermore, results derived from animal models and clinical studies in Type 1 diabetic patients suggest a salutary effect of C-peptide in the prevention and amelioration of diabetic nephropathy and neuropathy. Therefore a picture of Type 1 diabetes as a dual-hormone-deficiency disease is developing, suggesting that the replacement of C-peptide alongside insulin should be considered in its management.
Collapse
Affiliation(s)
- Claire E Hills
- Department of Infection, Immunity and Inflammation, University of Leicester School of Medicine, Leicester LE1 7RH, UK
| | | |
Collapse
|
41
|
Sabayan B, Foroughinia F, Mowla A, Borhanihaghighi A. Role of insulin metabolism disturbances in the development of Alzheimer disease: mini review. Am J Alzheimers Dis Other Demen 2008; 23:192-9. [PMID: 18198237 PMCID: PMC10846104 DOI: 10.1177/1533317507312623] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alzheimer disease (AD) is the most common form of dementia. Different pathogenic processes have been studied that underlie characteristic changes of AD, including A beta protein aggregation, tau phosphorylation, neurovascular dysfunction, and inflammatory processes. Insulin exerts pleiotropic effects in neurons, such as the regulation of neural proliferation, apoptosis, and synaptic transmission. In this setting, any disturbance in the metabolism of insulin in the central nervous system (CNS) may put unfavorable effects on CNS function. It seems that disturbances in insulin metabolism, especially insulin resistance, play a role in most pathogenic processes that promote the development of AD. In this article, the relationships of disturbances in the metabolism of insulin in CNS with A beta peptides aggregation, tau protein phosphorylation, inflammatory markers, neuron apoptosis, neurovascular dysfunction, and neurotransmitter modulation are discussed, and future research directions are provided.
Collapse
Affiliation(s)
- Behnam Sabayan
- Student Research Center, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran
| | | | | | | |
Collapse
|
42
|
Hills CE, Brunskill NJ. Intracellular signalling by C-peptide. EXPERIMENTAL DIABETES RESEARCH 2008; 2008:635158. [PMID: 18382618 PMCID: PMC2276616 DOI: 10.1155/2008/635158] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Accepted: 02/06/2008] [Indexed: 12/11/2022]
Abstract
C-peptide, a cleavage product of the proinsulin molecule, has long been regarded as biologically inert, serving merely as a surrogate marker for insulin release. Recent findings demonstrate both a physiological and protective role of C-peptide when administered to individuals with type I diabetes. Data indicate that C-peptide appears to bind in nanomolar concentrations to a cell surface receptor which is most likely to be G-protein coupled. Binding of C-peptide initiates multiple cellular effects, evoking a rise in intracellular calcium, increased PI-3-kinase activity, stimulation of the Na(+)/K(+) ATPase, increased eNOS transcription, and activation of the MAPK signalling pathway. These cell signalling effects have been studied in multiple cell types from multiple tissues. Overall these observations raise the possibility that C-peptide may serve as a potential therapeutic agent for the treatment or prevention of long-term complications associated with diabetes.
Collapse
Affiliation(s)
- Claire E. Hills
- Department of Infection,
Immunity and Inflammation,
University of Leicester, Leicester School of Medicine,
P.O. Box 138, Leicester LE1 7RH,
England,
UK
| | - Nigel J. Brunskill
- Department of Infection,
Immunity and Inflammation,
University of Leicester, Leicester School of Medicine,
P.O. Box 138, Leicester LE1 7RH,
England,
UK
- Department of Nephrology,
Leicester General Hospital,
Gwendolen Road,
Leicester LE5 4PW,
England,
UK
| |
Collapse
|
43
|
Nordquist L, Lai EY, Sjöquist M, Patzak A, Persson AEG. Proinsulin C-peptide constricts glomerular afferent arterioles in diabetic mice. A potential renoprotective mechanism. Am J Physiol Regul Integr Comp Physiol 2007; 294:R836-41. [PMID: 18077505 DOI: 10.1152/ajpregu.00811.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE an increased glomerular filtration rate (GFR) has been postulated as a potential mechanism involved in the progression of diabetic nephropathy. Studies suggest that C-peptide exerts a renoprotective effect on diabetes. The peptide decreases hyperfiltration in patients with type 1 diabetes, as well as in diabetic animal models. In this study, we investigated whether C-peptide causes a change in arteriolar diameter. RESEARCH DESIGN AND METHODS C57-Bl mice were made diabetic by means of a single intravenous injection of alloxan 2 wk prior to the experiment. Age-matched normoglycemic mice served as controls. Afferent arterioles, intact with the glomeruli, were dissected and microperfused. The effect of luminal application of C-peptide, compared with scrambled C-peptide or vehicle, was investigated. The effect of the Rho-kinase inhibitor Y-27632 was also investigated. RESULTS C-peptide constricted afferent arterioles in diabetic mice by -27% compared with the control value. Normoglycemic arterioles administered C-peptide displayed a delayed and minute response (-4%). Scrambled C-peptide or vehicle administration, whether administered to hyperglycemic or normoglycemic mice, did not induce any effect. Addition of Y-27632 abolished the effect of C-peptide. CONCLUSION C-peptide induces constriction of afferent arterioles in diabetic mice. This can reduce enhanced GFR and may be one of the mechanisms in the renoprotective action of C-peptide in diabetes.
Collapse
Affiliation(s)
- Lina Nordquist
- Department of Medical Cell Biology, Division of Physiology, University of Uppsala, Uppsala, Sweden
| | | | | | | | | |
Collapse
|
44
|
Yanamadala V, Negoro H, Gunaratnam L, Kong T, Denker BM. Galpha12 stimulates apoptosis in epithelial cells through JNK1-mediated Bcl-2 degradation and up-regulation of IkappaBalpha. J Biol Chem 2007; 282:24352-63. [PMID: 17565996 DOI: 10.1074/jbc.m702804200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Apoptosis is an essential mechanism for the maintenance of somatic tissues, and when dysregulated can lead to numerous pathological conditions. G proteins regulate apoptosis in addition to other cellular functions, but the roles of specific G proteins in apoptosis signaling are not well characterized. Galpha12 stimulates protein phosphatase 2A (PP2A), a serine/threonine phosphatase that modulates essential signaling pathways, including apoptosis. Herein, we examined whether Galpha12 regulates apoptosis in epithelial cells. Inducible expression of Galpha12 or constitutively active (QL)alpha12 in Madin-Darby canine kidney cells led to increased apoptosis with expression of QLalpha12, but not Galpha12. Inducing QLalpha12 led to degradation of the anti-apoptotic protein Bcl-2 (via the proteasome pathway), increased JNK activity, and up-regulated IkappaBalpha protein levels, a potent stimulator of apoptosis. Furthermore, the QLalpha12-stimulated activation of JNK was blocked by inhibiting PP2A. To characterize endogenous Galpha12 signaling pathways, non-transfected MDCK-II and HEK293 cells were stimulated with thrombin. Thrombin activated endogenous Galpha12 (confirmed by GST-tetratricopeptide repeat (TPR) pull-downs) and stimulated apoptosis in both cell types. The mechanisms of thrombin-stimulated apoptosis through endogenous Galpha12 were nearly identical to the mechanisms identified in QLalpha12-MDCK cells and included loss of Bcl-2, JNK activation, and up-regulation of IkappaBalpha. Knockdown of the PP2A catalytic subunit in HEK293 cells inhibited thrombin-stimulated apoptosis, prevented JNK activation, and blocked Bcl-2 degradation. In summary, Galpha12 has a major role in regulating epithelial cell apoptosis through PP2A and JNK activation leading to loss of Bcl-2 protein expression. Targeting these pathways in vivo may lead to new therapeutic strategies for a variety of disease processes.
Collapse
Affiliation(s)
- Vijay Yanamadala
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
45
|
Luzi L, Zerbini G, Caumo A. C-peptide: a redundant relative of insulin? Diabetologia 2007; 50:500-2. [PMID: 17225123 DOI: 10.1007/s00125-006-0576-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Accepted: 10/24/2006] [Indexed: 12/13/2022]
Affiliation(s)
- L Luzi
- Section of Nutrition and Metabolism, San Raffaele Scientific Institute, 20132, Milan, Italy.
| | | | | |
Collapse
|
46
|
Wahren J, Ekberg K, Jörnvall H. C-peptide is a bioactive peptide. Diabetologia 2007; 50:503-9. [PMID: 17235526 DOI: 10.1007/s00125-006-0559-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 10/24/2006] [Indexed: 11/24/2022]
Affiliation(s)
- J Wahren
- Department of Molecular Medicine and Surgery, Karolinska Hospital, 171 76, Stockholm, Sweden.
| | | | | |
Collapse
|
47
|
Bugliani M, Torri S, Lupi R, Del Guerra S, Grupillo M, Del Chiaro M, Mosca F, Boggi U, Del Prato S, Marchetti P. Effects of C-peptide on isolated human pancreatic islet cells. Diabetes Metab Res Rev 2007; 23:215-9. [PMID: 16874846 DOI: 10.1002/dmrr.674] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Recent data have demonstrated that pro-insulin-derived C-peptide can affect the function of several different cell types. We hypothesized that C-peptide might have an influence on the function and survival of isolated human islets. METHODS Islets were prepared by combining enzymatic digestion and density gradient centrifugation, and the effects of human C-peptide were evaluated acutely and after 24-h incubation. Insulin secretion, apoptosis, quantitative RT-PCR and western-blotting experiments were then performed. RESULTS Glucose-stimulated insulin secretion was not affected by C-peptide and, accordingly, mRNA expression of glucose transporter 2 and glucokinase did not differ between islets pre-cultured or not with the hormone. However, apoptosis was significantly lower in islets exposed to C-peptide than in control islets. This was accompanied by a significant increase of mRNA and protein expression of Bcl2, an anti-apoptotic molecule, with no change in the expression of Bax, a pro-apoptotic molecule. CONCLUSION These results show that in human islets pro-insulin C-peptide has no direct effects on insulin secretion, but it decreases islet cell apoptosis. A direct role of C-peptide on beta-cell mass regulation is therefore suggested.
Collapse
Affiliation(s)
- Marco Bugliani
- Department of Endocrinology and Metabolism - Metabolic Unit, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|