1
|
Ha Y, Kang W, Roh J, Jung Y, Lee H, Park T. Acetyl tributyl citrate attenuates 5-fluorouracil-induced inflammation, oxidative stress, and apoptosis in human keratinocytes. Biochem Pharmacol 2024; 230:116606. [PMID: 39489222 DOI: 10.1016/j.bcp.2024.116606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
5-Fluorouracil (5-FU) is a commonly used chemotherapy drug that effectively destroys cancer cells. Despite its widespread use and efficacy, it also presents considerable challenges, particularly with adverse effects on rapidly dividing normal cells, such as keratinocytes. These detrimental effects are attributed to inflammatory, oxidative, and apoptotic potentials, leading to severe skin disorders. Due to the lack of specific remedies for 5-FU-induced dermatological side effects, conventional treatments are applied instead, which provide limited relief and have drawbacks. This study investigated the impact of acetyl tributyl citrate (ATBC) in 5-FU-treated human keratinocytes. The findings indicated that ATBC substantially reduced inflammation caused by 5-FU, as demonstrated by nuclear translocation of nuclear factor kappa B and expression of its downstream genes, including tumor necrosis factor, interleukin 1 beta (IL1B), and IL6. ATBC also markedly decreased oxidative stress, indicated by reactive oxygen species levels and the antioxidant gene expression such as superoxide dismutase 1 (SOD1), SOD2, and heme oxygenase 1 in 5-FU-treated cells. Furthermore, ATBC attenuated 5-FU-induced apoptosis, as determined by lactate dehydrogenase release and Annexin V/propidium iodide flow cytometry, with the potential involvement of interferon-related genes. Following this, protein kinase C delta was predicted as a possible molecular target of ATBC. These findings propose ATBC as a therapeutic agent for managing the cutaneous side effects associated with 5-FU treatment.
Collapse
Affiliation(s)
- Yoojeong Ha
- Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Wesuk Kang
- Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jiyun Roh
- Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yearim Jung
- Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyunbin Lee
- Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Taesun Park
- Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
2
|
Fotros D, Tabar MS, Chegini M, Bahrizadeh M, Sadeghi A, Rabbani A, Yari Z, Hekmatdoost A. Adherence to the dietary approaches to stop hypertension (DASH) and risk of pancreatic steatosis. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:190. [PMID: 39567983 PMCID: PMC11580558 DOI: 10.1186/s41043-024-00628-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/20/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND The Dietary Approach to Stop Hypertension (DASH) has shown positive effects on various health factors that may be related to pancreatic steatosis (PS). This study aimed to investigate the association between adherence to the DASH diet and the risk of developing PS. METHODS This case-control study was conducted on 278 patients diagnosed with gallstone disease and referred to Taleghani Hospital (Tehran, Iran). Among the participants, 89 were diagnosed with PS based on an endoscopic ultrasound (EUS) examination, while 189 patients did not exhibit this condition. The dietary intake of patients was assessed using a validated food frequency questionnaire (FFQ). Participants were classified based on the DASH diet score. Multiple logistic regression models estimated crude and multivariable-adjusted odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The mean ± SD of DASH score in PS patients and controls was 23.68 ± 4.38 and 25.27 ± 4.2, respectively (P = 0.006). The risk of PS in the highest tertile of DASH score was 64% lower than the lowest tertile (OR = 0.36, 95%CI: 0.17-0.75, P = 0.005) after full adjustment for confounders. Also, more intake of vegetables and whole grains and less intake of sodium, red and processed meat were each significantly associated with reduced risk of PS. CONCLUSIONS Our data prove that adherence to the DASH diet was associated with a lower risk of PS. Further prospective studies are warranted to confirm these associations and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Danial Fotros
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Shaygan Tabar
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maedeh Chegini
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Bahrizadeh
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Research Institute for Gastroenterology and Liver Diseases of Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhassan Rabbani
- Department of General Surgery, Ayatollah Taleghani Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Zahra Yari
- Department of Nutrition Research, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
4
|
Collins J, Piscopio RA, Reyland ME, Johansen CG, Benninger RKP, Farnsworth NL. Cleavage of protein kinase c δ by caspase-3 mediates proinflammatory cytokine-induced apoptosis in pancreatic islets. J Biol Chem 2024; 300:107611. [PMID: 39074637 PMCID: PMC11381875 DOI: 10.1016/j.jbc.2024.107611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
In type 1 diabetes (T1D), autoreactive immune cells infiltrate the pancreas and secrete proinflammatory cytokines that initiate cell death in insulin producing islet β-cells. Protein kinase C δ (PKCδ) plays a role in mediating cytokine-induced β-cell death; however, the exact mechanisms are not well understood. To address this, we used an inducible β-cell specific PKCδ KO mouse as well as a small peptide inhibitor of PKCδ. We identified a role for PKCδ in mediating cytokine-induced β-cell death and have shown that inhibiting PKCδ protects pancreatic β-cells from cytokine-induced apoptosis in both mouse and human islets. We determined that cytokines induced nuclear translocation and activity of PKCδ and that caspase-3 cleavage of PKCδ may be required for cytokine-mediated islet apoptosis. Further, cytokine activated PKCδ increases activity both of proapoptotic Bax with acute treatment and C-Jun N-terminal kinase with prolonged treatment. Overall, our results suggest that PKCδ mediates cytokine-induced apoptosis via nuclear translocation, cleavage by caspase-3, and upregulation of proapoptotic signaling in pancreatic β-cells. Combined with the protective effects of PKCδ inhibition with δV1-1, the results of this study will aid in the development of novel therapies to prevent or delay β-cell death and preserve β-cell function in T1D.
Collapse
Affiliation(s)
- Jillian Collins
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Robert A Piscopio
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chelsea G Johansen
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Richard K P Benninger
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Nikki L Farnsworth
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA; Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
5
|
Hali M, Pinto N, Gleason N, Kowluru A. Regulatory Roles of Histone Deacetylation in Metabolic Stress-Induced Expression of Caspase Recruitment Domain-Containing Protein 9 (CARD9) in Pancreatic β-Cells. Int J Mol Sci 2023; 24:15994. [PMID: 37958977 PMCID: PMC10647342 DOI: 10.3390/ijms242115994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
CARD9, a scaffolding protein, has been implicated in the pathogenesis of metabolic diseases, including obesity and diabetes. We recently reported novel roles for CARD9 in islet β-cell dysregulation under duress of gluco (HG)- and glucolipotoxic (GLT) stress. CARD9 expression was also increased in β-cells following exposure to HG and GLT stress. The current study is aimed at understanding the putative roles of histone deacetylation in HG- and GLT-induced expression of CARD9. Using two structurally distinct inhibitors of histone deacetylases (HDACs), namely trichostatin (TSA) and suberoylanilide hydroxamic acid (SAHA), we provide the first evidence to suggest that the increased expression of CARD9 seen under duress of HG and GLT stress is under the regulatory control of histone deacetylation. Interestingly, the expression of protein kinase Cδ (PKCδ), a known upstream regulator of CARD9 activation, is also increased under conditions of metabolic stress. However, it is resistant to TSA and SAHA, suggesting that it is not regulated via histone deacetylation. Based on these data, we propose that targeting the appropriate HDACs, which mediate the expression (and function) of CARD9, might be the next step to further enhance our current understanding of the roles of CARD9 in islet dysfunction under metabolic stress and diabetes.
Collapse
Affiliation(s)
- Mirabela Hali
- Biomedical Research Service, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; (M.H.); (N.P.); (N.G.)
| | - Nelson Pinto
- Biomedical Research Service, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; (M.H.); (N.P.); (N.G.)
| | - Noah Gleason
- Biomedical Research Service, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; (M.H.); (N.P.); (N.G.)
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Anjaneyulu Kowluru
- Biomedical Research Service, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; (M.H.); (N.P.); (N.G.)
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Caldart F, de Pretis N, Luchini C, Ciccocioppo R, Frulloni L. Pancreatic steatosis and metabolic pancreatic disease: a new entity? Intern Emerg Med 2023; 18:2199-2208. [PMID: 37462859 PMCID: PMC10635967 DOI: 10.1007/s11739-023-03364-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/30/2023] [Indexed: 08/24/2023]
Abstract
Overweight and obesity are some of the most important health challenges. Many diseases are related to these metabolic disorders, and, among them, the pancreatic fat accumulation, also called "pancreatic steatosis" or "nonalcoholic fatty pancreas", seems to have an emerging role in different conditions. There are different method to evaluate the fat content in the pancreas, such as histology, different imaging techniques and endoscopic ultrasound, but there is no gold standard for the correct diagnosis and for the identification of "inter/intralobular" and "intra-acinar" pancreatic fat. However, the fat storage in the pancreas is linked to chronic inflammation and to several conditions, such as acute and chronic pancreatitis, type 2 diabetes mellitus and pancreatic cancer. In addition, pancreatic fat accumulation has also been demonstrated to play a role in surgical outcome after pancreatectomy, in particular for the development of postoperative pancreatic fistula. Different possible therapeutic approaches have been proposed, but there is still a lack of evidence. The aim of this review is to report the current evidence about the relationship between the obesity, the pancreatic fat accumulation and its potential role in pancreatic diseases.
Collapse
Affiliation(s)
- Federico Caldart
- Gastroenterology B Unit, University of Verona-Verona Hospital, Verona, Italy.
| | - Nicolò de Pretis
- Gastroenterology B Unit, University of Verona-Verona Hospital, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Rachele Ciccocioppo
- Gastroenterology B Unit, University of Verona-Verona Hospital, Verona, Italy
| | - Luca Frulloni
- Gastroenterology B Unit, University of Verona-Verona Hospital, Verona, Italy
| |
Collapse
|
7
|
Ciarambino T, Crispino P, Leto G, Minervini G, Para O, Giordano M. Microbiota and Glucidic Metabolism: A Link with Multiple Aspects and Perspectives. Int J Mol Sci 2023; 24:10409. [PMID: 37373556 DOI: 10.3390/ijms241210409] [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: 05/22/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The global prevalence of overweight and obesity has dramatically increased in the last few decades, with a significant socioeconomic burden. In this narrative review, we include clinical studies aiming to provide the necessary knowledge on the role of the gut microbiota in the development of diabetic pathology and glucose-metabolism-related disorders. In particular, the role of a certain microbial composition of the fermentative type seems to emerge without a specific link to the development in certain subjects of obesity and the chronic inflammation of the adipose tissues, which underlies the pathological development of all the diseases related to glucose metabolism and metabolic syndrome. The gut microbiota plays an important role in glucose tolerance. Conclusion. New knowledge and new information is presented on the development of individualized therapies for patients affected by all the conditions related to reduced glucose tolerance and insulin resistance.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, ASL Caserta, 81037 Caserta, Italy
| | - Pietro Crispino
- Internal Medicine Department, Hospital of Latina, ASL Latina, 04100 Latina, Italy
| | - Gaetano Leto
- Department of Experimental Medicine, University La Sapienza Roma, 00185 Rome, Italy
| | - Giovanni Minervini
- Internal Medicine Department, Hospital of Lagonegro, AOR San Carlo, 85042 Lagonegro, Italy
| | - Ombretta Para
- Internal Emergency Department, Hospital of Careggi, University of Florence, 50121 Florence, Italy
| | - Mauro Giordano
- Department of Medical Science, University of Campania, L. Vanvitelli, 81100 Naples, Italy
| |
Collapse
|
8
|
Biswas S, Mahapatra E, Das S, Roy M, Mukherjee S. PEITC: A resounding molecule averts metastasis in breast cancer cells in vitro by regulating PKCδ/Aurora A interplay. Heliyon 2022; 8:e11656. [PMID: 36458309 PMCID: PMC9706142 DOI: 10.1016/j.heliyon.2022.e11656] [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: 07/26/2021] [Revised: 01/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Background/aim Intricate association and aberrant activation of serine/threonine kinase (STK) family proteins like Polo-like kinase (PLK1) and Aurora kinase (Aurora A abruptly regulate mitotic entry whereas activation of PKCδ), another important member of STK family conversely induces apoptosis which is preceded by cell cycle arrest. These STKs are considered as major determinant of oncogenicity. Therefore, the contributory role of Aurora A/PLK-1 axis in mitotic control and PKCδ in apoptosis control and their reciprocity in cancer research is an emerging area to explore. The present study investigated the intricate involvement of STKs in breast cancer cells (MCF-7 and MDA-MB-231) and their disruption by PEITC. Methods Both MCF-7 and MDA-MB-231 cells were checked for clonogenic assay, cell-cycle analysis and the results were compared with normal MCF-10A, Western blotting, TUNEL & DNA-fragmentation assay, wound healing, transwell migration assays in presence and absence of PEITC. Results PEITC was found to increase the expression of PKCδ with subsequent nuclear translocation. Nuclear translocation of PKCδ was accompanied by inhibition of nuclear lamin vis a vis phosphorylation of Nrf2 at Ser 40 alongside nuclear accumulation of phospho-Nrf2. Activated PKCδ furthermore exerted its apoptotic effect by negatively regulating Aurora A and consequentially PLK1; indicating activation of PLK1 by Aurora A. Involvement of PEITC induced PKCδ activation and Aurora A inhibition was ascertained by using Rottlerin/Aurora A Inhibitor. Discussion & conclusion Natural isothiocyanates like PEITC efficiently altered the functional abilities of STKs concerning their entangled functional interplay. Such alterations in protein expression by PEITC was chaperoned with inhibition of the aggressiveness of breast cancer cells and ultimately induction of apoptosis.
Collapse
Affiliation(s)
- Souvick Biswas
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Elizabeth Mahapatra
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Salini Das
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Madhumita Roy
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| | - Sutapa Mukherjee
- Dept of Environmental Carcinogenesis & Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700 026, India
| |
Collapse
|
9
|
Effects of adrenergic-stimulated lipolysis and cytokine production on in vitro mouse adipose tissue-islet interactions. Sci Rep 2022; 12:15831. [PMID: 36138030 PMCID: PMC9499973 DOI: 10.1038/s41598-022-18262-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/08/2022] [Indexed: 11/08/2022] Open
Abstract
Inflammatory cytokines and non-esterified fatty acids (NEFAs) are obesity-linked factors that disturb insulin secretion. The aim of this study was to investigate whether pancreatic adipose tissue (pWAT) is able to generate a NEFA/cytokine overload within the pancreatic environment and as consequence to impact on insulin secretion. Pancreatic fat is a minor fat depot, therefore we used high-fat diet (HFD) feeding to induce pancreatic steatosis in mice. Relative Adipoq and Lep mRNA levels were higher in pWAT of HFD compared to chow diet mice. Regardless of HFD, Adipoq and Lep mRNA levels of pWAT were at least 10-times lower than those of epididymal fat (eWAT). Lipolysis stimulating receptors Adrb3 and Npr1 were expressed in pWAT and eWAT, and HFD reduced their expression in eWAT only. In accordance, HFD impaired lipolysis in eWAT but not in pWAT. Despite expression of Npr mRNA, lipolysis was stimulated solely by the adrenergic agonists, isoproterenol and adrenaline. Short term co-incubation of islets with CD/HFD pWAT did not alter insulin secretion. In the presence of CD/HFD eWAT, glucose stimulated insulin secretion only upon isoproterenol-induced lipolysis, i.e. in the presence of elevated NEFA. Isoproterenol augmented Il1b and Il6 mRNA levels both in pWAT and eWAT. These results suggest that an increased sympathetic activity enhances NEFA and cytokine load of the adipose microenvironment, including that of pancreatic fat, and by doing so it may alter beta-cell function.
Collapse
|
10
|
Free fatty acid-induced miR-181a-5p stimulates apoptosis by targeting XIAP and Bcl2 in hepatic cells. Life Sci 2022; 301:120625. [PMID: 35551953 DOI: 10.1016/j.lfs.2022.120625] [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: 03/11/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/22/2022]
Abstract
AIMS Non-alcoholic fatty liver disease is one of the major health concerns in the World. The dietary free fatty acids (FFAs) affect the metabolic status of the hepatocytes by modulating cellular pathways. In this study, we showed that free fatty acids stimulate apoptosis by upregulating miR-181a-5p expression, which in turn targets XIAP and Bcl2. METHODS Huh7 cells were incubated with FFAs for 72 h and the expression of XIAP, Bcl2, bax, pAkt, Akt, PTEN and β-actin were determined by Western blots, and miR-181a-5p expression was determined using real-time RT-PCR. The Huh7 cells were transfected with either miR-181a-5p pre-miRs or anti-miR-181a-5p and the regulation of apoptosis and proliferation was studied. Three groups of C57BL/6 mice (n = 6 per group) were fed with standard diet, CSAA or CDAA diet for 6, 18, 32 and 54 weeks. Total protein and RNA were isolated from the liver tissues and used for Western blots and real-time RT-PCR respectively. KEY FINDINGS FFAs inhibited Akt phosphorylation, expression of XIAP and Bcl2, while upregulating the expression of PTEN, bax, and miR-181a-5p in Huh7 cells. Similar results were observed when the Huh7 cells were transfected with miR-181a-5p premiRs, while these changes were reversed in anti-miR-181a-5p-transfected, FFA-treated Huh7 cells. The CDAA-fed mice showed a significant inhibition of Akt phosphorylation, XIAP and Bcl2, whereas PTEN and bax expression were upregulated. The expression of miR-181a-5p was also significantly higher in CDAA-fed mice. SIGNIFICANCE These findings showed that free fatty acids induced apoptosis via upregulating miR-181a-5p in hepatic cells.
Collapse
|
11
|
Wagner R, Eckstein SS, Yamazaki H, Gerst F, Machann J, Jaghutriz BA, Schürmann A, Solimena M, Singer S, Königsrainer A, Birkenfeld AL, Häring HU, Fritsche A, Ullrich S, Heni M. Metabolic implications of pancreatic fat accumulation. Nat Rev Endocrinol 2022; 18:43-54. [PMID: 34671102 DOI: 10.1038/s41574-021-00573-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/15/2022]
Abstract
Fat accumulation outside subcutaneous adipose tissue often has unfavourable effects on systemic metabolism. In addition to non-alcoholic fatty liver disease, which has received considerable attention, pancreatic fat has become an important area of research throughout the past 10 years. While a number of diagnostic approaches are available to quantify pancreatic fat, multi-echo Dixon MRI is currently the most developed method. Initial studies have shown associations between pancreatic fat and the metabolic syndrome, impaired glucose metabolism and type 2 diabetes mellitus. Pancreatic fat is linked to reduced insulin secretion, at least under specific circumstances such as prediabetes, low BMI and increased genetic risk of type 2 diabetes mellitus. This Review summarizes the possible causes and metabolic consequences of pancreatic fat accumulation. In addition, potential therapeutic approaches for addressing pancreatic fat accumulation are discussed.
Collapse
Affiliation(s)
- Robert Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sabine S Eckstein
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Felicia Gerst
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Benjamin Assad Jaghutriz
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Potsdam-Rehbrücke, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Michele Solimena
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stephan Singer
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral, and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Susanne Ullrich
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Tübingen, Germany.
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
12
|
Martínez-Montoro JI, Damas-Fuentes M, Fernández-García JC, Tinahones FJ. Role of the Gut Microbiome in Beta Cell and Adipose Tissue Crosstalk: A Review. Front Endocrinol (Lausanne) 2022; 13:869951. [PMID: 35634505 PMCID: PMC9133559 DOI: 10.3389/fendo.2022.869951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decades, obesity has reached epidemic proportions worldwide. Obesity is a chronic disease associated with a wide range of comorbidities, including insulin resistance and type 2 diabetes mellitus (T2D), which results in significant burden of disease and major consequences on health care systems. Of note, intricate interactions, including different signaling pathways, are necessary for the establishment and progression of these two closely related conditions. Altered cell-to-cell communication among the different players implicated in this equation leads to the perpetuation of a vicious circle associated with an increased risk for the development of obesity-related complications, such as T2D, which in turn contributes to the development of cardiovascular disease. In this regard, the dialogue between the adipocyte and pancreatic beta cells has been extensively studied, although some connections are yet to be fully elucidated. In this review, we explore the potential pathological mechanisms linking adipocyte dysfunction and pancreatic beta cell impairment/insulin resistance. In addition, we evaluate the role of emerging actors, such as the gut microbiome, in this complex crosstalk.
Collapse
Affiliation(s)
- José Ignacio Martínez-Montoro
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
- *Correspondence: José Ignacio Martínez-Montoro, ; Francisco J. Tinahones,
| | - Miguel Damas-Fuentes
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - José Carlos Fernández-García
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: José Ignacio Martínez-Montoro, ; Francisco J. Tinahones,
| |
Collapse
|
13
|
Sato T, Ishiwatari C, Kaneko YK, Ishikawa Y, Kimura Y, Watanabe N, Aoshima I, Matsuda Y, Nakayama T, Chiba R, Fujinuki T, Iwata K, Lu Q, Usuki T, Sakane F, Ishikawa T. Diacylglycerol kinase δ functions as a proliferation suppressor in pancreatic β-cells. FASEB J 2021; 35:e21420. [PMID: 33774855 DOI: 10.1096/fj.202001279rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/11/2022]
Abstract
Although an aberrant reduction in pancreatic β-cell mass contributes to the pathogenesis of diabetes, the mechanism underlying the regulation of β-cell mass is poorly understood. Here, we show that diacylglycerol kinase δ (DGKδ) is a key enzyme in the regulation of β-cell mass. DGKδ expression was detected in the nucleus of β-cells. We developed β-cell-specific DGKδ knockout (βDGKδ KO) mice, which showed lower blood glucose, higher plasma insulin levels, and better glucose tolerance compared to control mice. Moreover, an increased number of small islets and Ki-67-positive islet cells, as well as elevated cyclin B1 expression in the islets, were detected in the pancreas of βDGKδ KO mice. DGKδ knockdown in the β-cell line MIN6 induced significant increases in bromodeoxyuridine (BrdU) incorporation and cyclin B1 expression. Finally, we confirmed that streptozotocin-induced hyperglycemia and β-cell loss were alleviated in βDGKδ KO mice. Thus, suppressing the expression or enzymatic activity of DGKδ that functions as a suppressor of β-cell proliferation could be a novel therapeutic approach to increase β-cell mass for the treatment of diabetes.
Collapse
Affiliation(s)
- Taiji Sato
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Chihiro Ishiwatari
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yoko Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yuki Kimura
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Naoya Watanabe
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Ikumi Aoshima
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yukari Matsuda
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takahiro Nakayama
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Rina Chiba
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takahiro Fujinuki
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kai Iwata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Qiang Lu
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Takako Usuki
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
14
|
Šrámek J, Němcová-Fürstová V, Kovář J. Molecular Mechanisms of Apoptosis Induction and Its Regulation by Fatty Acids in Pancreatic β-Cells. Int J Mol Sci 2021; 22:4285. [PMID: 33924206 PMCID: PMC8074590 DOI: 10.3390/ijms22084285] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic β-cell failure and death contribute significantly to the pathogenesis of type 2 diabetes. One of the main factors responsible for β-cell dysfunction and subsequent cell death is chronic exposure to increased concentrations of FAs (fatty acids). The effect of FAs seems to depend particularly on the degree of their saturation. Saturated FAs induce apoptosis in pancreatic β-cells, whereas unsaturated FAs are well tolerated and are even capable of inhibiting the pro-apoptotic effect of saturated FAs. Molecular mechanisms of apoptosis induction by saturated FAs in β-cells are not completely elucidated. Saturated FAs induce ER stress, which in turn leads to activation of all ER stress pathways. When ER stress is severe or prolonged, apoptosis is induced. The main mediator seems to be the CHOP transcription factor. Via regulation of expression/activity of pro- and anti-apoptotic Bcl-2 family members, and potentially also through the increase in ROS production, CHOP switches on the mitochondrial pathway of apoptosis induction. ER stress signalling also possibly leads to autophagy signalling, which may activate caspase-8. Saturated FAs activate or inhibit various signalling pathways, i.e., p38 MAPK signalling, ERK signalling, ceramide signalling, Akt signalling and PKCδ signalling. This may lead to the activation of the mitochondrial pathway of apoptosis, as well. Particularly, the inhibition of the pro-survival Akt signalling seems to play an important role. This inhibition may be mediated by multiple pathways (e.g., ER stress signalling, PKCδ and ceramide) and could also consequence in autophagy signalling. Experimental evidence indicates the involvement of certain miRNAs in mechanisms of FA-induced β-cell apoptosis, as well. In the rather rare situations when unsaturated FAs are also shown to be pro-apoptotic, the mechanisms mediating this effect in β-cells seem to be the same as for saturated FAs. To conclude, FA-induced apoptosis rather appears to be preceded by complex cross talks of multiple signalling pathways. Some of these pathways may be regulated by decreased membrane fluidity due to saturated FA incorporation. Few data are available concerning molecular mechanisms mediating the protective effect of unsaturated FAs on the effect of saturated FAs. It seems that the main possible mechanism represents a rather inhibitory intervention into saturated FA-induced pro-apoptotic signalling than activation of some pro-survival signalling pathway(s) or metabolic interference in β-cells. This inhibitory intervention may be due to an increase of membrane fluidity.
Collapse
Affiliation(s)
- Jan Šrámek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | - Vlasta Němcová-Fürstová
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | | |
Collapse
|
15
|
Benito-Vicente A, Jebari-Benslaiman S, Galicia-Garcia U, Larrea-Sebal A, Uribe KB, Martin C. Molecular mechanisms of lipotoxicity-induced pancreatic β-cell dysfunction. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:357-402. [PMID: 33832653 DOI: 10.1016/bs.ircmb.2021.02.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes (T2D), a heterogeneous disorder derived from metabolic dysfunctions, leads to a glucose overflow in the circulation due to both defective insulin secretion and peripheral insulin resistance. One of the critical risk factor for T2D is obesity, which represents a global epidemic that has nearly tripled since 1975. Obesity is characterized by chronically elevated free fatty acid (FFA) levels, which cause deleterious effects on glucose homeostasis referred to as lipotoxicity. Here, we review the physiological FFA roles onto glucose-stimulated insulin secretion (GSIS) and the pathological ones affecting many steps of the mechanisms and modulation of GSIS. We also describe in vitro and in vivo experimental evidences addressing lipotoxicity in β-cells and the role of saturation and chain length of FFA on the potency of GSIS stimulation. The molecular mechanisms underpinning lipotoxic-β-cell dysfunction are also reviewed. Among them, endoplasmic reticulum stress, oxidative stress and mitochondrial dysfunction, inflammation, impaired autophagy and β-cell dedifferentiation. Finally therapeutic strategies for the β-cells dysfunctions such as the use of metformin, glucagon-like peptide 1, thiazolidinediones, anti-inflammatory drugs, chemical chaperones and weight are discussed.
Collapse
Affiliation(s)
- Asier Benito-Vicente
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Shifa Jebari-Benslaiman
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Unai Galicia-Garcia
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Molecular Biophysics, Fundación Biofísica Bizkaia, Leioa, Spain
| | - Asier Larrea-Sebal
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Molecular Biophysics, Fundación Biofísica Bizkaia, Leioa, Spain
| | - Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia San Sebastián, Spain
| | - Cesar Martin
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
16
|
Engin AB, Engin A. Protein Kinases Signaling in Pancreatic Beta-cells Death and Type 2 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:195-227. [PMID: 33539017 DOI: 10.1007/978-3-030-49844-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes (T2D) is a worldwide serious public health problem. Insulin resistance and β-cell failure are the two major components of T2D pathology. In addition to defective endoplasmic reticulum (ER) stress signaling due to glucolipotoxicity, β-cell dysfunction or β-cell death initiates the deleterious vicious cycle observed in T2D. Although the primary cause is still unknown, overnutrition that contributes to the induction of the state of low-grade inflammation, and the activation of various protein kinases-related metabolic pathways are main factors leading to T2D. In this chapter following subjects, which have critical checkpoints regarding β-cell fate and protein kinases pathways are discussed; hyperglycemia-induced β-cell failure, chronic accumulation of unfolded protein in β-cells, the effect of intracellular reactive oxygen species (ROS) signaling to insulin secretion, excessive saturated free fatty acid-induced β-cell apoptosis, mitophagy dysfunction, proinflammatory responses and insulin resistance, and the reprogramming of β-cell for differentiation or dedifferentiation in T2D. There is much debate about selecting proposed therapeutic strategies to maintain or enhance optimal β-cell viability for adequate insulin secretion in T2D. However, in order to achieve an effective solution in the treatment of T2D, more intensive clinical trials are required on newer therapeutic options based on protein kinases signaling pathways.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
17
|
Kolczynska K, Loza-Valdes A, Hawro I, Sumara G. Diacylglycerol-evoked activation of PKC and PKD isoforms in regulation of glucose and lipid metabolism: a review. Lipids Health Dis 2020; 19:113. [PMID: 32466765 PMCID: PMC7257441 DOI: 10.1186/s12944-020-01286-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Protein kinase C (PKC) and Protein kinase D (PKD) isoforms can sense diacylglycerol (DAG) generated in the different cellular compartments in various physiological processes. DAG accumulates in multiple organs of the obese subjects, which leads to the disruption of metabolic homeostasis and the development of diabetes as well as associated diseases. Multiple studies proved that aberrant activation of PKCs and PKDs contributes to the development of metabolic diseases. DAG-sensing PKC and PKD isoforms play a crucial role in the regulation of metabolic homeostasis and therefore might serve as targets for the treatment of metabolic disorders such as obesity and diabetes.
Collapse
Affiliation(s)
- Katarzyna Kolczynska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Angel Loza-Valdes
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Izabela Hawro
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Grzegorz Sumara
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland.
| |
Collapse
|
18
|
Oshima M, Pechberty S, Bellini L, Göpel SO, Campana M, Rouch C, Dairou J, Cosentino C, Fantuzzi F, Toivonen S, Marchetti P, Magnan C, Cnop M, Le Stunff H, Scharfmann R. Stearoyl CoA desaturase is a gatekeeper that protects human beta cells against lipotoxicity and maintains their identity. Diabetologia 2020; 63:395-409. [PMID: 31796987 PMCID: PMC6946759 DOI: 10.1007/s00125-019-05046-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/14/2019] [Indexed: 01/02/2023]
Abstract
AIMS/HYPOTHESIS During the onset of type 2 diabetes, excessive dietary intake of saturated NEFA and fructose lead to impaired insulin production and secretion by insulin-producing pancreatic beta cells. The majority of data on the deleterious effects of lipids on functional beta cell mass were obtained either in vivo in rodent models or in vitro using rodent islets and beta cell lines. Translating data from rodent to human beta cells remains challenging. Here, we used the human beta cell line EndoC-βH1 and analysed its sensitivity to a lipotoxic and glucolipotoxic (high palmitate with or without high glucose) insult, as a way to model human beta cells in a type 2 diabetes environment. METHODS EndoC-βH1 cells were exposed to palmitate after knockdown of genes related to saturated NEFA metabolism. We analysed whether and how palmitate induces apoptosis, stress and inflammation and modulates beta cell identity. RESULTS EndoC-βH1 cells were insensitive to the deleterious effects of saturated NEFA (palmitate and stearate) unless stearoyl CoA desaturase (SCD) was silenced. SCD was abundantly expressed in EndoC-βH1 cells, as well as in human islets and human induced pluripotent stem cell-derived beta cells. SCD silencing induced markers of inflammation and endoplasmic reticulum stress and also IAPP mRNA. Treatment with the SCD products oleate or palmitoleate reversed inflammation and endoplasmic reticulum stress. Upon SCD knockdown, palmitate induced expression of dedifferentiation markers such as SOX9, MYC and HES1. Interestingly, SCD knockdown by itself disrupted beta cell identity with a decrease in mature beta cell markers INS, MAFA and SLC30A8 and decreased insulin content and glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION The present study delineates an important role for SCD in the protection against lipotoxicity and in the maintenance of human beta cell identity. DATA AVAILABILITY Microarray data and all experimental details that support the findings of this study have been deposited in in the GEO database with the GSE130208 accession code.
Collapse
Affiliation(s)
- Masaya Oshima
- Université Paris Descartes, Institut Cochin, Inserm U1016, 123 bd du Port-Royal, 75014, Paris, France
| | - Séverine Pechberty
- Université Paris Descartes, Institut Cochin, Inserm U1016, 123 bd du Port-Royal, 75014, Paris, France
| | - Lara Bellini
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Sven O Göpel
- Bioscience Metabolism, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Mélanie Campana
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Claude Rouch
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Julien Dairou
- Université Paris Descartes CNRS UMR 8601, Paris, France
| | - Cristina Cosentino
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Federica Fantuzzi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Sanna Toivonen
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Piero Marchetti
- University of Pisa, Department of Clinical and Experimental Medicine, Pisa, Italy
| | - Christophe Magnan
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology, ULB Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Hervé Le Stunff
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
- Université Paris-Sud, CNRS UMR 9197, Institut des Neurosciences Paris-Saclay (Neuro-PSI) - CNRS UMR 9197, Orsay, France
| | - Raphaël Scharfmann
- Université Paris Descartes, Institut Cochin, Inserm U1016, 123 bd du Port-Royal, 75014, Paris, France.
| |
Collapse
|
19
|
Las G, Oliveira MF, Shirihai OS. Emerging roles of β-cell mitochondria in type-2-diabetes. Mol Aspects Med 2020; 71:100843. [PMID: 31918997 DOI: 10.1016/j.mam.2019.100843] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 10/25/2022]
Abstract
Type-2-Diabetes (T2D) is the most common metabolic disease in the world today. It erupts as a result of peripheral insulin resistance combined with hyperinsulinemia followed by suppression of insulin secretion from pancreatic β-cells. Mitochondria play a central role in β-cells by sensing glucose and also by mediating the suppression of insulin secretion in T2D. Here, we will summarize the evidence accumulated for the roles of β-cells mitochondria in T2D. We will present an updated view on how mitochondria in β-cells have been associated with T2D, from the genetic, bioenergetic, redox and structural points of view. The emerging picture is that mitochondrial structure and dysfunction directly contribute to β-cell function and in the pathogenesis of T2D.
Collapse
Affiliation(s)
- Guy Las
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Marcus F Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, RJ, Brazil.
| | - Orian S Shirihai
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Urbaniak SK, Boguszewska K, Szewczuk M, Kaźmierczak-Barańska J, Karwowski BT. 8-Oxo-7,8-Dihydro-2'-Deoxyguanosine (8-oxodG) and 8-Hydroxy-2'-Deoxyguanosine (8-OHdG) as a Potential Biomarker for Gestational Diabetes Mellitus (GDM) Development. Molecules 2020; 25:molecules25010202. [PMID: 31947819 PMCID: PMC6982778 DOI: 10.3390/molecules25010202] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/30/2019] [Accepted: 01/01/2020] [Indexed: 12/12/2022] Open
Abstract
The growing clinical and epidemiological significance of gestational diabetes mellitus results from its constantly increasing worldwide prevalence, obesity, and overall unhealthy lifestyle among women of childbearing age. Oxidative stress seems to be the most important predictor of gestational diabetes mellitus development. Disturbances in the cell caused by oxidative stress lead to different changes in biomolecules, including DNA. The nucleobase which is most susceptible to oxidative stress is guanine. Its damage results in two main modifications: 8-hydroxy-2′-deoxyguanosineor 8-oxo-7,8-dihydro-2′-deoxyguanosine. Their significant level can indicate pathological processes during pregnancy, like gestational diabetes mellitus and probably, type 2 diabetes mellitus after pregnancy. This review provides an overview of current knowledge on the use of 8-hydroxy-2′-deoxyguanosineand/or 8-oxo-7,8-dihydro-2′-deoxyguanosine as a biomarker in gestational diabetes mellitus and allows us to understand the mechanism of 8-hydroxy-2′-deoxyguanosineand/or 8-oxo-7,8-dihydro-2′-deoxyguanosine generation during this disease.
Collapse
|
21
|
Xue Y, Guo C, Hu F, Zhu W, Mao S. Maternal undernutrition induces fetal hepatic lipid metabolism disorder and affects the development of fetal liver in a sheep model. FASEB J 2019; 33:9990-10004. [PMID: 31167079 DOI: 10.1096/fj.201900406r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Undernutrition accelerates body fat mobilization to alleviate negative energy balance, which disrupts homeostasis of lipid metabolism in maternal liver. However, little is known about its effect on fetal metabolism and development. Here, a sheep model was used to explore whether maternal undernutrition induces fetal lipid metabolism disorder and further inhibits fetal hepatic development. Twenty pregnant ewes were either fed normally or restricted to 30% level for 15 d, after which fetal hepatic samples were collected to conduct transcriptome, metabolome, histomorphology, and biochemical analysis. Results showed that maternal undernutrition altered the general transcriptome profile and metabolic mode in fetal liver. Fatty acid oxidation and ketogenesis were enhanced in fetal livers of undernourished ewes, which might be promoted by the activated peroxisome proliferator-activated receptor α signaling pathway, whereas cholesterol, steroid, and fatty acid synthesis were repressed. Maternal undernutrition increased triglyceride synthesis, decreased triglyceride degradation, and inhibited phospholipid degradation and synthesis in fetal liver. In addition, our data revealed that maternal undernutrition extremely inhibited DNA replication, cell cycle progression, and antiapoptosis and broke the balance between cell proliferation and apoptosis in fetal liver, indicating that maternal undernutrition affects the growth and development of fetal liver. Generally, these findings provide evidence that maternal undernutrition during pregnancy disturbs fetal lipid metabolism and inhibits fetal hepatic development in sheep, which greatly contribute to the further study of fetal metabolism and development in human beings.-Xue, Y., Guo, C., Hu, F., Zhu, W., Mao, S. Maternal undernutrition induces fetal hepatic lipid metabolism disorder and affects the development of fetal liver in a sheep model.
Collapse
Affiliation(s)
- Yanfeng Xue
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Changzheng Guo
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Fan Hu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Shengyong Mao
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
22
|
Gerst F, Wagner R, Oquendo MB, Siegel-Axel D, Fritsche A, Heni M, Staiger H, Häring HU, Ullrich S. What role do fat cells play in pancreatic tissue? Mol Metab 2019; 25:1-10. [PMID: 31113756 PMCID: PMC6600604 DOI: 10.1016/j.molmet.2019.05.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/10/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023] Open
Abstract
Background It is now generally accepted that obesity is a major risk factor for type 2 diabetes mellitus (T2DM). Hepatic steatosis in particular, as well as visceral and ectopic fat accumulation within tissues, is associated with the development of the disease. We recently presented the first study on isolated human pancreatic adipocytes and their interaction with islets [Gerst, F., Wagner, R., Kaiser, G., Panse, M., Heni, M., Machann, J., et al., 2017. Metabolic crosstalk between fatty pancreas and fatty liver: effects on local inflammation and insulin secretion. Diabetologia 60(11):2240–2251.]. The results indicate that the function of adipocytes depends on the overall metabolic status in humans which, in turn, differentially affects islet hormone release. Scope of Review This review summarizes former and recent studies on factors derived from adipocytes and their effects on insulin-secreting β-cells, with particular emphasis on the human pancreas. The adipocyte secretome is discussed with a special focus on its influence on insulin secretion, β-cell survival and apoptotic β-cell death. Major Conclusions Human pancreatic adipocytes store lipids and release adipokines, metabolites, and pro-inflammatory molecules in response to the overall metabolic, humoral, and neuronal status. The differentially regulated adipocyte secretome impacts on endocrine function, i.e., insulin secretion, β-cell survival and death which interferes with glycemic control. This review attempts to explain why the extent of pancreatic steatosis is associated with reduced insulin secretion in some studies but not in others.
Collapse
Affiliation(s)
- Felicia Gerst
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Morgana Barroso Oquendo
- German Center for Diabetes Research (DZD), Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Dorothea Siegel-Axel
- German Center for Diabetes Research (DZD), Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Harald Staiger
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Susanne Ullrich
- German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, Tübingen, Germany.
| |
Collapse
|
23
|
Farnsworth NL, Walter R, Piscopio RA, Schleicher WE, Benninger RKP. Exendin-4 overcomes cytokine-induced decreases in gap junction coupling via protein kinase A and Epac2 in mouse and human islets. J Physiol 2019; 597:431-447. [PMID: 30412665 PMCID: PMC6332825 DOI: 10.1113/jp276106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 11/07/2018] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS The pancreatic islets of Langerhans maintain glucose homeostasis through insulin secretion, where insulin secretion dynamics are regulated by intracellular Ca2+ signalling and electrical coupling of the insulin producing β-cells in the islet. We have previously shown that cytokines decrease β-cell coupling and that compounds which increase cAMP can increase coupling. In both mouse and human islets exendin-4, which increases cAMP, protected against cytokine-induced decreases in coupling and in mouse islets preserved glucose-stimulated calcium signalling by increasing connexin36 gap junction levels on the plasma membrane. Our data indicate that protein kinase A regulates β-cell coupling through a fast mechanism, such as channel gating or membrane organization, while Epac2 regulates slower mechanisms of regulation, such as gap junction turnover. Increases in β-cell coupling with exendin-4 may protect against cytokine-mediated β-cell death as well as preserve insulin secretion dynamics during the development of diabetes. ABSTRACT The pancreatic islets of Langerhans maintain glucose homeostasis. Insulin secretion from islet β-cells is driven by glucose metabolism, depolarization of the cell membrane and an influx of calcium, which initiates the release of insulin. Gap junctions composed of connexin36 (Cx36) electrically couple β-cells, regulating calcium signalling and insulin secretion dynamics. Cx36 coupling is decreased in pre-diabetic mice, suggesting a role for altered coupling in diabetes. Our previous work has shown that pro-inflammatory cytokines decrease Cx36 coupling and that compounds which increase cAMP can increase Cx36 coupling. The goal of this study was to determine if exendin-4, which increases cAMP, can protect against cytokine-induced decreases in Cx36 coupling and altered islet function. In both mouse and human islets, exendin-4 protected against cytokine-induced decreases in coupling and preserved glucose-stimulated calcium signalling. Exendin-4 also protected against protein kinase Cδ-mediated decreases in Cx36 coupling. Exendin-4 preserved coupling in mouse islets by preserving Cx36 levels on the plasma membrane. Exendin-4 regulated Cx36 coupling via both protein kinase A (PKA)- and Epac2-mediated mechanisms in cytokine-treated islets. In mouse islets, modulating Epac2 had a greater impact in mediating Cx36 coupling, while in human islets modulating PKA had a greater impact on Cx36 coupling. Our data indicate that PKA regulates Cx36 coupling through a fast mechanism, such as channel gating, while Epac2 regulates slower mechanisms of regulation, such as Cx36 turnover in the membrane. Increases in Cx36 coupling with exendin-4 may protect against cytokine-mediated β-cell dysfunction to insulin secretion dynamics during the development of diabetes.
Collapse
Affiliation(s)
- Nikki L. Farnsworth
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Rachelle Walter
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Robert A. Piscopio
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Wolfgang E. Schleicher
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Richard K. P. Benninger
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| |
Collapse
|
24
|
Turk J, White TD, Nelson AJ, Lei X, Ramanadham S. iPLA 2β and its role in male fertility, neurological disorders, metabolic disorders, and inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:846-860. [PMID: 30408523 DOI: 10.1016/j.bbalip.2018.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023]
Abstract
The Ca2+-independent phospholipases, designated as group VI iPLA2s, also referred to as PNPLAs due to their shared homology with patatin, include the β, γ, δ, ε, ζ, and η forms of the enzyme. The iPLA2s are ubiquitously expressed, share a consensus GXSXG catalytic motif, and exhibit organelle/cell-specific localization. Among the iPLA2s, iPLA2β has received wide attention as it is recognized to be involved in membrane remodeling, cell proliferation, cell death, and signal transduction. Ongoing studies implicate participation of iPLA2β in a variety of disease processes including cancer, cardiovascular abnormalities, glaucoma, and peridonditis. This review will focus on iPLA2β and its links to male fertility, neurological disorders, metabolic disorders, and inflammation.
Collapse
Affiliation(s)
- John Turk
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Tayleur D White
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Alexander J Nelson
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States of America; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| |
Collapse
|
25
|
Hu X, Ge X, Liang W, Shao Y, Jing J, Wang C, Zeng R, Yao B. Effects of saturated palmitic acid and omega-3 polyunsaturated fatty acids on Sertoli cell apoptosis. Syst Biol Reprod Med 2018; 64:368-380. [PMID: 29798686 DOI: 10.1080/19396368.2018.1471554] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is believed to negatively affect male semen quality and is accompanied by dysregulation of free fatty acid (FFA) metabolism in plasma. However, the implication of dysregulated FFA on semen quality and the involvement of Sertoli cells remain unclear. In the present study, we report obesity decreased Sertoli cell viability through dysregulated FFAs. We observed an increased rate of apoptosis in Sertoli cells, accompanied with elevated FFA levels, in the testes of obese mice that were provided a high-fat diet (HFD). Moreover, the levels of reactive oxygen species were elevated. Furthermore, we demonstrated by in vitro assays that saturated palmitic acid (PA), which is the most common saturated FFA in plasma, led to decreased cell viability of TM4 Sertoli cells in a time- and dose-dependent manner. A similar finding was noted in primary mouse Sertoli cells. In contrast to saturated FFA, omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) protected Sertoli cells from PA-induced lipotoxicity at the physiologically relevant levels. These results indicated that the lipotoxicity of saturated fatty acids might be the cause of obesity-induced Sertoli cell apoptosis, which leads to decreased semen quality. In addition, ω-3 PUFAs could be classified as protective FFAs. ABBREVIATIONS FFA: free fatty acid; HFD: high-fat diet; SD: standard diet; PA: palmitic acid; PUFA: polyunsaturated fatty acid; AI: apoptotic index; MTT: 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide; ROS: reactive oxygen species; HE: Hematoxylin and eosin; WT1: Wilm Tumor 1; NAFLD: non- alcoholic fatty liver disease; DCFH-DA: 2', 7' dichlorofluorescin diacetate; 36B4: acidic ribosomal phosphoprotein P0; SD: standard deviation; EPA: eicosapentaenoic acid; PI: propidium iodide; DHA: docosahexenoic acid.
Collapse
Affiliation(s)
- Xuechun Hu
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| | - Xie Ge
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| | - Wei Liang
- b Traditional Chinese Medicine Department , Nanjing No. 454 Hospital , Nanjing , PR China
| | - Yong Shao
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| | - Jun Jing
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| | - Cencen Wang
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| | - Rong Zeng
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| | - Bing Yao
- a Jinling Hospital, School of Medicine , Nanjing University , Nanjing , PR China
| |
Collapse
|
26
|
Velasquez C, Vasquez JS, Balcazar N. In Vitro Effect of Fatty Acids Identified in the Plasma of Obese Adolescents on the Function of Pancreatic β-Cells. Diabetes Metab J 2017; 41:303-315. [PMID: 28868828 PMCID: PMC5583408 DOI: 10.4093/dmj.2017.41.4.303] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/31/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The increase in circulating free fatty acid (FFA) levels is a major factor that induces malfunction in pancreatic β-cells. We evaluated the effect of FFAs reconstituted according to the profile of circulating fatty acids found in obese adolescents on the viability and function of the murine insulinoma cell line (mouse insulinoma [MIN6]). METHODS From fatty acids obtained commercially, plasma-FFA profiles of three different youth populations were reconstituted: obese with metabolic syndrome; obese without metabolic syndrome; and normal weight without metabolic syndrome. MIN6 cells were treated for 24 or 48 hours with the three FFA profiles, and glucose-stimulated insulin secretion, cell viability, mitochondrial function and antioxidant activity were evaluated. RESULTS The high FFA content and high polyunsaturated ω6/ω3 ratio, present in plasma of obese adolescents with metabolic syndrome had a toxic effect on MIN6 cell viability and function, increasing oxidative stress and decreasing glucose-dependent insulin secretion. CONCLUSION These results could help to guide nutritional management of obese young individuals, encouraging the increase of ω-3-rich food consumption in order to reduce the likelihood of deterioration of β-cells and the possible development of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Claudia Velasquez
- Research Group in Food and Human Nutrition, School of Dietetics and Human Nutrition, University of Antioquia, Medellin, Colombia
| | | | - Norman Balcazar
- Genetics Molecular Group, University of Antioquia, Medellin, Colombia
- Department of Physiology and Biochemistry, School of Medicine, University of Antioquia, Medellin, Colombia.
| |
Collapse
|
27
|
Chowdhury AI, Bergsten P. GLP-1 analogue recovers impaired insulin secretion from human islets treated with palmitate via down-regulation of SOCS2. Mol Cell Endocrinol 2017; 439:194-202. [PMID: 27566229 DOI: 10.1016/j.mce.2016.08.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 08/22/2016] [Accepted: 08/22/2016] [Indexed: 12/31/2022]
Abstract
Elevated circulating palmitate levels have been connected with type 2 diabetes mellitus. GLP-1 has favorable effects on beta-cells function. The aim was to identify mechanisms for decreased GSIS after long-term palmitate exposure and restoration by GLP-1 by analyzing changes in G-protein coupled receptor (GPCR) pathway signaling. Insulin secretory response to 20 mM glucose was attenuated after 7 days in islets exposed to palmitate but inclusion of exendin-4 restored secretion. Palmitate treatment altered genes of several GPCR signaling pathways including inflammatory pathways with up-regulated IL-1B, SOCS1 and SOCS2 transcript levels. Protein level of SOCS2 was also up-regulated by palmitate and accompanied by down-regulation of pAkt(T308), which was restored by exendin-4 treatment. When SOCS2 was knocked down, palmitate-induced down-regulation of IRS-1 and pAkt(T308) was prevented and GSIS, proinsulin to insulin ratio and apoptosis was restored. Long-term palmitate treatment up-regulates SOCS2 and reduces PI3K activity, thereby impairing GSIS. GLP-1 reverts the palmitate-induced effects.
Collapse
Affiliation(s)
- Azazul Islam Chowdhury
- Department of Medical Cell Biology, Uppsala University, Box 571, SE-75123, Uppsala, Sweden.
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Box 571, SE-75123, Uppsala, Sweden
| |
Collapse
|
28
|
Kinase Signaling in Apoptosis Induced by Saturated Fatty Acids in Pancreatic β-Cells. Int J Mol Sci 2016; 17:ijms17091400. [PMID: 27626409 PMCID: PMC5037680 DOI: 10.3390/ijms17091400] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β-cell failure and death is considered to be one of the main factors responsible for type 2 diabetes. It is caused by, in addition to hyperglycemia, chronic exposure to increased concentrations of fatty acids, mainly saturated fatty acids. Molecular mechanisms of apoptosis induction by saturated fatty acids in β-cells are not completely clear. It has been proposed that kinase signaling could be involved, particularly, c-Jun N-terminal kinase (JNK), protein kinase C (PKC), p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinase (ERK), and Akt kinases and their pathways. In this review, we discuss these kinases and their signaling pathways with respect to their possible role in apoptosis induction by saturated fatty acids in pancreatic β-cells.
Collapse
|
29
|
Abstract
Type two diabetes (T2D) is a challenging metabolic disorder for which a cure has not yet been found. Its etiology is associated with several phenomena, including significant loss of insulin-producing, beta cell (β cell) mass via progressive programmed cell death and disrupted cellular autophagy. In diabetes, the etiology of β cell death and the role of mitochondria are complex and involve several layers of mechanisms. Understanding the dynamics of those mechanisms could permit researchers to develop an intervention for the progressive loss of β cells. Currently, diabetes research has shifted toward rejuvenation and plasticity technology and away from the simplified approach of hormonal compensation. Diabetes research is currently challenged by questions such as how to enhance cell survival, decrease apoptosis and replenish β cell mass in diabetic patients. In this review, we discuss evidence that β cell development and mass formation are guided by specific signaling systems, particularly hormones, transcription factors, and growth factors, all of which could be manipulated to enhance mass growth. There is also strong evidence that β cells are dynamically active cells, which, under specific conditions such as obesity, can increase in size and subsequently increase insulin secretion. In certain cases of aggressive or advanced forms of T2D, β cells become markedly impaired, and the only alternatives for maintaining glucose homeostasis are through partial or complete cell grafting (the Edmonton protocol). In these cases, the harvesting of an enriched population of viable β cells is required for transplantation. This task necessitates a deep understanding of the pharmacological agents that affect β cell survival, mass, and function. The aim of this review is to initiate discussion about the important signals in pancreatic β cell development and mass formation and to highlight the process by which cell death occurs in diabetes. This review also examines the attempts that have been made to recover or increase cell mass in diabetic patients by using various pharmacological agents.
Collapse
Affiliation(s)
- Husnia I Marrif
- Department of Pharmacology, Faculty of Medicine, University of Benghazi Benghazi, Libya
| | - Salma I Al-Sunousi
- Department of Histology and Anatomy, Faculty of Medicine, University of Benghazi Benghazi, Libya
| |
Collapse
|
30
|
Yoon JS, Moon JS, Kim YW, Won KC, Lee HW. The Glucotoxicity Protecting Effect of Ezetimibe in Pancreatic Beta Cells via Inhibition of CD36. J Korean Med Sci 2016; 31:547-52. [PMID: 27051238 PMCID: PMC4810337 DOI: 10.3346/jkms.2016.31.4.547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/23/2015] [Indexed: 11/20/2022] Open
Abstract
Inhibition of CD36, a fatty acid transporter, has been reported to prevent glucotoxicity and ameliorate high glucose induced beta cell dysfunction. Ezetimibe is a selective cholesterol absorption inhibitor that blocks Niemann Pick C1-like 1 protein, but may exert its effect through suppression of CD36. We attempted to clarify the beneficial effect of ezetimibe on insulin secreting cells and to determine whether this effect is related to change of CD36 expression. mRNA expression of insulin and CD36, intracellular peroxide level and glucose stimulated insulin secretion (GSIS) under normal (5.6 mM) or high glucose (30 mM) condition in INS-1 cells and primary rat islet cells were compared. Changes of the aforementioned factors with treatment with ezetimibe (20 μM) under normal or high glucose condition were also assessed. mRNA expression of insulin was decreased with high glucose, which was reversed by ezetimibe in both INS-1 cells and primary rat islets. CD36 mRNA expression was increased with high glucose, but decreased by ezetimibe in INS-1 cells and primary rat islets. Three-day treatment with high glucose resulted in an increase in intracellular peroxide level; however, it was decreased by treatment with ezetimibe. Decrease in GSIS by three-day treatment with high glucose was reversed by ezetimibe. Palmitate uptake following exposure to high glucose conditions for three days was significantly elevated, which was reversed by ezetimibe in INS-1 cells. Ezetimibe may prevent glucotoxicity in pancreatic β-cells through a decrease in fatty acid influx via inhibition of CD36.
Collapse
Affiliation(s)
- Ji Sung Yoon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Yong-Woon Kim
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyu Chang Won
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Hyoung Woo Lee
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
31
|
Granulocyte colony-stimulating factor (G-CSF): A saturated fatty acid-induced myokine with insulin-desensitizing properties in humans. Mol Metab 2016; 5:305-316. [PMID: 27069870 PMCID: PMC4812007 DOI: 10.1016/j.molmet.2016.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 01/01/2023] Open
Abstract
Objective Circulating long-chain free fatty acids (FFAs) are important metabolic signals that acutely enhance fatty acid oxidation, thermogenesis, energy expenditure, and insulin secretion. However, if chronically elevated, they provoke inflammation, insulin resistance, and β-cell failure. Moreover, FFAs act via multiple signaling pathways as very potent regulators of gene expression. In human skeletal muscle cells differentiated in vitro (myotubes), we have shown in previous studies that the expression of CSF3, the gene encoding granulocyte colony-stimulating factor (G-CSF), is markedly induced upon FFA treatment and exercise. Methods and results We now report that CSF3 is induced in human myotubes by saturated, but not unsaturated, FFAs via Toll-like receptor 4-dependent and -independent pathways including activation of Rel-A, AP-1, C/EBPα, Src, and stress kinases. Furthermore, we show that human adipocytes and myotubes treated with G-CSF become insulin-resistant. In line with this, a functional polymorphism in the CSF3 gene affects adipose tissue- and whole-body insulin sensitivity and glucose tolerance in human subjects with elevated plasma FFA concentrations. Conclusion G-CSF emerges as a new player in FFA-induced insulin resistance and thus may be of interest as a target for prevention and treatment of type 2 diabetes. CSF3, the gene encoding G-CSF, is induced in human myotubes by saturated, but not unsaturated, FFAs. CSF3 expression is induced via Toll-like receptor 4-dependent and -independent pathways. Human adipocytes and myotubes treated with G-CSF become insulin-resistant. A CSF3 SNP affects insulin sensitivity and glucose tolerance in human subjects with elevated plasma FFA concentrations. G-CSF emerges as a new player in FFA-induced insulin resistance.
Collapse
|
32
|
p38 MAPK Is Activated but Does Not Play a Key Role during Apoptosis Induction by Saturated Fatty Acid in Human Pancreatic β-Cells. Int J Mol Sci 2016; 17:159. [PMID: 26861294 PMCID: PMC4783893 DOI: 10.3390/ijms17020159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 12/29/2015] [Accepted: 01/22/2016] [Indexed: 12/25/2022] Open
Abstract
Saturated stearic acid (SA) induces apoptosis in the human pancreatic β-cells NES2Y. However, the molecular mechanisms involved are unclear. We showed that apoptosis-inducing concentrations of SA activate the p38 MAPK signaling pathway in these cells. Therefore, we tested the role of p38 MAPK signaling pathway activation in apoptosis induction by SA in NES2Y cells. Crosstalk between p38 MAPK pathway activation and accompanying ERK pathway inhibition after SA application was also tested. The inhibition of p38 MAPK expression by siRNA silencing resulted in a decrease in MAPKAPK-2 activation after SA application, but it had no significant effect on cell viability or the level of phosphorylated ERK pathway members. The inhibition of p38 MAPK activity by the specific inhibitor SB202190 resulted in inhibition of MAPKAPK-2 activation and noticeable activation of ERK pathway members after SA treatment but in no significant effect on cell viability. p38 MAPK overexpression by plasmid transfection produced an increase in MAPKAPK-2 activation after SA exposure but no significant influence on cell viability or ERK pathway activation. The activation of p38 MAPK by the specific activator anisomycin resulted in significant activation of MAPKAPK-2. Concerning the effect on cell viability, application of the activator led to apoptosis induction similar to application of SA (PARP cleavage and caspase-7, -8, and -9 activation) and in inhibition of ERK pathway members. We demonstrated that apoptosis-inducing concentrations of SA activate the p38 MAPK signaling pathway and that this activation could be involved in apoptosis induction by SA in the human pancreatic β-cells NES2Y. However, this involvement does not seem to play a key role. Crosstalk between p38 MAPK pathway activation and ERK pathway inhibition in NES2Y cells seems likely. Thus, the ERK pathway inhibition by p38 MAPK activation does not also seem to be essential for SA-induced apoptosis.
Collapse
|
33
|
Hozumi Y, Nakano T, Tanaka T, Goto K. Localization of diacylglycerol kinase ζ in rat pancreatic islet cells under normal and streptozotocin-induced stress conditions . ACTA ACUST UNITED AC 2016. [DOI: 10.1679/aohc.76.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yasukazu Hozumi
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine,
| | - Tomoyuki Nakano
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine,
| | - Toshiaki Tanaka
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine,
| | - Kaoru Goto
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine,
| |
Collapse
|
34
|
Gerst F, Kaiser G, Panse M, Sartorius T, Pujol A, Hennige AM, Machicao F, Lammers R, Bosch F, Häring HU, Ullrich S. Protein kinase Cδ regulates nuclear export of FOXO1 through phosphorylation of the chaperone 14-3-3ζ. Diabetologia 2015; 58:2819-31. [PMID: 26363783 DOI: 10.1007/s00125-015-3744-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/03/2015] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Forkhead box protein O1 (FOXO1) is a transcription factor essential for beta cell fate. Protein kinase B-dependent phosphorylation of FOXO1 at S256 (P-FOXO1) enables its binding to 14-3-3 dimers and nuclear export. Dephosphorylated FOXO1 enters nuclei and activates pro-apoptotic genes. Since our previous observations suggest that protein kinase C delta (PKCδ) induces nuclear accumulation of FOXO1, the underlying mechanism was examined. METHODS In human islets, genetically modified mice and INS-1E cells apoptosis was assessed by TUNEL staining. Subcellular translocation of proteins was examined by confocal microscopy and signalling pathways were analysed by western blotting and overlay assay. RESULTS In PKCδ-overexpressing (PKCδ-tg) mouse islet cells and INS-1E cells FOXO1 accumulated in nuclei, surprisingly, as P-FOXO1. PKCδ-tg decelerated IGF-1-dependent stimulation of nuclear export, indicating that changes in export caused nuclear retention of P-FOXO1. Nuclear accumulation of P-FOXO1 was accompanied by increased phosphorylation of 14-3-3ζ at S58 and reduced dimerisation of 14-3-3ζ. Palmitic acid further augmented phosphorylation of 14-3-3ζ and triggered nuclear accumulation of FOXO1 in both INS-1E and human islet cells. Furthermore, the overexpression of a phosphomimicking mutant of 14-3-3ζ (S58D) enhanced nuclear FOXO1. In accordance with the nuclear accumulation of P-FOXO1, PKCδ overexpression alone did not increase apoptotic cell death. Additionally, insulin secretion and glucose homeostasis in PKCδ-overexpressing mice remained unaffected. CONCLUSIONS/INTERPRETATION These results suggest that PKCδ-mediated phosphorylation of 14-3-3ζ contributes to the nuclear retention of FOXO1, even when FOXO1 is phosphorylated as under non-stress conditions. P-FOXO1 does not induce pro-apoptotic genes, but may rather exert beneficial effects on beta cells.
Collapse
Affiliation(s)
- Felicia Gerst
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Gabriele Kaiser
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Madhura Panse
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Tina Sartorius
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Anna Pujol
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Anita M Hennige
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Fausto Machicao
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Reiner Lammers
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Susanne Ullrich
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, D-72076, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Partner in the German Center for Diabetes Research (DZD), Tübingen, Germany.
| |
Collapse
|
35
|
Lu S, Mott JL, Harrison-Findik DD. Saturated fatty acids induce post-transcriptional regulation of HAMP mRNA via AU-rich element-binding protein, human antigen R (HuR). J Biol Chem 2015; 290:24178-89. [PMID: 26304124 DOI: 10.1074/jbc.m115.648212] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 12/11/2022] Open
Abstract
Iron is implicated in fatty liver disease pathogenesis. The human hepcidin gene, HAMP, is the master switch of iron metabolism. The aim of this study is to investigate the regulation of HAMP expression by fatty acids in HepG2 cells. For these studies, both saturated fatty acids (palmitic acid (PA) and stearic acid (SA)) and unsaturated fatty acid (oleic acid (OA)) were used. PA and, to a lesser extent, SA, but not OA, up-regulated HAMP mRNA levels, as determined by real-time PCR. To understand whether PA regulates HAMP mRNA at the transcriptional or post-transcriptional level, the transcription inhibitor actinomycin D was employed. PA-mediated induction of HAMP mRNA expression was not blocked by actinomycin D. Furthermore, PA activated HAMP 3'-UTR, but not promoter, activity, as shown by reporter assays. HAMP 3'-UTR harbors a single AU-rich element (ARE). Mutation of this ARE abolished the effect of PA, suggesting the involvement of ARE-binding proteins. The ARE-binding protein human antigen R (HuR) stabilizes mRNA through direct interaction with AREs on 3'-UTR. HuR is regulated by phosphorylation-mediated nucleo-cytoplasmic shuttling. PA activated this process. The binding of HuR to HAMP mRNA was also induced by PA in HepG2 cells. Silencing of HuR by siRNA abolished PA-mediated up-regulation of HAMP mRNA levels. PKC is known to phosphorylate HuR. Staurosporine, a broad-spectrum PKC inhibitor, inhibited both PA-mediated translocation of HuR and induction of HAMP expression. Similarly, rottlerin, a novel class PKC inhibitor, abrogated PA-mediated up-regulation of HAMP expression. In conclusion, lipids mediate post-transcriptional regulation of HAMP throughPKC- and HuR-dependent mechanisms.
Collapse
Affiliation(s)
- Sizhao Lu
- From the Department of Biochemistry and Molecular Biology and
| | - Justin L Mott
- From the Department of Biochemistry and Molecular Biology and
| | - Duygu Dee Harrison-Findik
- the Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198-5820
| |
Collapse
|
36
|
Caputa G, Zhao S, Criado AEG, Ory DS, Duncan JG, Schaffer JE. RNASET2 is required for ROS propagation during oxidative stress-mediated cell death. Cell Death Differ 2015. [PMID: 26206090 DOI: 10.1038/cdd.2015.105] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
RNASET2 is a ubiquitously expressed acidic ribonuclease that has been implicated in diverse pathophysiological processes including tumorigeneis, vitiligo, asthenozoospermia, and neurodegeneration. Prior studies indicate that RNASET2 is induced in response to oxidative stress and that overexpression of RNASET2 sensitizes cells to reactive oxygen species (ROS)-induced cell death through a mechanism that is independent of catalytic activity. Herein, we report a loss-of-function genetic screen that identified RNASET2 as an essential gene for lipotoxic cell death. Haploinsufficiency of RNASET2 confers increased antioxidant capacity and generalized resistance to oxidative stress-mediated cell death in cultured cells. This function is critically dependent on catalytic activity. Furthermore, knockdown of RNASET2 in the Drosophila fat body confers increased survival in the setting of oxidative stress inducers. Together, these findings demonstrate that RNASET2 regulates antioxidant tone and is required for physiological ROS responses.
Collapse
Affiliation(s)
- G Caputa
- Diabetic Cardiovascular Disease Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - S Zhao
- Diabetic Cardiovascular Disease Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - A E G Criado
- Diabetic Cardiovascular Disease Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - D S Ory
- Diabetic Cardiovascular Disease Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - J G Duncan
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - J E Schaffer
- Diabetic Cardiovascular Disease Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
37
|
Natalicchio A, Tortosa F, Labarbuta R, Biondi G, Marrano N, Carchia E, Leonardini A, Cignarelli A, Bugliani M, Marchetti P, Fadini GP, Giorgio M, Avogaro A, Perrini S, Laviola L, Giorgino F. The p66(Shc) redox adaptor protein is induced by saturated fatty acids and mediates lipotoxicity-induced apoptosis in pancreatic beta cells. Diabetologia 2015; 58:1260-71. [PMID: 25810038 DOI: 10.1007/s00125-015-3563-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/27/2015] [Indexed: 01/03/2023]
Abstract
AIMS/HYPOTHESIS The role of the redox adaptor protein p66(Shc) as a potential mediator of saturated fatty acid (FA)-induced beta cell death was investigated. METHODS The effects of the FA palmitate on p66(Shc) expression were evaluated in human and murine islets and in rat insulin-secreting INS-1E cells. p66(Shc) expression was also measured in islets from mice fed a high-fat diet (HFD) and from human donors with different BMIs. Cell apoptosis was quantified by two independent assays. The role of p66(Shc) was investigated using pancreatic islets from p66 (Shc-/-) mice and in INS-1E cells with knockdown of p66(Shc) or overexpression of wild-type and phosphorylation-defective p66(Shc). Production of reactive oxygen species (ROS) was evaluated by the dihydroethidium oxidation method. RESULTS Palmitate induced a selective increase in p66(Shc) protein expression and phosphorylation on Ser(36) and augmented apoptosis in human and mouse islets and in INS-1E cells. Inhibiting the tumour suppressor protein p53 prevented both the palmitate-induced increase in p66(Shc) expression and beta cell apoptosis. Palmitate-induced apoptosis was abrogated in islets from p66 (Shc-/-) mice and following p66 (Shc) knockdown in INS-1E cells; by contrast, overexpression of p66(Shc), but not that of the phosphorylation-defective p66(Shc) mutant, enhanced palmitate-induced apoptosis. The pro-apoptotic effects of p66(Shc) were dependent upon its c-Jun N-terminal kinase-mediated phosphorylation on Ser(36) and associated with generation of ROS. p66(Shc) protein expression and function were also elevated in islets from HFD-fed mice and from obese/overweight cadaveric human donors. CONCLUSIONS/INTERPRETATION p53-dependent augmentation of p66(Shc) expression and function represents a key signalling response contributing to beta cell apoptosis under conditions of lipotoxicity.
Collapse
Affiliation(s)
- Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sartorius T, Häring HU. The Metabolic Role of Saturated and Monounsaturated Dietary Fatty Acids. MODULATION OF SLEEP BY OBESITY, DIABETES, AGE, AND DIET 2015:203-210. [DOI: 10.1016/b978-0-12-420168-2.00022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
39
|
Kowluru A. On a sugary-relationship between caspases and lamins. Cell Cycle 2014; 13:3787-8. [PMID: 25470565 DOI: 10.4161/15384101.2014.989948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Anjaneyulu Kowluru
- a Beta-Cell Biochemistry Laboratory; John D. Dingell VA Medical Center; Department of Pharmaceutical Sciences ; Wayne State University ; Detroit , MI USA
| |
Collapse
|
40
|
Gorgani-Firuzjaee S, Ahmadi S, Meshkani R. Palmitate induces SHIP2 expression via the ceramide-mediated activation of NF-κB, and JNK in skeletal muscle cells. Biochem Biophys Res Commun 2014; 450:494-9. [DOI: 10.1016/j.bbrc.2014.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 06/01/2014] [Indexed: 11/17/2022]
|
41
|
Abstract
Over 200 million people worldwide suffer from diabetes, a disorder of glucose homeostasis. The majority of these individuals are diagnosed with type 2 diabetes. It has traditionally been thought that tissue resistance to the action of insulin is the primary defect in type 2 diabetes. However, recent longitudinal and genome‐wide association studies have shown that insulin resistance is more likely to be a precondition, and that the failure of the pancreatic β cell to meet the increased insulin requirements is the triggering factor in the development of type 2 diabetes. A major emphasis in diabetes research has therefore shifted to understanding the causes of β cell failure. Collectively, these studies have implicated a complex network of triggers, which activate intersecting execution pathways leading to β cell dysfunction and death. In the present review, we discuss these triggers (glucotoxicity, lipotoxicity, amyloid and cytokines) with respect to the pathways they activate (oxidative stress, inflammation and endoplasmic reticulum stress) and propose a model for understanding β cell failure in type 2 diabetes. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00021.x, 2010)
Collapse
Affiliation(s)
- Takeshi Ogihara
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research
| | - Raghavendra G Mirmira
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research ; Departments of Medicine and Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
42
|
Chen KC, Yang TY, Wu CC, Cheng CC, Hsu SL, Hung HW, Chen JW, Chang GC. Pemetrexed induces S-phase arrest and apoptosis via a deregulated activation of Akt signaling pathway. PLoS One 2014; 9:e97888. [PMID: 24847863 PMCID: PMC4029963 DOI: 10.1371/journal.pone.0097888] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/25/2014] [Indexed: 12/16/2022] Open
Abstract
Pemetrexed is approved for first-line and maintenance treatment of patients with advanced or metastatic non-small-cell lung cancer (NSCLC). The protein kinase Akt/protein kinase B is a well-known regulator of cell survival which is activated by pemetrexed, but its role in pemetrexed-mediated cell death and its molecular mechanisms are unclear. This study showed that stimulation with pemetrexed induced S-phase arrest and cell apoptosis and a parallel increase in sustained Akt phosphorylation and nuclear accumulation in the NSCLC A549 cell line. Inhibition of Akt expression by Akt specific siRNA blocked S-phase arrest and protected cells from apoptosis, indicating an unexpected proapoptotic role of Akt in the pemetrexed-mediated toxicity. Treatment of A549 cells with pharmacological inhibitors of phosphatidylinositol 3-kinase (PI3K), wortmannin and Ly294002, similarly inhibited pemetrexed-induced S-phase arrest and apoptosis and Akt phosphorylation, indicating that PI3K is an upstream mediator of Akt and is involved in pemetrexed-mediated cell death. Previously, we identified cyclin A-associated cyclin-dependent kinase 2 (Cdk2) as the principal kinase that was required for pemetrexed-induced S-phase arrest and apoptosis. The current study showed that inhibition of Akt function and expression by pharmacological inhibitors as well as Akt siRNA drastically inhibited cyclin A/Cdk2 activation. These pemetrexed-mediated biological and molecular events were also observed in a H1299 cell line. Overall, our results indicate that, in contrast to its normal prosurvival role, the activated Akt plays a proapoptotic role in pemetrexed-mediated S-phase arrest and cell death through a mechanism that involves Cdk2/cyclin A activation.
Collapse
Affiliation(s)
- Kun-Chieh Chen
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Tsung-Ying Yang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Chun-Chi Wu
- Institute of Medicine, Chung Shang Medical University, Taichung, Taiwan, Republic of China
- Department of Medical Research, Chung-Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Chi-Chih Cheng
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Shih-Lan Hsu
- Department of Medical Research, Chung-Shan Medical University Hospital, Taichung, Taiwan, Republic of China
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Hsiao-Wen Hung
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Jian-Wei Chen
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Gee-Chen Chang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| |
Collapse
|
43
|
Qin J, Fang N, Lou J, Zhang W, Xu S, Liu H, Fang Q, Wang Z, Liu J, Men X, Peng L, Chen L. TRB3 is involved in free fatty acid-induced INS-1-derived cell apoptosis via the protein kinase C δ pathway. PLoS One 2014; 9:e96089. [PMID: 24824999 PMCID: PMC4019472 DOI: 10.1371/journal.pone.0096089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/02/2014] [Indexed: 12/16/2022] Open
Abstract
Chronic exposure to free fatty acids (FFAs) may induce β cell apoptosis in type 2 diabetes. However, the precise mechanism by which FFAs trigger β cell apoptosis is still unclear. Tribbles homolog 3 (TRB3) is a pseudokinase inhibiting Akt, a key mediator of insulin signaling, and contributes to insulin resistance in insulin target tissues. This paper outlined the role of TRB3 in FFAs-induced INS-1 β cell apoptosis. TRB3 was promptly induced in INS-1 cells after stimulation by FFAs, and this was accompanied by enhanced INS-1 cell apoptosis. The overexpression of TRB3 led to exacerbated apoptosis triggered by FFAs in INS-1-derived cell line and the subrenal capsular transplantation animal model. In contrast, cell apoptosis induced by FFAs was attenuated when TRB3 was knocked down. Moreover, we observed that activation and nuclear accumulation of protein kinase C (PKC) δ was enhanced by upregulation of TRB3. Preventing PKCδ nuclear translocation and PKCδ selective antagonist both significantly lessened the pro-apoptotic effect. These findings suggest that TRB3 was involved in lipoapoptosis of INS-1 β cell, and thus could be an attractive pharmacological target in the prevention and treatment of T2DM.
Collapse
Affiliation(s)
- Jun Qin
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ni Fang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jinning Lou
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Wenjian Zhang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Shiqing Xu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Honglin Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Qing Fang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jiang Liu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Xiuli Men
- Department of Pathophysiology, North China Coal Medical University, Tangshan, China
| | - Liang Peng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
44
|
Lee JH, Jung IR, Choi SE, Lee SM, Lee SJ, Han SJ, Kim HJ, Kim DJ, Lee KW, Kang Y. Toxicity generated through inhibition of pyruvate carboxylase and carnitine palmitoyl transferase-1 is similar to high glucose/palmitate-induced glucolipotoxicity in INS-1 beta cells. Mol Cell Endocrinol 2014; 383:48-59. [PMID: 24333689 DOI: 10.1016/j.mce.2013.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 11/14/2013] [Accepted: 12/01/2013] [Indexed: 01/22/2023]
Abstract
This work was initiated to determine whether toxicity generated through inhibition of mitochondrial fuel metabolism is similar to high glucose/palmitate (HG/PA)-induced glucolipotoxicity. Influx of glucose and free fatty acids into the tricarboxylic acid (TCA) cycle was inhibited by treatment with the pyruvate carboxylase (PC) inhibitor phenylacetic acid (PAA) and carnitine palmitoyl transferase-1 (CPT-1) inhibitor etomoxir (Eto), or knockdown of PC and CPT-1. Treatment of PAA/Eto or knockdown of PC/CPT-1 induced apoptotic death in INS-1 beta cells. Similar to HG/PA treatment, PAA/Eto increased endoplasmic reticulum stress responses but decreased the Akt signal. JNK inhibitor or chemical chaperone was protective against both PAA/Eto- and HG/PA-induced cell death. All attempts to reduce [Ca²⁺](i), stimulate lipid metabolism, and increase the TCA cycle intermediate pool protected PAA/Eto-induced death as well as HG/PA-induced death. These data suggest that signals induced from impaired mitochondrial fuel metabolism play a critical role in HG/PA-induced glucolipotoxicity.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Life Science, Korea University Seoul 136-701, Republic of Korea
| | - Ik-Rak Jung
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-E Choi
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-Mi Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Soo-Jin Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Kwan-Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Yup Kang
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea.
| |
Collapse
|
45
|
Wagner R, Staiger H, Ullrich S, Stefan N, Fritsche A, Häring HU. Untangling the interplay of genetic and metabolic influences on beta-cell function: Examples of potential therapeutic implications involving TCF7L2 and FFAR1. Mol Metab 2014; 3:261-7. [PMID: 24749055 PMCID: PMC3986492 DOI: 10.1016/j.molmet.2014.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 01/07/2014] [Accepted: 01/07/2014] [Indexed: 12/24/2022] Open
Abstract
Deteriorating beta-cell function is a common feature of type 2 diabetes. In this review, we briefly address the regulation of beta-cell function, and discuss some of the main determinants of beta-cell failure. We will focus on the role of interactions between the genetic background and metabolic environment (insulin resistance, fuel supply and flux as well as metabolic signaling). We present data on the function of the strongest common diabetes risk variant, the single nucleotide polymorphism (SNP) rs7903146 in TCF7L2. As also mirrored by its interaction with glycemia on insulin secretion, this SNP in large part confers resistance against the incretin effect. Genetic influence on insulin secretion also interacts with free fatty acids, as evidenced by data on rs1573611 in FFAR1. Several medications marketed by now or currently under development for diabetes treatment engage these pathways, and therapeutic implications from these findings are soon to be expected.
Collapse
Affiliation(s)
- Robert Wagner
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University Hospital of the Eberhard Karls University, Tübingen, Germany ; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Harald Staiger
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University Hospital of the Eberhard Karls University, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich at the University of Tübingen (IDM), Tübingen, Germany ; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Susanne Ullrich
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University Hospital of the Eberhard Karls University, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich at the University of Tübingen (IDM), Tübingen, Germany ; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Norbert Stefan
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University Hospital of the Eberhard Karls University, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich at the University of Tübingen (IDM), Tübingen, Germany ; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andreas Fritsche
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University Hospital of the Eberhard Karls University, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich at the University of Tübingen (IDM), Tübingen, Germany ; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, University Hospital of the Eberhard Karls University, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich at the University of Tübingen (IDM), Tübingen, Germany ; German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
46
|
Kurohane Kaneko Y, Kobayashi Y, Motoki K, Nakata K, Miyagawa S, Yamamoto M, Hayashi D, Shirai Y, Sakane F, Ishikawa T. Depression of type I diacylglycerol kinases in pancreatic β-cells from male mice results in impaired insulin secretion. Endocrinology 2013; 154:4089-98. [PMID: 24035999 DOI: 10.1210/en.2013-1356] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Diacylglycerol kinase (DGK) catalyzes the conversion of diacylglycerol (DAG) to phosphatidic acid. This study investigated the expression and function of DGK in pancreatic β-cells. mRNA expression of type I DGK isoforms (α, β, γ) was detected in mouse pancreatic islets and the β-cell line MIN6. Protein expression of DGKα and DGKγ was also detected in mouse β-cells and MIN6 cells. The type I DGK inhibitor R59949 inhibited high K(+)- and glucose-induced insulin secretion in MIN6 cells. Moreover, single knockdown of DGKα or DGKγ by small interfering RNA slightly but significantly decreased glucose- and high K(+)-induced insulin secretions, and the double knockdown further decreased them to the levels comparable with those induced by R59949. R59949 and DiC8, a membrane permeable DAG analog, decreased intracellular Ca(2+) concentration elevated by glucose and high K(+) in MIN6 cells. Real-time imaging in MIN6 cells expressing green fluorescent protein-tagged DGKα or DGKγ showed that the DGK activator phorbol 12-myristate 13-acetate rapidly induced translocation of DGKγ to the plasma membrane, whereas high K(+) slowly translocated DGKα and DGKγ to the plasma membrane. R59949 increased the DAG content in MIN6 cells when stimulated with high KCl, whereas it did not increase the DAG content but decreased the phosphatidic acid content when stimulated with high glucose. Finally, R59949 was confirmed to inhibit high K(+)-induced insulin secretion from mouse islets and glucose-induced insulin secretion from rat islets. These results suggest that DGKα and DGKγ are present in β-cells and that the depression of these DGKs causes a decrease in intracellular Ca(2+) concentration, thereby reducing insulin secretion.
Collapse
|
47
|
Natalicchio A, Labarbuta R, Tortosa F, Biondi G, Marrano N, Peschechera A, Carchia E, Orlando MR, Leonardini A, Cignarelli A, Marchetti P, Perrini S, Laviola L, Giorgino F. Exendin-4 protects pancreatic beta cells from palmitate-induced apoptosis by interfering with GPR40 and the MKK4/7 stress kinase signalling pathway. Diabetologia 2013; 56:2456-66. [PMID: 23995397 DOI: 10.1007/s00125-013-3028-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/29/2013] [Indexed: 12/24/2022]
Abstract
AIMS/HYPOTHESIS The mechanisms of the protective effects of exendin-4 on NEFA-induced beta cell apoptosis were investigated. METHODS The effects of exendin-4 and palmitate were evaluated in human and murine islets, rat insulin-secreting INS-1E cells and murine glucagon-secreting alpha-TC1-6 cells. mRNA and protein expression/phosphorylation were measured by real-time RT-PCR and immunoblotting or immunofluorescence, respectively. Small interfering (si)RNAs for Ib1 and Gpr40 were used. Cell apoptosis was quantified by two independent assays. Insulin release was assessed with an insulin ELISA. RESULTS Exposure of human and murine primary islets and INS-1E cells, but not alpha-TC1-6 cells, to exendin-4 inhibited phosphorylation of the stress kinases, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK), and prevented apoptosis in response to palmitate. Exendin-4 increased the protein content of islet-brain 1 (IB1), an endogenous JNK blocker; however, siRNA-mediated reduction of IB1 did not impair the ability of exendin-4 to inhibit JNK and prevent apoptosis. Exendin-4 reduced G-protein-coupled receptor 40 (GPR40) expression and inhibited palmitate-induced phosphorylation of mitogen-activated kinase kinase (MKK)4 and MKK7. The effects of exendin-4 were abrogated in the presence of the protein kinase A (PKA) inhibitors, H89 and KT5720. Knockdown of GPR40, as well as use of a specific GPR40 antagonist, resulted in diminished palmitate-induced JNK and p38 MAPK phosphorylation and apoptosis. Furthermore, inhibition of JNK and p38 MAPK activity prevented palmitate-induced apoptosis. CONCLUSIONS/INTERPRETATION Exendin-4 counteracts the proapoptotic effects of palmitate in beta cells by reducing GPR40 expression and inhibiting MKK7- and MKK4-dependent phosphorylation of the stress kinases, JNK and p38 MAPK, in a PKA-dependent manner.
Collapse
Affiliation(s)
- Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhu Y, You W, Wang H, Li Y, Qiao N, Shi Y, Zhang C, Bleich D, Han X. MicroRNA-24/MODY gene regulatory pathway mediates pancreatic β-cell dysfunction. Diabetes 2013; 62:3194-206. [PMID: 23761103 PMCID: PMC3749364 DOI: 10.2337/db13-0151] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Overnutrition and genetics both contribute separately to pancreatic β-cell dysfunction, but how these factors interact is unclear. This study was aimed at determining whether microRNAs (miRNAs) provide a link between these factors. In this study, miRNA-24 (miR-24) was highly expressed in pancreatic β-cells and further upregulated in islets from genetic fatty (db/db) or mice fed a high-fat diet, and islets subject to oxidative stress. Overexpression of miR-24 inhibited insulin secretion and β-cell proliferation, potentially involving 351 downregulated genes. By using bioinformatic analysis combined with luciferase-based promoter activity assays and quantitative real-time PCR assays, we identified two maturity-onset diabetes of the young (MODY) genes as direct targets of miR-24. Silencing either of these MODY genes (Hnf1a and Neurod1) mimicked the cellular phenotype caused by miR-24 overexpression, whereas restoring their expression rescued β-cell function. Our findings functionally link the miR-24/MODY gene regulatory pathway to the onset of type 2 diabetes and create a novel network between nutrient overload and genetic diabetes via miR-24.
Collapse
Affiliation(s)
- Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Center, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Weiyan You
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Center, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Hongdong Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Center, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Center, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Nan Qiao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Center, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yuguang Shi
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Chenyu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - David Bleich
- University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Jiangsu Diabetes Center, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
- Corresponding author: Xiao Han,
| |
Collapse
|
49
|
Rué L, Alcalá-Vida R, López-Soop G, Creus-Muncunill J, Alberch J, Pérez-Navarro E. Early down-regulation of PKCδ as a pro-survival mechanism in Huntington's disease. Neuromolecular Med 2013; 16:25-37. [PMID: 23896721 DOI: 10.1007/s12017-013-8248-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 07/12/2013] [Indexed: 11/29/2022]
Abstract
A balance between cell survival and apoptosis is crucial to avoid neurodegeneration. Here, we analyzed whether the pro-apoptotic protein PKCδ, and the pro-survival PKCα and βII, were dysregulated in the brain of R6/1 mouse model of Huntington's disease (HD). Protein levels of the three PKCs examined were reduced in all the brain regions analyzed being PKCδ the most affected isoform. Interestingly, PKCδ protein levels were also decreased in the striatum and cortex of R6/2 and Hdh(Q111/Q111) mice, and in the putamen of HD patients. Nuclear PKCδ induces apoptosis, but we detected reduced PKCδ in both cytoplasmic and nuclear enriched fractions from R6/1 mouse striatum, cortex and hippocampus. In addition, we show that phosphorylation and ubiquitination of PKCδ are increased in 30-week-old R6/1 mouse brain. All together these results suggest a pro-survival role of reduced PKCδ levels in response to mutant huntingtin-induced toxicity. In fact, we show that over-expression of PKCδ increases mutant huntingtin-induced cell death in vitro, whereas over-expression of a PKCδ dominant negative form or silencing of endogenous PKCδ partially blocks mutant huntingtin-induced cell death. Finally, we show that the analysis of lamin B protein levels could be a good marker of PKCδ activity, but it is not involved in PKCδ-mediated cell death in mutant huntingtin-expressing cells. In conclusion, our results suggest that neurons increase the degradation of PKCδ as a compensatory pro-survival mechanism in response to mutant huntingtin-induced toxicity that can help to understand why cell death appears late in the disease.
Collapse
Affiliation(s)
- Laura Rué
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Casanova 143, Barcelona, 08036, Spain
| | | | | | | | | | | |
Collapse
|
50
|
Yang F, Cai W, Yang K, Chen M. PKCδ knockdown inhibits free fatty acid induction of endothelial cell apoptosis. Cell Biochem Funct 2012; 31:380-4. [PMID: 23086745 DOI: 10.1002/cbf.2908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 08/16/2012] [Accepted: 09/10/2012] [Indexed: 11/11/2022]
Abstract
The mechanisms whereby free fatty acids induce endothelial cell apoptosis are not yet understood. The present study aimed to investigate the role of PKCδ in free fatty acid-induced endothelial cell apoptosis. In addition, we looked for evidence of apoptosis-related interactions between PKCδ and Fas signal pathway. Human umbilical vein endothelial cells were treated with various concentrations of free fatty acids and transiently transfected with PKCδ siRNA or Fas siRNA to inhibit PKCδ or Fas expression. Cell proliferation was determined through colorimetric assays, and apoptosis was quantified using flow cytometry. Protein expression was determined from cell lysates using Western blots with antibodies against p-PKCδTyr512, PKCδ, and Fas. Statistical analyses were performed. Free fatty acids had multiple effects on human umbilical vein endothelial cells, including concentration-dependent inhibition of cell proliferation, induction of apoptosis, increased Fas expression, and increased PKCδ expression and phosphorylation. Inhibition of PKCδ mRNA expression by PKCδ siRNA led to a reduction in both free fatty acid-induced apoptosis and Fas expression. However, Fas siRNA treatment inhibited Fas, but not PKCδ, expression in human umbilical vein endothelial cells. The free fatty acid-induced apoptosis in endothelial cells are possibly mediated by PKCδ and may involve upregulation of its downstream Fas.
Collapse
Affiliation(s)
- Feiyan Yang
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, Hubei Province, China, 430014
| | | | | | | |
Collapse
|