51
|
Mustapha S, Mohammed M, Azemi AK, Yunusa I, Shehu A, Mustapha L, Wada Y, Ahmad MH, Ahmad WANW, Rasool AHG, Mokhtar SS. Potential Roles of Endoplasmic Reticulum Stress and Cellular Proteins Implicated in Diabesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8830880. [PMID: 33995826 PMCID: PMC8099518 DOI: 10.1155/2021/8830880] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
The role of the endoplasmic reticulum (ER) has evolved from protein synthesis, processing, and other secretory pathways to forming a foundation for lipid biosynthesis and other metabolic functions. Maintaining ER homeostasis is essential for normal cellular function and survival. An imbalance in the ER implied stressful conditions such as metabolic distress, which activates a protective process called unfolded protein response (UPR). This response is activated through some canonical branches of ER stress, i.e., the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6). Therefore, chronic hyperglycemia, hyperinsulinemia, increased proinflammatory cytokines, and free fatty acids (FFAs) found in diabesity (a pathophysiological link between obesity and diabetes) could lead to ER stress. However, limited data exist regarding ER stress and its association with diabesity, particularly the implicated proteins and molecular mechanisms. Thus, this review highlights the role of ER stress in relation to some proteins involved in diabesity pathogenesis and provides insight into possible pathways that could serve as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Pulau Pinang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, College of Pharmacy, Columbia, SC, USA
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna, Nigeria
| | - Yusuf Wada
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mubarak Hussaini Ahmad
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
- School of Pharmacy Technician, Aminu Dabo College of Health Sciences and Technology, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
52
|
Abstract
Improved stem cell-derived pancreatic islet (SC-islet) differentiation protocols robustly generate insulin-secreting β cells from patient induced pluripotent stem cells (iPSCs). These advances are enabling in vitro disease modeling studies and the development of an autologous diabetes cell replacement therapy. SC-islet technology elucidates key features of human pancreas development and diabetes disease progression through the generation of pancreatic progenitors, endocrine progenitors, and β cells derived from diabetic and nondiabetic iPSCs. Combining disease modeling with gene editing and next-generation sequencing reveals the impact of diabetes-causing mutations and diabetic phenotypes on multiple islet cell types. In addition, the supply of SC-islets, containing β and other islet cell types, is unlimited, presenting an opportunity for personalized medicine and overcoming several disadvantages posed by donor islets. This review highlights relevant studies involving iPSC-β cells and progenitors, encompassing new conclusions involving cells from patients with diabetes and the therapeutic potential of iPSC-β cells. Improved differentiation protocols generate pancreatic islet from patient stem cells Diabetic stem cell-derived islet studies identified key markers for cell function Gene editing aims to address unmet needs for stem cell therapy field Stem cell-derived islets are a promising source for diabetes stem cell therapy
Collapse
|
53
|
Vig S, Lambooij JM, Zaldumbide A, Guigas B. Endoplasmic Reticulum-Mitochondria Crosstalk and Beta-Cell Destruction in Type 1 Diabetes. Front Immunol 2021; 12:669492. [PMID: 33936111 PMCID: PMC8085402 DOI: 10.3389/fimmu.2021.669492] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Beta-cell destruction in type 1 diabetes (T1D) results from the combined effect of inflammation and recurrent autoimmunity. In response to inflammatory signals, beta-cells engage adaptive mechanisms where the endoplasmic reticulum (ER) and mitochondria act in concert to restore cellular homeostasis. In the recent years it has become clear that this adaptive phase may trigger the development of autoimmunity by the generation of autoantigens recognized by autoreactive CD8 T cells. The participation of the ER stress and the unfolded protein response to the increased visibility of beta-cells to the immune system has been largely described. However, the role of the other cellular organelles, and in particular the mitochondria that are central mediator for beta-cell survival and function, remains poorly investigated. In this review we will dissect the crosstalk between the ER and mitochondria in the context of T1D, highlighting the key role played by this interaction in beta-cell dysfunctions and immune activation, especially through regulation of calcium homeostasis, oxidative stress and generation of mitochondrial-derived factors.
Collapse
Affiliation(s)
- Saurabh Vig
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Joost M. Lambooij
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
54
|
Esakov E, Nandedkar-Kulkarni N, Al-Dieri AG, Hafner H, Gregg B, McInerney MF. Islet Dysfunction in a Novel Transgenic Model of T Cell Insulitis. Biomolecules 2021; 11:biom11040552. [PMID: 33918805 PMCID: PMC8070091 DOI: 10.3390/biom11040552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 11/25/2022] Open
Abstract
The newly established CD3FLAG-mIR transgenic mouse model on a C57Bl/6 background has a FLAG tag on the mouse Insulin Receptor (mIR), specifically on T cells, as the FLAG-tagged mIR gene was engineered behind CD3 promoter and enhancer. The IR is a chemotactic molecule for insulin and the Flag-tagged mIR T cells in the BL/6-CD3FLAGmIR transgenic mice can migrate into the pancreas, as shown by immunofluorescent staining. While the transgenic mice do not become diabetic, there are phenotypic and metabolic changes in the islets. The transgenic islets become enlarged and disorganized by 15 weeks and those phenotypes continue out to 35 weeks of age. We examined the islets by RT-PCR for cell markers, ER stress markers, beta cell proliferation markers, and cytokines, as well as measuring serum insulin and insulin content in the pancreas at 15, 25, and 35 weeks of age. In transgenic mice, insulin in serum was increased at 15 weeks of age and glucose intolerance developed by 25 weeks of age. Passage of transgenic spleen cells into C57Bl/6 RAG−/− mice resulted in enlarged and disorganized islets with T infiltration by 4 to 5 weeks post-transfer, replicating the transgenic mouse studies. Therefore, migration of non-antigen-specific T cells into islets has ramifications for islet organization and function.
Collapse
Affiliation(s)
- Emily Esakov
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
| | - Neha Nandedkar-Kulkarni
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
| | - Ali G. Al-Dieri
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
| | - Hannah Hafner
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (H.H.); (B.G.)
| | - Brigid Gregg
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (H.H.); (B.G.)
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcia F. McInerney
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
- Correspondence: ; Tel.: +1-419-517-3638
| |
Collapse
|
55
|
Honzawa N, Fujimoto K. The Plasticity of Pancreatic β-Cells. Metabolites 2021; 11:metabo11040218. [PMID: 33918379 PMCID: PMC8065544 DOI: 10.3390/metabo11040218] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes is caused by impaired insulin secretion and/or insulin resistance. Loss of pancreatic β-cell mass detected in human diabetic patients has been considered to be a major cause of impaired insulin secretion. Additionally, apoptosis is found in pancreatic β-cells; β-cell mass loss is induced when cell death exceeds proliferation. Recently, however, β-cell dedifferentiation to pancreatic endocrine progenitor cells and β-cell transdifferentiation to α-cell was reported in human islets, which led to a new underlying molecular mechanism. Hyperglycemia inhibits nuclear translocation and expression of forkhead box-O1 (FoxO1) and induces the expression of neurogenin-3 (Ngn3), which is required for the development and maintenance of pancreatic endocrine progenitor cells. This new hypothesis (Foxology) is attracting attention because it explains molecular mechanism(s) underlying β-cell plasticity. The lineage tracing technique revealed that the contribution of dedifferentiation is higher than that of β-cell apoptosis retaining to β-cell mass loss. In addition, islet cells transdifferentiate each other, such as transdifferentiation of pancreatic β-cell to α-cell and vice versa. Islet cells can exhibit plasticity, and they may have the ability to redifferentiate into any cell type. This review describes recent findings in the dedifferentiation and transdifferentiation of β-cells. We outline novel treatment(s) for diabetes targeting islet cell plasticity.
Collapse
Affiliation(s)
- Norikiyo Honzawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, 3-25-8, Nishishinbashi, Minato-ku, Tokyo 105-8461, Japan;
| | - Kei Fujimoto
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University Kashiwa Hospital, 163-1, Kashiwashita, Kshiwa-shi, Chiba 277-8567, Japan
- Correspondence: ; Tel.: +81-04-7164-1111
| |
Collapse
|
56
|
Montaser H, Patel KA, Balboa D, Ibrahim H, Lithovius V, Näätänen A, Chandra V, Demir K, Acar S, Ben-Omran T, Colclough K, Locke JM, Wakeling M, Lindahl M, Hattersley AT, Saarimäki-Vire J, Otonkoski T. Loss of MANF Causes Childhood-Onset Syndromic Diabetes Due to Increased Endoplasmic Reticulum Stress. Diabetes 2021; 70:1006-1018. [PMID: 33500254 PMCID: PMC7610619 DOI: 10.2337/db20-1174] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-resident protein that plays a crucial role in attenuating ER stress responses. Although MANF is indispensable for the survival and function of mouse β-cells, its precise role in human β-cell development and function is unknown. In this study, we show that lack of MANF in humans results in diabetes due to increased ER stress, leading to impaired β-cell function. We identified two patients from different families with childhood diabetes and a neurodevelopmental disorder associated with homozygous loss-of-function mutations in the MANF gene. To study the role of MANF in human β-cell development and function, we knocked out the MANF gene in human embryonic stem cells and differentiated them into pancreatic endocrine cells. Loss of MANF induced mild ER stress and impaired insulin-processing capacity of β-cells in vitro. Upon implantation to immunocompromised mice, the MANF knockout grafts presented elevated ER stress and functional failure, particularly in recipients with diabetes. By describing a new form of monogenic neurodevelopmental diabetes syndrome caused by disturbed ER function, we highlight the importance of adequate ER stress regulation for proper human β-cell function and demonstrate the crucial role of MANF in this process.
Collapse
Affiliation(s)
- Hossam Montaser
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, U.K.
| | - Diego Balboa
- Bioinformatics and Genomics Program, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Väinö Lithovius
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anna Näätänen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vikash Chandra
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Korcan Demir
- Department of Pediatric Endocrinology, Dokuz Eylül University, Izmir, Turkey
| | - Sezer Acar
- Department of Pediatric Endocrinology, Dokuz Eylül University, Izmir, Turkey
| | - Tawfeg Ben-Omran
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, Doha, Qatar
- Department of Pediatrics, Weill Cornell Medical College, Doha, Qatar
- Division of Genetic and Genomic Medicine, Sidra Medicine, Doha, Qatar
| | - Kevin Colclough
- Department of Molecular Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter, U.K
| | - Jonathan M Locke
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, U.K
| | - Matthew Wakeling
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, U.K
| | - Maria Lindahl
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, U.K
| | - Jonna Saarimäki-Vire
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
57
|
Miranda MA, Macias-Velasco JF, Lawson HA. Pancreatic β-cell heterogeneity in health and diabetes: classes, sources, and subtypes. Am J Physiol Endocrinol Metab 2021; 320:E716-E731. [PMID: 33586491 PMCID: PMC8238131 DOI: 10.1152/ajpendo.00649.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pancreatic β-cells perform glucose-stimulated insulin secretion, a process at the center of type 2 diabetes etiology. Efforts to understand how β-cells behave in healthy and stressful conditions have revealed a wide degree of morphological, functional, and transcriptional heterogeneity. Sources of heterogeneity include β-cell topography, developmental origin, maturation state, and stress response. Advances in sequencing and imaging technologies have led to the identification of β-cell subtypes, which play distinct roles in the islet niche. This review examines β-cell heterogeneity from morphological, functional, and transcriptional perspectives, and considers the relevance of topography, maturation, development, and stress response. It also discusses how these factors have been used to identify β-cell subtypes, and how heterogeneity is impacted by diabetes. We examine open questions in the field and discuss recent technological innovations that could advance understanding of β-cell heterogeneity in health and disease.
Collapse
Affiliation(s)
- Mario A Miranda
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Juan F Macias-Velasco
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
58
|
Zhu B, Chen Y, Xu F, Shen X, Chen X, Lv J, Zhang S. Androgens impair β-cell function in a mouse model of polycystic ovary syndrome by activating endoplasmic reticulum stress. Endocr Connect 2021; 10:265-272. [PMID: 33543730 PMCID: PMC8052571 DOI: 10.1530/ec-20-0608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Androgens excess results in endoplasmic reticulum (ER) stress, which is an important cause of β cells dysfunction. Here, we investigated the molecular regulation of androgens excess, ER stress, and β-cell function in polycystic ovary syndrome (PCOS). METHODS PCOS mouse model was established by injection of DHEA. Primary cultured mouse islets were used to detect testosterone (TE)-induced ER stress. The response of ER stress, apoptosis, and hyperinsulinemia were analyzed in INS-1 cells with or without TE exposure. Androgen receptor (AR) antagonist and ER stress inhibitor treatment was performed to evaluate the role of TE in ER stress and proinsulin secretion of PCOS mice. RESULTS PCOS mice had higher ER stress in islets. TE exposure induced ER stress and apoptosis significantly through sustaining insulin overexpression in β cells, which in turn impaired proinsulin maturation and secretion. Blocking this process could significantly relieve ER stress and apoptosis and improve insulin homeostasis. CONCLUSION ER stress activated by androgens excess in PCOS contributes to β cell dysfunction and hyperinsulinemia.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Obstetrics and Gynecology, Assisted Reproduction Unit, Sir Run Run ShawHospital, Zhejiang University School of Medicine Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Gynecology and Obstetrics, Wenzhou People’s Hospital, Wenzhou Women and Children Health, Wenzhou, Zhejiang, China
| | - Yumei Chen
- Department of Gynecology and Obstetrics, Wenzhou People’s Hospital, Wenzhou Women and Children Health, Wenzhou, Zhejiang, China
| | - Fang Xu
- Department of Gynecology and Obstetrics, Wenzhou People’s Hospital, Wenzhou Women and Children Health, Wenzhou, Zhejiang, China
| | - Xiaolu Shen
- Department of Gynecology and Obstetrics, Wenzhou People’s Hospital, Wenzhou Women and Children Health, Wenzhou, Zhejiang, China
| | - Xuanyu Chen
- Department of Gynecology and Obstetrics, Wenzhou People’s Hospital, Wenzhou Women and Children Health, Wenzhou, Zhejiang, China
| | - Jieqiang Lv
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songying Zhang
- Department of Obstetrics and Gynecology, Assisted Reproduction Unit, Sir Run Run ShawHospital, Zhejiang University School of Medicine Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
59
|
Huang JS, Guo BB, Wang GH, Zeng LM, Hu YH, Wang T, Wang HY. DGAT1 inhibitors protect pancreatic β-cells from palmitic acid-induced apoptosis. Acta Pharmacol Sin 2021; 42:264-271. [PMID: 32737468 DOI: 10.1038/s41401-020-0482-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
Previous studies demonstrated that prolonged exposure to elevated levels of free fatty acids (FFA), especially saturated fatty acids, could lead to pancreatic β-cell apoptosis, which plays an important role in the progression of type 2 diabetes (T2D). Diacylglycerol acyltransferase 1 (DGAT1), an enzyme that catalyzes the final step of triglyceride (TG) synthesis, has been reported as a novel target for the treatment of multiple metabolic diseases. In this study we evaluated the potential beneficial effects of DGAT1 inhibitors on pancreatic β-cells, and further verified their antidiabetic effects in db/db mice. We showed that DGAT1 inhibitors (4a and LCQ908) at the concentration of 1 μM significantly ameliorated palmitic acid (PA)-induced apoptosis in MIN6 pancreatic β-cells and primary cultured mouse islets; oral administration of a DGAT1 inhibitor (4a) (100 mg/kg) for 4 weeks significantly reduced the apoptosis of pancreatic islets in db/db mice. Meanwhile, 4a administration significantly decreased fasting blood glucose and TG levels, and improved glucose tolerance and insulin tolerance in db/db mice. Furthermore, we revealed that pretreatment with 4a (1 μM) significantly alleviated PA-induced intracellular lipid accumulation, endoplasmic reticulum (ER) stress, and proinflammatory responses in MIN6 cells, which might contribute to the protective effects of DGAT1 inhibitors on pancreatic β-cells. These findings provided a better understanding of the antidiabetic effects of DGAT1 inhibitors.
Collapse
|
60
|
Xiong Y, Jin E, Yin Q, Che C, He S. Boron Attenuates Heat Stress-Induced Apoptosis by Inhibiting Endoplasmic Reticulum Stress in Mouse Granulosa Cells. Biol Trace Elem Res 2021; 199:611-621. [PMID: 32385716 DOI: 10.1007/s12011-020-02180-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/28/2020] [Indexed: 01/25/2023]
Abstract
Heat stress-induced apoptosis in granulosa cells is mediated by multiple apoptotic signaling pathways, including endoplasmic reticulum (ER) stress. Boron is a naturally occurring trace element with several cytoprotective properties. Nonetheless, the molecular mechanisms involved in the protective functions of boron in granulosa cells undergoing apoptosis caused by heat stress (HS) remain unclear. In this study, we investigated the role of boric acid, a predominant chemical form of boron, in HS-induced apoptotic damage in mouse granulosa cells (mGCs) and explored the underlying mechanisms. We found that HS treatment suppressed cell viability; increased the apoptotic rate of cells; potentiated the activity of caspase-3, a key player in the caspase-mediated apoptotic signaling pathway; and activated ER stress markers, including glucose-regulated protein 78 (GRP78) and CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) in mGCs. However, boric acid treatment effectively alleviated the effects of both HS-induced and thapsigargin (an ER stress agonist)-induced apoptosis, such as the enhanced activity of caspase-3 and increase in GRP78 and CHOP expression. Moreover, treatment with 4-phenylbutyrate (4-PBA), an ER stress antagonist, significantly attenuated these HS-induced adverse effects in mGCs. In addition, boric acid supplementation in the culture medium significantly restored the decreased estradiol levels in heat-treated mGCs. The administration of boric acid to female mice previously exposed to hyperthermal conditions effectively restored the levels of serum estradiol in vivo. Collectively, these findings suggest that HS induces apoptosis in mGCs via ER stress pathways and that boron has a protective effect against these adverse effects. This study provides novel insights into the benefits of using boron against heat-induced apoptosis.
Collapse
Affiliation(s)
- Yongjie Xiong
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
- Key Laboratory of the Quality and Safety Control for Pork of the Ministry of Agriculture, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
- Key Laboratory of the Quality and Safety Control for Pork of the Ministry of Agriculture, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
| | - Qirun Yin
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
- Key Laboratory of the Quality and Safety Control for Pork of the Ministry of Agriculture, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
| | - Chuanyan Che
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
- Key Laboratory of the Quality and Safety Control for Pork of the Ministry of Agriculture, Anhui Science and Technology University, Fengyang, 233100, Anhui, China
| | - Shaojun He
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100, Anhui, China.
- Key Laboratory of the Quality and Safety Control for Pork of the Ministry of Agriculture, Anhui Science and Technology University, Fengyang, 233100, Anhui, China.
| |
Collapse
|
61
|
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: 0.7] [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
|
62
|
Avrahami D, Wang YJ, Schug J, Feleke E, Gao L, Liu C, Naji A, Glaser B, Kaestner KH. Single-cell transcriptomics of human islet ontogeny defines the molecular basis of β-cell dedifferentiation in T2D. Mol Metab 2020; 42:101057. [PMID: 32739450 PMCID: PMC7471622 DOI: 10.1016/j.molmet.2020.101057] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Dedifferentiation of pancreatic β-cells may reduce islet function in type 2 diabetes (T2D). However, the prevalence, plasticity and functional consequences of this cellular state remain unknown. METHODS We employed single-cell RNAseq to detail the maturation program of α- and β-cells during human ontogeny. We also compared islets from non-diabetic and T2D individuals. RESULTS Both α- and β-cells mature in part by repressing non-endocrine genes; however, α-cells retain hallmarks of an immature state, while β-cells attain a full β-cell specific gene expression program. In islets from T2D donors, both α- and β-cells have a less mature expression profile, de-repressing the juvenile genetic program and exocrine genes and increasing expression of exocytosis, inflammation and stress response signalling pathways. These changes are consistent with the increased proportion of β-cells displaying suboptimal function observed in T2D islets. CONCLUSIONS These findings provide new insights into the molecular program underlying islet cell maturation during human ontogeny and the loss of transcriptomic maturity that occurs in islets of type 2 diabetics.
Collapse
Affiliation(s)
- Dana Avrahami
- Endocrinology and Metabolism Department, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Yue J Wang
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Eseye Feleke
- Endocrinology and Metabolism Department, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Long Gao
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Department of Surgery and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ali Naji
- Department of Surgery and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin Glaser
- Endocrinology and Metabolism Department, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel.
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
63
|
Hayden MR. An Immediate and Long-Term Complication of COVID-19 May Be Type 2 Diabetes Mellitus: The Central Role of β-Cell Dysfunction, Apoptosis and Exploration of Possible Mechanisms. Cells 2020; 9:E2475. [PMID: 33202960 PMCID: PMC7697826 DOI: 10.3390/cells9112475] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was declared a pandemic by the WHO on 19 March 2020. This pandemic is associated with markedly elevated blood glucose levels and a remarkable degree of insulin resistance, which suggests pancreatic islet β-cell dysfunction or apoptosis and insulin's inability to dispose of glucose into cellular tissues. Diabetes is known to be one of the top pre-existing co-morbidities associated with the severity of COVID-19 along with hypertension, cardiocerebrovascular disease, advanced age, male gender, and recently obesity. This review focuses on how COVID-19 may be responsible for the accelerated development of type 2 diabetes mellitus (T2DM) as one of its acute and suspected long-term complications. These observations implicate an active role of metabolic syndrome, systemic and tissue islet renin-angiotensin-aldosterone system, redox stress, inflammation, islet fibrosis, amyloid deposition along with β-cell dysfunction and apoptosis in those who develop T2DM. Utilizing light and electron microscopy in preclinical rodent models and human islets may help to better understand how COVID-19 accelerates islet and β-cell injury and remodeling to result in the long-term complications of T2DM.
Collapse
Affiliation(s)
- Melvin R Hayden
- Departments of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri-Columbia School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
64
|
Buday K, Conrad M. Emerging roles for non-selenium containing ER-resident glutathione peroxidases in cell signaling and disease. Biol Chem 2020; 402:271-287. [PMID: 33055310 DOI: 10.1515/hsz-2020-0286] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/08/2020] [Indexed: 12/16/2022]
Abstract
Maintenance of cellular redox control is pivotal for normal cellular functions and cell fate decisions including cell death. Among the key cellular redox systems in mammals, the glutathione peroxidase (GPX) family of proteins is the largest conferring multifaceted functions and affecting virtually all cellular processes. The endoplasmic reticulum (ER)-resident GPXs, designated as GPX7 and GPX8, are the most recently added members of this family of enzymes. Recent studies have provided exciting insights how both enzymes support critical processes of the ER including oxidative protein folding, maintenance of ER redox control by eliminating H2O2, and preventing palmitic acid-induced lipotoxicity. Consequently, numerous pathological conditions, such as neurodegeneration, cancer and metabolic diseases have been linked with altered GPX7 and GPX8 expression. Studies in mice have demonstrated that loss of GPX7 leads to increased differentiation of preadipocytes, increased tumorigenesis and shortened lifespan. By contrast, GPX8 deficiency in mice results in enhanced caspase-4/11 activation and increased endotoxic shock in colitis model. With the increasing recognition that both types of enzymes are dysregulated in various tumor entities in man, we deem a review of the emerging roles played by GPX7 and GPX8 in health and disease development timely and appropriate.
Collapse
Affiliation(s)
- Katalin Buday
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764Neuherberg, Germany.,National Research Medical University, Laboratory of Experimental Oncology, Ostrovityanova 1, 117997Moscow, Russia
| |
Collapse
|
65
|
Giri KR, de Beaurepaire L, Jegou D, Lavy M, Mosser M, Dupont A, Fleurisson R, Dubreil L, Collot M, Van Endert P, Bach JM, Mignot G, Bosch S. Molecular and Functional Diversity of Distinct Subpopulations of the Stressed Insulin-Secreting Cell's Vesiculome. Front Immunol 2020; 11:1814. [PMID: 33101266 PMCID: PMC7556286 DOI: 10.3389/fimmu.2020.01814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Beta cell failure and apoptosis following islet inflammation have been associated with autoimmune type 1 diabetes pathogenesis. As conveyors of biological active material, extracellular vesicles (EV) act as mediators in communication with immune effectors fostering the idea that EV from inflamed beta cells may contribute to autoimmunity. Evidence accumulates that beta exosomes promote diabetogenic responses, but relative contributions of larger vesicles as well as variations in the composition of the beta cell's vesiculome due to environmental changes have not been explored yet. Here, we made side-by-side comparisons of the phenotype and function of apoptotic bodies (AB), microvesicles (MV) and small EV (sEV) isolated from an equal amount of MIN6 beta cells exposed to inflammatory, hypoxic or genotoxic stressors. Under normal conditions, large vesicles represent 93% of the volume, but only 2% of the number of the vesicles. Our data reveal a consistently higher release of AB and sEV and to a lesser extent of MV, exclusively under inflammatory conditions commensurate with a 4-fold increase in the total volume of the vesiculome and enhanced export of immune-stimulatory material including the autoantigen insulin, microRNA, and cytokines. Whilst inflammation does not change the concentration of insulin inside the EV, specific Toll-like receptor-binding microRNA sequences preferentially partition into sEV. Exposure to inflammatory stress engenders drastic increases in the expression of monocyte chemoattractant protein 1 in all EV and of interleukin-27 solely in AB suggesting selective sorting toward EV subspecies. Functional in vitro assays in mouse dendritic cells and macrophages reveal further differences in the aptitude of EV to modulate expression of cytokines and maturation markers. These findings highlight the different quantitative and qualitative imprints of environmental changes in subpopulations of beta EV that may contribute to the spread of inflammation and sustained immune cell recruitment at the inception of the (auto-) immune response.
Collapse
Affiliation(s)
| | | | | | - Margot Lavy
- IECM, ONIRIS, INRAE, USC1383, Nantes, France
| | | | - Aurelien Dupont
- MRic, Biosit, UMS3480 CNRS, University of Rennes 1, Rennes, France
| | | | - Laurence Dubreil
- PAnTher, INRAE, Oniris, Université Bretagne Loire, Nantes, France
| | - Mayeul Collot
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Université de Strasbourg, Illkirch, France
| | - Peter Van Endert
- Université Paris Descartes, Paris, France.,INSERM, U1151, Institut Necker-Enfants Malades, Paris, France
| | | | | | | |
Collapse
|
66
|
Gaddam RR, Kim YR, Li Q, Jacobs JS, Gabani M, Mishra A, Promes JA, Imai Y, Irani K, Vikram A. Genetic deletion of miR-204 improves glycemic control despite obesity in db/db mice. Biochem Biophys Res Commun 2020; 532:167-172. [PMID: 32950230 DOI: 10.1016/j.bbrc.2020.08.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRs) are small non-coding RNAs that regulate the target gene expression. A change in miR profile in the pancreatic islets during diabetes is known, and multiple studies have demonstrated that miRs influence the pancreatic β-cell function. The miR-204 is highly expressed in the β-cells and reported to regulate insulin synthesis. Here we investigated whether the absence of miR-204 rescues the impaired glycemic control and obesity in the genetically diabetic (db/db) mice. We found that the db/db mice overexpressed miR-204 in the islets. The db/db mice lacking miR-204 (db/db-204-/-) initially develops hyperglycemia and obesity like the control (db/db) mice but later displayed a gradual improvement in glycemic control despite remaining obese. The db/db-204-/- mice had a lower fasting blood glucose and higher serum insulin level compared to the db/db mice. A homeostatic model assessment (HOMA) suggests the improvement of β-cell function contributes to the improvement in glycemic control in db/db-204-/- mice. Next, we examined the cellular proliferation and endoplasmic reticulum (ER) stress and found an increased frequency of proliferating cells (PCNA + ve) and a decreased CHOP expression in the islets of db/db-204-/- mice. Next, we determined the effect of systemic miR-204 inhibition in improving glycemic control in the high-fat diet (HFD)-fed insulin-resistant mice. MiR-204 inhibition for 6 weeks improved the HFD-triggered impairment in glucose disposal. In conclusion, the absence of miR-204 improves β-cell proliferation, decreases islet ER stress, and improves glycemic control with limited change in body weight in obese mice.
Collapse
Affiliation(s)
- Ravinder Reddy Gaddam
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Young-Rae Kim
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Quixia Li
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Julia S Jacobs
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mohanad Gabani
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Akansha Mishra
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Joseph A Promes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Yumi Imai
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| | - Ajit Vikram
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
67
|
Kang HJ, Yoo EJ, Lee HH, An SM, Park H, Lee-Kwon W, Choi SY, Kwon HM. TonEBP Promotes β-Cell Survival under ER Stress by Enhancing Autophagy. Cells 2020; 9:cells9091928. [PMID: 32825390 PMCID: PMC7563687 DOI: 10.3390/cells9091928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/16/2022] Open
Abstract
The endoplasmic reticulum (ER) stress response and autophagy are important cellular responses that determine cell fate and whose dysregulation is implicated in the perturbation of homeostasis and diseases. Tonicity-responsive enhancer-binding protein (TonEBP, also called NFAT5) is a pleiotropic stress protein that mediates both protective and pathological cellular responses. Here, we examined the role of TonEBP in β-cell survival under ER stress. We found that TonEBP increases β-cell survival under ER stress by enhancing autophagy. The level of TonEBP protein increased under ER stress due to a reduction in its degradation via the ubiquitin–proteasome pathway. In response to ER stress, TonEBP increased autophagosome formations and suppressed the accumulation of protein aggregates and β-cell death. The Rel-homology domain of TonEBP interacted with FIP200, which is essential for the initiation of autophagy, and was required for autophagy and cell survival upon exposure to ER stress. Mice in which TonEBP was specifically deleted in pancreatic endocrine progenitor cells exhibited defective glucose homeostasis and a loss of islet mass. Taken together, these findings demonstrate that TonEBP protects against ER stress-induced β-cell death by enhancing autophagy.
Collapse
|
68
|
Sharma RB, Darko C, Alonso LC. Intersection of the ATF6 and XBP1 ER stress pathways in mouse islet cells. J Biol Chem 2020; 295:14164-14177. [PMID: 32788214 DOI: 10.1074/jbc.ra120.014173] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/06/2020] [Indexed: 12/20/2022] Open
Abstract
Success or failure of pancreatic beta cell adaptation to ER stress is a determinant of diabetes susceptibility. The ATF6 and IRE1/XBP1 pathways are separate ER stress-response effectors important to beta cell health and function. ATF6α. and XBP1 direct overlapping transcriptional responses in some cell types. However, the signaling dynamics and interdependence of ATF6α and XBP1 in pancreatic beta cells have not been explored. To assess pathway-specific signal onset, we performed timed exposures of primary mouse islet cells to ER stressors and measured the early transcriptional response. Comparing the time course of induction of ATF6 and XBP1 targets suggested that the two pathways have similar response dynamics. The role of ATF6α in target induction was assessed by acute knockdown using islet cells from Atf6α flox/flox mice transduced with adenovirus expressing Cre recombinase. Surprisingly, given the mild impact of chronic deletion in mice, acute ATF6α knockdown markedly reduced ATF6-pathway target gene expression under both basal and stressed conditions. Intriguingly, although ATF6α knockdown did not alter Xbp1 splicing dynamics or intensity, it did reduce induction of XBP1 targets. Inhibition of Xbp1 splicing did not decrease induction of ATF6α targets. Taken together, these data suggest that the XBP1 and ATF6 pathways are simultaneously activated in islet cells in response to acute stress and that ATF6α is required for full activation of XBP1 targets, but XBP1 is not required for activation of ATF6α targets. These observations improve understanding of the ER stress transcriptional response in pancreatic islets.
Collapse
Affiliation(s)
- Rohit B Sharma
- Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine, New York, New York, USA .,Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, USA
| | - Christine Darko
- Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine, New York, New York, USA.,Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, USA
| | - Laura C Alonso
- Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine, New York, New York, USA .,Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
69
|
Abreu D, Asada R, Revilla JMP, Lavagnino Z, Kries K, Piston DW, Urano F. Wolfram syndrome 1 gene regulates pathways maintaining beta-cell health and survival. J Transl Med 2020; 100:849-862. [PMID: 32060407 PMCID: PMC7286786 DOI: 10.1038/s41374-020-0408-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
Wolfram Syndrome 1 (WFS1) protein is an endoplasmic reticulum (ER) factor whose deficiency results in juvenile-onset diabetes secondary to cellular dysfunction and apoptosis. The mechanisms guiding β-cell outcomes secondary to WFS1 function, however, remain unclear. Here, we show that WFS1 preserves normal β-cell physiology by promoting insulin biosynthesis and negatively regulating ER stress. Depletion of Wfs1 in vivo and in vitro causes functional defects in glucose-stimulated insulin secretion and insulin content, triggering Chop-mediated apoptotic pathways. Genetic proof of concept studies coupled with RNA-seq reveal that increasing WFS1 confers a functional and a survival advantage to β-cells under ER stress by increasing insulin gene expression and downregulating the Chop-Trib3 axis, thereby activating Akt pathways. Remarkably, WFS1 and INS levels are reduced in type-2 diabetic (T2DM) islets, suggesting that WFS1 may contribute to T2DM β-cell pathology. Taken together, this work reveals essential pathways regulated by WFS1 to control β-cell survival and function primarily through preservation of ER homeostasis.
Collapse
Affiliation(s)
- Damien Abreu
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA,Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, U.S.A
| | - Rie Asada
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Biochemistry, Institute of Biomedical & Health Science, Hiroshima University, Hiroshima 734-8553, Japan
| | - John M. P. Revilla
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zeno Lavagnino
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA,Experimental Imaging Center DIBIT, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Kelly Kries
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David W. Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
70
|
The Perlman syndrome DIS3L2 exoribonuclease safeguards endoplasmic reticulum-targeted mRNA translation and calcium ion homeostasis. Nat Commun 2020; 11:2619. [PMID: 32457326 PMCID: PMC7250864 DOI: 10.1038/s41467-020-16418-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 04/30/2020] [Indexed: 11/16/2022] Open
Abstract
DIS3L2-mediated decay (DMD) is a surveillance pathway for certain non-coding RNAs (ncRNAs) including ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), small nuclear RNAs (snRNAs), and RMRP. While mutations in DIS3L2 are associated with Perlman syndrome, the biological significance of impaired DMD is obscure and pathological RNAs have not been identified. Here, by ribosome profiling (Ribo-seq) we find specific dysregulation of endoplasmic reticulum (ER)-targeted mRNA translation in DIS3L2-deficient cells. Mechanistically, DMD functions in the quality control of the 7SL ncRNA component of the signal recognition particle (SRP) required for ER-targeted translation. Upon DIS3L2 loss, sustained 3’-end uridylation of aberrant 7SL RNA impacts ER-targeted translation and causes ER calcium leakage. Consequently, elevated intracellular calcium in DIS3L2-deficient cells activates calcium signaling response genes and perturbs ESC differentiation. Thus, DMD is required to safeguard ER-targeted mRNA translation, intracellular calcium homeostasis, and stem cell differentiation. The DIS3L2 exonuclease degrades aberrant 7SL RNAs tagged by an oligouridine 3′-tail. Here the authors analyze DIS3L2 knockout mouse embryonic stem cells and suggest that DIS3L2-mediated quality control of 7SL RNA is important for ER-mediated translation and calcium ion homeostasis.
Collapse
|
71
|
AKT1 Regulates Endoplasmic Reticulum Stress and Mediates the Adaptive Response of Pancreatic β Cells. Mol Cell Biol 2020; 40:MCB.00031-20. [PMID: 32179553 DOI: 10.1128/mcb.00031-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022] Open
Abstract
Isoforms of protein kinase B (also known as AKT) play important roles in mediating insulin and growth factor signals. Previous studies have suggested that the AKT2 isoform is critical for insulin-regulated glucose metabolism, while the role of the AKT1 isoform remains less clear. This study focuses on the effects of AKT1 on the adaptive response of pancreatic β cells. Using a mouse model with inducible β-cell-specific deletion of the Akt1 gene (βA1KO mice), we showed that AKT1 is involved in high-fat-diet (HFD)-induced growth and survival of β cells but is unnecessary for them to maintain a population in the absence of metabolic stress. When unchallenged, βA1KO mice presented the same metabolic profile and β-cell phenotype as the control mice with an intact Akt1 gene. When metabolic stress was induced by HFD, β cells in control mice with intact Akt1 proliferated as a compensatory mechanism for metabolic overload. Similar effects were not observed in βA1KO mice. We further demonstrated that AKT1 protein deficiency caused endoplasmic reticulum (ER) stress and potentiated β cells to undergo apoptosis. Our results revealed that AKT1 protein loss led to the induction of eukaryotic initiation factor 2 α subunit (eIF2α) signaling and ER stress markers under normal-chow-fed conditions, indicating chronic low-level ER stress. Together, these data established a role for AKT1 as a growth and survival factor for adaptive β-cell response and suggest that ER stress induction is responsible for this effect of AKT1.
Collapse
|
72
|
Hu R, Walker E, Huang C, Xu Y, Weng C, Erickson GE, Coldren A, Yang X, Brissova M, Kaverina I, Balamurugan AN, Wright CVE, Li Y, Stein R, Gu G. Myt Transcription Factors Prevent Stress-Response Gene Overactivation to Enable Postnatal Pancreatic β Cell Proliferation, Function, and Survival. Dev Cell 2020; 53:390-405.e10. [PMID: 32359405 PMCID: PMC7278035 DOI: 10.1016/j.devcel.2020.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 03/06/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Although cellular stress response is important for maintaining function and survival, overactivation of late-stage stress effectors cause dysfunction and death. We show that the myelin transcription factors (TFs) Myt1 (Nzf2), Myt2 (Myt1l, Nztf1, and Png-1), and Myt3 (St18 and Nzf3) prevent such overactivation in islet β cells. Thus, we found that co-inactivating the Myt TFs in mouse pancreatic progenitors compromised postnatal β cell function, proliferation, and survival, preceded by upregulation of late-stage stress-response genes activating transcription factors (e.g., Atf4) and heat-shock proteins (Hsps). Myt1 binds putative enhancers of Atf4 and Hsps, whose overexpression largely recapitulated the Myt-mutant phenotypes. Moreover, Myt(MYT)-TF levels were upregulated in mouse and human β cells during metabolic stress-induced compensation but downregulated in dysfunctional type 2 diabetic (T2D) human β cells. Lastly, MYT knockdown caused stress-gene overactivation and death in human EndoC-βH1 cells. These findings suggest that Myt TFs are essential restrictors of stress-response overactivity.
Collapse
Affiliation(s)
- Ruiying Hu
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Emily Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Chen Huang
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yanwen Xu
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Chen Weng
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Gillian E Erickson
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Anastasia Coldren
- Department of Medicine, Vanderbilt Medical Center, Nashville, TN 27232, USA
| | - Xiaodun Yang
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Marcela Brissova
- Department of Medicine, Vanderbilt Medical Center, Nashville, TN 27232, USA
| | - Irina Kaverina
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Appakalai N Balamurugan
- Department of Surgery, Clinical Islet Transplantation Laboratory, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40202, USA
| | - Christopher V E Wright
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yan Li
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Guoqiang Gu
- Vanderbilt Program in Developmental Biology, Department of Cell and Developmental Biology, and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
73
|
Constantin A, Dumitrescu M, Nemecz M, Picu A, Smeu B, Guja C, Alexandru N, Georgescu A, Tanko G. Sera of Obese Type 2 Diabetic Patients Undergoing Metabolic Surgery Instead of Conventional Treatment Exert Beneficial Effects on Beta Cell Survival and Function: Results of a Randomized Clinical Study. Obes Surg 2020; 29:1485-1497. [PMID: 30701387 DOI: 10.1007/s11695-019-03710-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pancreatic beta cells are highly sensitive to oxidative and endoplasmic reticulum (ER) stress, commonly occurring in type 2 diabetes (T2D) and obesity. OBJECTIVE We aimed at investigating cellular responses of human beta cells exposed to sera from obese T2D patients treated differently, namely by conventional therapy or laparoscopic sleeve gastrectomy (LSG). METHODS Serum samples from obese T2D men randomized to conventional treatment or LSG were taken at baseline and 6 months later. After exposing 1.1B4 cells to study patients' sera, the following were assessed: cellular viability and proliferation (by MTT and xCELLigence assays), reactive oxygen species (ROS) production (with DCFH-DA), and expression of ER stress markers, oxidative- or autophagy-related proteins and insulin (by real-time PCR and Western blot). RESULTS At 6-month follow-up, patients undergoing LSG achieved an adequate glycemic control, whereas conventionally treated patients did not. As compared to 1.1B4 cells incubated with baseline sera (control), cells exposed to sera from LSG-treated participants exhibited (i) increased viability and proliferation (p < 0.05); (ii) diminished levels of ROS and p53 (p < 0.05); (iii) enhanced protein expression of autophagy-related SIRT1 and p62/SQSTM1 (p < 0.05); (iv) significantly decreased transcript levels of ER stress markers (p < 0.05); and (v) augmented insulin expression (p < 0.05). Conversely, the 6-month conventional therapy appeared not to impact on circulating redox status. Moreover, 1.1B4 cells exposed to sera from conventionally treated patients experienced mild ER stress. CONCLUSION Circulating factors in patients with improved diabetes after metabolic surgery exerted favorable effects on beta cell function and survival.
Collapse
Affiliation(s)
- Alina Constantin
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, 8, BP Hasdeu Street, PO Box 35-14, 050568, Bucharest, Romania
| | - Mădălina Dumitrescu
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, 8, BP Hasdeu Street, PO Box 35-14, 050568, Bucharest, Romania
| | - Miruna Nemecz
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, 8, BP Hasdeu Street, PO Box 35-14, 050568, Bucharest, Romania
| | - Ariana Picu
- Institute of Diabetes, Nutrition and Metabolic Diseases "Prof. Dr N. Paulescu", Bucharest, Romania
| | | | - Cristian Guja
- Institute of Diabetes, Nutrition and Metabolic Diseases "Prof. Dr N. Paulescu", Bucharest, Romania
| | - Nicoleta Alexandru
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, 8, BP Hasdeu Street, PO Box 35-14, 050568, Bucharest, Romania
| | - Adriana Georgescu
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, 8, BP Hasdeu Street, PO Box 35-14, 050568, Bucharest, Romania
| | - Gabriela Tanko
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, 8, BP Hasdeu Street, PO Box 35-14, 050568, Bucharest, Romania.
| |
Collapse
|
74
|
Hu Y, Yang W, Xie L, Liu T, Liu H, Liu B. Endoplasmic reticulum stress and pulmonary hypertension. Pulm Circ 2020; 10:2045894019900121. [PMID: 32110387 PMCID: PMC7000863 DOI: 10.1177/2045894019900121] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022] Open
Abstract
Pulmonary hypertension is a fatal disease of which pulmonary vasculopathy is the main pathological feature resulting in the mean pulmonary arterial pressure higher than 25 mmHg. Moreover, pulmonary hypertension remains a tough problem with unclear molecular mechanisms. There have been dozens of studies about endoplasmic reticulum stress during the onset of pulmonary hypertension in patients, suggesting that endoplasmic reticulum stress may have a critical effect on the pathogenesis of pulmonary hypertension. The review aims to summarize the rationale to elucidate the role of endoplasmic reticulum stress in pulmonary hypertension. Started by reviewing the mechanisms responsible for the unfolded protein response following endoplasmic reticulum stress, the potential link between endoplasmic reticulum stress and pulmonary hypertension were introduced, and the contributions of endoplasmic reticulum stress to different vascular cells, mitochondria, and inflammation were described, and finally the potential therapies of attenuating endoplasmic reticulum stress for pulmonary hypertension were discussed.
Collapse
Affiliation(s)
- Yanan Hu
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenhao Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,The Vascular Remodeling and Developmental Defects Research Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liang Xie
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,The Vascular Remodeling and Developmental Defects Research Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Tao Liu
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hanmin Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.,The Vascular Remodeling and Developmental Defects Research Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Bin Liu
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
75
|
Zhang IX, Raghavan M, Satin LS. The Endoplasmic Reticulum and Calcium Homeostasis in Pancreatic Beta Cells. Endocrinology 2020; 161:bqz028. [PMID: 31796960 PMCID: PMC7028010 DOI: 10.1210/endocr/bqz028] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 12/01/2019] [Indexed: 12/14/2022]
Abstract
The endoplasmic reticulum (ER) mediates the first steps of protein assembly within the secretory pathway and is the site where protein folding and quality control are initiated. The storage and release of Ca2+ are critical physiological functions of the ER. Disrupted ER homeostasis activates the unfolded protein response (UPR), a pathway which attempts to restore cellular equilibrium in the face of ER stress. Unremitting ER stress, and insufficient compensation for it results in beta-cell apoptosis, a process that has been linked to both type 1 diabetes (T1D) and type 2 diabetes (T2D). Both types are characterized by progressive beta-cell failure and a loss of beta-cell mass, although the underlying causes are different. The reduction of mass occurs secondary to apoptosis in the case of T2D, while beta cells undergo autoimmune destruction in T1D. In this review, we examine recent findings that link the UPR pathway and ER Ca2+ to beta cell dysfunction. We also discuss how UPR activation in beta cells favors cell survival versus apoptosis and death, and how ER protein chaperones are involved in regulating ER Ca2+ levels. Abbreviations: BiP, Binding immunoglobulin Protein ER; endoplasmic reticulum; ERAD, ER-associated protein degradation; IFN, interferon; IL, interleukin; JNK, c-Jun N-terminal kinase; KHE, proton-K+ exchanger; MODY, maturity-onset diabetes of young; PERK, PRKR-like ER kinase; SERCA, Sarco/Endoplasmic Reticulum Ca2+-ATPases; T1D, type 1 diabetes; T2D, type 2 diabetes; TNF, tumor necrosis factor; UPR, unfolded protein response; WRS, Wolcott-Rallison syndrome.
Collapse
Affiliation(s)
- Irina X Zhang
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan, Ann Arbor, MI
| | - Malini Raghavan
- Department of Microbiology and Immunology Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Leslie S Satin
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
76
|
Sachs S, Bastidas-Ponce A, Tritschler S, Bakhti M, Böttcher A, Sánchez-Garrido MA, Tarquis-Medina M, Kleinert M, Fischer K, Jall S, Harger A, Bader E, Roscioni S, Ussar S, Feuchtinger A, Yesildag B, Neelakandhan A, Jensen CB, Cornu M, Yang B, Finan B, DiMarchi RD, Tschöp MH, Theis FJ, Hofmann SM, Müller TD, Lickert H. Targeted pharmacological therapy restores β-cell function for diabetes remission. Nat Metab 2020; 2:192-209. [PMID: 32694693 DOI: 10.1038/s42255-020-0171-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/15/2020] [Indexed: 12/27/2022]
Abstract
Dedifferentiation of insulin-secreting β cells in the islets of Langerhans has been proposed to be a major mechanism of β-cell dysfunction. Whether dedifferentiated β cells can be targeted by pharmacological intervention for diabetes remission, and ways in which this could be accomplished, are unknown as yet. Here we report the use of streptozotocin-induced diabetes to study β-cell dedifferentiation in mice. Single-cell RNA sequencing (scRNA-seq) of islets identified markers and pathways associated with β-cell dedifferentiation and dysfunction. Single and combinatorial pharmacology further show that insulin treatment triggers insulin receptor pathway activation in β cells and restores maturation and function for diabetes remission. Additional β-cell selective delivery of oestrogen by Glucagon-like peptide-1 (GLP-1-oestrogen conjugate) decreases daily insulin requirements by 60%, triggers oestrogen-specific activation of the endoplasmic-reticulum-associated protein degradation system, and further increases β-cell survival and regeneration. GLP-1-oestrogen also protects human β cells against cytokine-induced dysfunction. This study not only describes mechanisms of β-cell dedifferentiation and regeneration, but also reveals pharmacological entry points to target dedifferentiated β cells for diabetes remission.
Collapse
Affiliation(s)
- Stephan Sachs
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Miguel A Sánchez-Garrido
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Marta Tarquis-Medina
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Maximilian Kleinert
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Katrin Fischer
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| | - Sigrid Jall
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| | - Alexandra Harger
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Erik Bader
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Sara Roscioni
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Siegfried Ussar
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | | | - Marion Cornu
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Bin Yang
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - Matthias H Tschöp
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Munich, Germany.
| | - Susanna M Hofmann
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medical Clinic and Polyclinic IV, Ludwig Maximilian University of Munich, Munich, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany.
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany.
- Department of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
77
|
Mahadevan J, Morikawa S, Yagi T, Abreu D, Lu S, Kanekura K, Brown CM, Urano F. A soluble endoplasmic reticulum factor as regenerative therapy for Wolfram syndrome. J Transl Med 2020; 100:1197-1207. [PMID: 32366942 PMCID: PMC7438202 DOI: 10.1038/s41374-020-0436-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 01/24/2023] Open
Abstract
Endoplasmic reticulum (ER) stress-mediated cell death is an emerging target for human chronic disorders, including neurodegeneration and diabetes. However, there is currently no treatment for preventing ER stress-mediated cell death. Here, we show that mesencephalic astrocyte-derived neurotrophic factor (MANF), a neurotrophic factor secreted from ER stressed cells, prevents ER stress-mediated β cell death and enhances β cell proliferation in cell and mouse models of Wolfram syndrome, a prototype of ER disorders. Our results indicate that molecular pathways regulated by MANF are promising therapeutic targets for regenerative therapy of ER stress-related disorders, including diabetes, retinal degeneration, neurodegeneration, and Wolfram syndrome.
Collapse
Affiliation(s)
- Jana Mahadevan
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Shuntaro Morikawa
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Takuya Yagi
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Damien Abreu
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Simin Lu
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Kohsuke Kanekura
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.410793.80000 0001 0663 3325Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Cris M. Brown
- grid.4367.60000 0001 2355 7002Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
78
|
Beetch M, Harandi-Zadeh S, Shen K, Lubecka K, Kitts DD, O'Hagan HM, Stefanska B. Dietary antioxidants remodel DNA methylation patterns in chronic disease. Br J Pharmacol 2019; 177:1382-1408. [PMID: 31626338 DOI: 10.1111/bph.14888] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic diseases account for over 60% of all deaths worldwide according to the World Health Organization reports. Majority of cases are triggered by environmental exposures that lead to aberrant changes in the epigenome, specifically, the DNA methylation patterns. These changes result in altered expression of gene networks and activity of signalling pathways. Dietary antioxidants, including catechins, flavonoids, anthocyanins, stilbenes and carotenoids, demonstrate benefits in the prevention and/or support of therapy in chronic diseases. This review provides a comprehensive discussion of potential epigenetic mechanisms of antioxidant compounds in reversing altered patterns of DNA methylation in chronic disease. Antioxidants remodel the DNA methylation patterns through multiple mechanisms, including regulation of epigenetic enzymes and chromatin remodelling complexes. These effects can further contribute to antioxidant properties of the compounds. On the other hand, decrease in oxidative stress itself can impact DNA methylation delivering additional link between antioxidant mechanisms and epigenetic effects of the compounds. LINKED ARTICLES: This article is part of a themed section on The Pharmacology of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.6/issuetoc.
Collapse
Affiliation(s)
- Megan Beetch
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Sadaf Harandi-Zadeh
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Kate Shen
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Katarzyna Lubecka
- Department of Biomedical Chemistry, Medical University of Lodz, Lodz, Poland
| | - David D Kitts
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Heather M O'Hagan
- Cell, Molecular and Cancer Biology, Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
79
|
Kim HJ, Moon JH, Chung H, Shin JS, Kim B, Kim JM, Kim JS, Yoon IH, Min BH, Kang SJ, Kim YH, Jo K, Choi J, Chae H, Lee WW, Kim S, Park CG. Bioinformatic analysis of peripheral blood RNA-sequencing sensitively detects the cause of late graft loss following overt hyperglycemia in pig-to-nonhuman primate islet xenotransplantation. Sci Rep 2019; 9:18835. [PMID: 31827198 PMCID: PMC6906328 DOI: 10.1038/s41598-019-55417-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 11/12/2019] [Indexed: 01/19/2023] Open
Abstract
Clinical islet transplantation has recently been a promising treatment option for intractable type 1 diabetes patients. Although early graft loss has been well studied and controlled, the mechanisms of late graft loss largely remains obscure. Since long-term islet graft survival had not been achieved in islet xenotransplantation, it has been impossible to explore the mechanism of late islet graft loss. Fortunately, recent advances where consistent long-term survival (≥6 months) of adult porcine islet grafts was achieved in five independent, diabetic nonhuman primates (NHPs) enabled us to investigate on the late graft loss. Regardless of the conventional immune monitoring methods applied in the post-transplant period, the initiation of late graft loss could rarely be detected before the overt graft loss observed via uncontrolled blood glucose level. Thus, we retrospectively analyzed the gene expression profiles in 2 rhesus monkey recipients using peripheral blood RNA-sequencing (RNA-seq) data to find out the potential cause(s) of late graft loss. Bioinformatic analyses showed that highly relevant immunological pathways were activated in the animal which experienced late graft failure. Further connectivity analyses revealed that the activation of T cell signaling pathways was the most prominent, suggesting that T cell-mediated graft rejection could be the cause of the late-phase islet loss. Indeed, the porcine islets in the biopsied monkey liver samples were heavily infiltrated with CD3+ T cells. Furthermore, hypothesis test using a computational experiment reinforced our conclusion. Taken together, we suggest that bioinformatics analyses with peripheral blood RNA-seq could unveil the cause of insidious late islet graft loss.
Collapse
Affiliation(s)
- Hyun-Je Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Republic of Korea
- Department of Dermatology and the Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ji Hwan Moon
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 14260, USA
| | - Hyunwoo Chung
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Republic of Korea
| | - Jun-Seop Shin
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Bongi Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jong-Min Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jung-Sik Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Il-Hee Yoon
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Byoung-Hoon Min
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seong-Jun Kang
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Republic of Korea
| | - Yong-Hee Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Kyuri Jo
- Department of Computer Engineering, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Joungmin Choi
- Division of Computer Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Heejoon Chae
- Division of Computer Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Won-Woo Lee
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea.
- Bioinformatics Institute, Department of Computer Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Department of Computer Science & Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| |
Collapse
|
80
|
Eeda V, Herlea-Pana O, Lim HY, Wang W. Discovery of N-(2-(Benzylamino)-2-oxoethyl)benzamide analogs as a novel scaffold of pancreatic β-cell protective agents against endoplasmic reticulum stress. Chem Biol Drug Des 2019; 95:388-393. [PMID: 31755655 DOI: 10.1111/cbdd.13650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/25/2019] [Accepted: 11/10/2019] [Indexed: 12/30/2022]
Abstract
Endoplasmic reticulum (ER) stress-induced pancreatic β-cell dysfunction and death play important roles in the development of diabetes. The 1,2,3-triazole derivative 1 is one of only a few structures that have thus far been identified that protect β cells against ER stress. However, this compound has narrow activity range and limited aqueous solubility. To overcome these, we designed and synthesized a new scaffold in which the triazole pharmacophore was substituted with a glycine-like amino acid. Structure-activity relationship studies on this scaffold identified a N-(2-(Benzylamino)-2-oxoethyl)benzamide analog WO5m that possesses β-cell protective activity against ER stress with much improved potency (maximal activity at 100% with EC50 at 0.1 ± 0.01 µm) and water solubility. Identification of this novel β-cell protective scaffold thus provides a new promising modality for the treatment of diabetes.
Collapse
Affiliation(s)
- Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Hui-Ying Lim
- Department of Physiology, The University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| |
Collapse
|
81
|
Men L, Sun J, Luo G, Ren D. Acute Deletion of METTL14 in β-Cells of Adult Mice Results in Glucose Intolerance. Endocrinology 2019; 160:2388-2394. [PMID: 31369074 PMCID: PMC6760293 DOI: 10.1210/en.2019-00350] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/26/2019] [Indexed: 01/11/2023]
Abstract
N6-Methyladenosine (m6A) is the most common and abundant mRNA modification that involves regulating the RNA metabolism. However, the role of m6A in regulating the β-cell function is unclear. Methyltransferase-like 14 (METTL14) is a key component of the m6A methyltransferase complex. To define the role of m6A in regulating the β-cell function, we generated β-cell METTL14-specific knockout (βKO) mice by tamoxifen administration. Acute deletion of Mettl14 in β-cells results in glucose intolerance as a result of a reduction in insulin secretion in β-cells even though β-cell mass is increased, which is related to increased β-cell proliferation. To define the molecular mechanism, we performed RNA sequencing to detect the gene expression in βKO islets. The genes responsible for endoplasmic reticulum stress, such as Ire1α, were among the top upregulated genes. Both mRNA and protein levels of IRE1α and spliced X-box protein binding 1 (sXBP-1) were increased in βKO islets. The protein levels of proinsulin and insulin were decreased in βKO islets. These results suggest that acute METTL14 deficiency in β-cells induces glucose intolerance by increasing the IRE1α/sXBP-1 pathway.
Collapse
Affiliation(s)
- Lili Men
- Department of Endocrinology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Juan Sun
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Guanzheng Luo
- School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Decheng Ren
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Correspondence: Decheng Ren, PhD, Eli Lilly and Company, 893 Delaware Street, Indianapolis, Indiana 46225. E-mail:
| |
Collapse
|
82
|
Abstract
The loss of functional beta cell mass characterises all forms of diabetes. Beta cells are highly susceptible to stress, including cytokine, endoplasmic reticulum (ER) and oxidative stress. This study examined the role of pleckstrin homology-like, domain family A, member 3 (Phlda3) in beta cell survival under stress conditions and the regulatory basis. We found that the mRNA levels of Phlda3 were markedly upregulated in vivo in the islets of diabetic humans and mice. In vitro, exposure of MIN6 cells or islets to cytokines, palmitate, thapsigargin or ribose upregulated Phlda3 mRNA and protein levels, concurrent with the induction of ER stress (Ddit3 and Trb3) and antioxidant (Hmox1) genes. Furthermore, H2O2 treatment markedly increased PHLDA3 immunostaining in human islets. Phlda3 expression was differentially regulated by adaptive (Xbp1) and apoptotic (Ddit3) unfolded protein response (UPR) mediators. siRNA-mediated knockdown of Xbp1 inhibited the induction of Phlda3 by cytokines and palmitate, whereas knockdown of Ddit3 upregulated Phlda3. Moreover, knockdown of Phlda3 potentiated cytokine-induced apoptosis in association with upregulation of inflammatory genes (iNos, IL1β and IκBα) and NFκB phosphorylation and downregulation of antioxidant (Gpx1 and Srxn1) and adaptive UPR (Xbp1, Hspa5 and Fkbp11) genes. Knockdown of Phlda3 also potentiated apoptosis under oxidative stress conditions induced by ribose treatment. These findings suggest that Phlda3 is crucial for beta cell survival under stress conditions. Phlda3 regulates the cytokine, oxidative and ER stress responses in beta cells via the repression of inflammatory gene expression and the maintenance of antioxidant and adaptive UPR gene expression. Phlda3 may promote beta cell survival in diabetes.
Collapse
|
83
|
Aslam AS, Fuwad A, Ryu H, Selvaraj B, Song JW, Kim DW, Kim SM, Lee JW, Jeon TJ, Cho DG. Synthetic Anion Transporters as Endoplasmic Reticulum (ER) Stress Inducers. Org Lett 2019; 21:7828-7832. [DOI: 10.1021/acs.orglett.9b02823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Adil S. Aslam
- Department of Chemistry and Chemical Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Ahmed Fuwad
- Department of Mechanical Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Hyunil Ryu
- Department of Biological Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Baskar Selvaraj
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung Institute, Gangneung 25451, Republic of Korea
| | - Jae-Won Song
- Department of Chemistry and Chemical Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Science, College of Dentistry, Gangneung Wonju National University, Gangneung 25457, Republic of Korea
| | - Sun Min Kim
- Department of Mechanical Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jae Wook Lee
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung Institute, Gangneung 25451, Republic of Korea
| | - Tae-Joon Jeon
- Department of Biological Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Gyu Cho
- Department of Chemistry and Chemical Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
84
|
Jacobson DA, Shyng SL. Ion Channels of the Islets in Type 2 Diabetes. J Mol Biol 2019; 432:1326-1346. [PMID: 31473158 DOI: 10.1016/j.jmb.2019.08.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
Abstract
Ca2+ is an essential signal for pancreatic β-cell function. Ca2+ plays critical roles in numerous β-cell pathways such as insulin secretion, transcription, metabolism, endoplasmic reticulum function, and the stress response. Therefore, β-cell Ca2+ handling is tightly controlled. At the plasma membrane, Ca2+ entry primarily occurs through voltage-dependent Ca2+ channels. Voltage-dependent Ca2+ channel activity is dependent on orchestrated fluctuations in the plasma membrane potential or voltage, which are mediated via the activity of many ion channels. During the pathogenesis of type 2 diabetes the β-cell is exposed to stressful conditions, which result in alterations of Ca2+ handling. Some of the changes in β-cell Ca2+ handling that occur under stress result from perturbations in ion channel activity, expression or localization. Defective Ca2+ signaling in the diabetic β-cell alters function, limits insulin secretion and exacerbates hyperglycemia. In this review, we focus on the β-cell ion channels that control Ca2+ handling and how they impact β-cell dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7415 MRB4 (Langford), 2213 Garland Avenue, Nashville, TN 37232, USA.
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, L224, MRB 624, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
85
|
Zhu Y, Zhang X, Zhang L, Zhang M, Li L, Luo D, Zhong Y. Perilipin5 protects against lipotoxicity and alleviates endoplasmic reticulum stress in pancreatic β-cells. Nutr Metab (Lond) 2019; 16:50. [PMID: 31384284 PMCID: PMC6668071 DOI: 10.1186/s12986-019-0375-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 07/19/2019] [Indexed: 12/27/2022] Open
Abstract
Background Chronic exposure of pancreatic β-cells to excess free fatty acids is thought to contribute to type 2 diabetes pathogenesis in obesity by impairing β-cell function and even leading to apoptosis. In β-cells, lipid droplet-associated protein perilipin 5 (PLIN5) has been shown to enhance insulin secretion by regulating intracellular lipid metabolism; the roles of PLIN5 in response to lipotoxicity remain poorly understood. Methods INS-1 β-cells were transfected with PLIN5-overexpression adenovirus (Ad-PLIN5) and treated with palmitate. C57BL/6 J male mice were fed with high fat diet and tail intravenous injected with adeno-associated virus overexpressing PLIN5 (AAV-PLIN5) in β-cells. Results Our data showed that palmitate and PPAR agonists including WY14643 (PPARα), GW501516 (PPARβ/δ), rosiglitazone (PPARγ) in vitro all induced PLIN5 expression in INS-1 cells. Under palmitate overload, although upregulating PLIN5 promoted lipid droplet storage, it alleviated lipotoxicity in INS-1 β-cells with improved cell viability, cell apoptosis and β-cell function. The protection role of PLIN5 in β-cell function observed in cell experiments were further verified in in vivo study indicated by mitigated glucose intolerance in high fat diet fed mice with β-cell-specific overexpression of PLIN5. Mechanistic experiments revealed that enhanced FAO induced by elevation of PLIN5, followed by decreased ER stress may be a major mechanism responsible for alleviation of lipotoxicity observed in the present study. Conclusions Our finding substantiated the important role of PLIN5 in protection against lipotoxicity in β-cells. Electronic supplementary material The online version of this article (10.1186/s12986-019-0375-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunxia Zhu
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| | - Xiaoyan Zhang
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| | - Li Zhang
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| | - Mingliang Zhang
- 2Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233 China
| | - Ling Li
- 3Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Deng Luo
- 4Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Yuan Zhong
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| |
Collapse
|
86
|
Panganiban RA, Park HR, Sun M, Shumyatcher M, Himes BE, Lu Q. Genome-wide CRISPR screen identifies suppressors of endoplasmic reticulum stress-induced apoptosis. Proc Natl Acad Sci U S A 2019; 116:13384-13393. [PMID: 31213543 PMCID: PMC6613086 DOI: 10.1073/pnas.1906275116] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sensing misfolded proteins in the endoplasmic reticulum (ER), cells initiate the ER stress response and, when overwhelmed, undergo apoptosis. However, little is known about how cells prevent excessive ER stress response and cell death to restore homeostasis. Here, we report the identification and characterization of cellular suppressors of ER stress-induced apoptosis. Using a genome-wide CRISPR library, we screen for genes whose inactivation further increases ER stress-induced up-regulation of C/EBP homologous protein 10 (CHOP)-the transcription factor central to ER stress-associated apoptosis. Among the top validated hits are two interacting components of the polycomb repressive complex (L3MBTL2 [L(3)Mbt-Like 2] and MGA [MAX gene associated]), and microRNA-124-3 (miR-124-3). CRISPR knockout of these genes increases CHOP expression and sensitizes cells to apoptosis induced by multiple ER stressors, while overexpression confers the opposite effects. L3MBTL2 associates with the CHOP promoter in unstressed cells to repress CHOP induction but dissociates from the promoter in the presence of ER stress, whereas miR-124-3 directly targets the IRE1 branch of the ER stress pathway. Our study reveals distinct mechanisms that suppress ER stress-induced apoptosis and may lead to a better understanding of diseases whose pathogenesis is linked to overactive ER stress response.
Collapse
Affiliation(s)
- Ronald A Panganiban
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Hae-Ryung Park
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Maoyun Sun
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115;
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| |
Collapse
|
87
|
Arunagiri A, Haataja L, Pottekat A, Pamenan F, Kim S, Zeltser LM, Paton AW, Paton JC, Tsai B, Itkin-Ansari P, Kaufman RJ, Liu M, Arvan P. Proinsulin misfolding is an early event in the progression to type 2 diabetes. eLife 2019; 8:44532. [PMID: 31184302 PMCID: PMC6559786 DOI: 10.7554/elife.44532] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Biosynthesis of insulin – critical to metabolic homeostasis – begins with folding of the proinsulin precursor, including formation of three evolutionarily conserved intramolecular disulfide bonds. Remarkably, normal pancreatic islets contain a subset of proinsulin molecules bearing at least one free cysteine thiol. In human (or rodent) islets with a perturbed endoplasmic reticulum folding environment, non-native proinsulin enters intermolecular disulfide-linked complexes. In genetically obese mice with otherwise wild-type islets, disulfide-linked complexes of proinsulin are more abundant, and leptin receptor-deficient mice, the further increase of such complexes tracks with the onset of islet insulin deficiency and diabetes. Proinsulin-Cys(B19) and Cys(A20) are necessary and sufficient for the formation of proinsulin disulfide-linked complexes; indeed, proinsulin Cys(B19)-Cys(B19) covalent homodimers resist reductive dissociation, highlighting a structural basis for aberrant proinsulin complex formation. We conclude that increased proinsulin misfolding via disulfide-linked complexes is an early event associated with prediabetes that worsens with ß-cell dysfunction in type two diabetes. Our body fine-tunes the amount of sugar in our blood thanks to specialized ‘beta cells’ in the pancreas, which can release a hormone called insulin. To produce insulin, the beta cells first need to build an early version of the molecule – known as proinsulin – inside a cellular compartment called the endoplasmic reticulum. This process involves the formation of internal staples that keep the molecule of proinsulin folded correctly. Individuals developing type 2 diabetes have spikes of sugar in their blood, and so their bodies often respond by trying to make large amounts of insulin. After a while, the beta cells can fail to keep up, which brings on the full-blown disease. However, scientists have discovered that early in type 2 diabetes, the endoplasmic reticulum of beta cells can already show signs of stress; yet, the exact causes of this early damage are still unknown. To investigate this, Arunagiri et al. looked into whether proinsulin folds correctly during the earliest stages of type 2 diabetes. Biochemical experiments showed that even healthy beta cells contained some misfolded proinsulin molecules, where the molecular staples that should fold proinsulin internally were instead abnormally linking proinsulin molecules together. Further work revealed that the misfolded proinsulin was accumulating inside the endoplasmic reticulum. Finally, obese mice that were in the earliest stages of type 2 diabetes had the highest levels of abnormal proinsulin in their beta cells. Overall, the work by Arunagiri et al. suggests that large amounts of proinsulin molecules stapling themselves to each other in the endoplasmic reticulum of beta cells could be an early hallmark of the disease, and could make it get worse. A separate study by Jang et al. also shows that a protein that limits the misfolding of proinsulin is key to maintain successful insulin production in animals eating a Western-style, high fat diet. Hundreds of millions of people around the world have type 2 diabetes, and this number is rising quickly. Detecting and then fixing early problems associated with the condition may help to stop the disease in its track.
Collapse
Affiliation(s)
- Anoop Arunagiri
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Anita Pottekat
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Fawnnie Pamenan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| | - Soohyun Kim
- Department of Biomedical Science and Technology, Konkuk University, Gwangjin-gu, Republic of Korea
| | - Lori M Zeltser
- Department of Pathology and Cell Biology, Naomi Berrie Diabetes Center, Columbia University, New York, United States
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Pamela Itkin-Ansari
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, United States
| | - Ming Liu
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States.,Department of Endocrinology and Metabolism, Tianjin Medical University, Tianjin, China
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
88
|
Lalwani A, Warren J, Liuwantara D, Hawthorne WJ, O'Connell PJ, Gonzalez FJ, Stokes RA, Chen J, Laybutt DR, Craig ME, Swarbrick MM, King C, Gunton JE. β Cell Hypoxia-Inducible Factor-1α Is Required for the Prevention of Type 1 Diabetes. Cell Rep 2019; 27:2370-2384.e6. [PMID: 31116982 PMCID: PMC6661122 DOI: 10.1016/j.celrep.2019.04.086] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 01/31/2019] [Accepted: 04/18/2019] [Indexed: 12/28/2022] Open
Abstract
The development of autoimmune disease type 1 diabetes (T1D) is determined by both genetic background and environmental factors. Environmental triggers include RNA viruses, particularly coxsackievirus (CV), but how they induce T1D is not understood. Here, we demonstrate that deletion of the transcription factor hypoxia-inducible factor-1α (HIF-1α) from β cells increases the susceptibility of non-obese diabetic (NOD) mice to environmentally triggered T1D from coxsackieviruses and the β cell toxin streptozotocin. Similarly, knockdown of HIF-1α in human islets leads to a poorer response to coxsackievirus infection. Studies in coxsackievirus-infected islets demonstrate that lack of HIF-1α leads to impaired viral clearance, increased viral load, inflammation, pancreatitis, and loss of β cell mass. These findings show an important role for β cells and, specifically, lack of β cell HIF-1α in the development of T1D. These data suggest new strategies for the prevention of T1D.
Collapse
Affiliation(s)
- Amit Lalwani
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Joanna Warren
- Mucosal Autoimmunity, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - David Liuwantara
- National Pancreas Transplant Unit (NPTU), Westmead Hospital, Sydney, NSW, Australia
| | - Wayne J Hawthorne
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; National Pancreas Transplant Unit (NPTU), Westmead Hospital, Sydney, NSW, Australia
| | - Philip J O'Connell
- National Pancreas Transplant Unit (NPTU), Westmead Hospital, Sydney, NSW, Australia
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD, USA
| | - Rebecca A Stokes
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia
| | - Jennifer Chen
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia
| | - D Ross Laybutt
- Islet Biology, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Maria E Craig
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; The Children's Hospital at Westmead, Sydney, NSW, Australia; School of Women's and Children's Health, University of New South Wales, Kensington, NSW, Australia
| | - Michael M Swarbrick
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Kensington, NSW, Australia
| | - Cecile King
- Mucosal Autoimmunity, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Jenny E Gunton
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; St Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia; Department of Diabetes and Endocrinology, Westmead Hospital, Sydney, NSW, Australia.
| |
Collapse
|
89
|
Lee ES, Kim JS, Lee H, Ryu JY, Lee HJ, Sonn JK, Lim YB. Auranofin, an Anti-rheumatic Gold Drug, Aggravates the Radiation-Induced Acute Intestinal Injury in Mice. Front Pharmacol 2019; 10:417. [PMID: 31105565 PMCID: PMC6492527 DOI: 10.3389/fphar.2019.00417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/02/2019] [Indexed: 12/20/2022] Open
Abstract
Pelvic and abdominal radiotherapy plays an important role in eradication of malignant cells; however, it also results in slight intestinal injury. The apoptosis of cells in the intestinal epithelium is a primary pathological factor that initiates radiation-induced intestinal injury. Auranofin, a gold-containing triethylphosphine, was approved for the treatment of rheumatoid arthritis, and its therapeutic application has been expanded to a number of other diseases, such as parasitic infections, neurodegenerative disorders, AIDS, and bacterial infections. Recently, a treatment strategy combining the use of auranofin and ionizing radiation aimed at increasing the radiosensitivity of cancer cells was proposed for improving the control of local cancers. In this study, we evaluated the effect of auranofin on the radiosensitivity of intestinal epithelial cells. The treatment with a combination of 1 μM auranofin and 5 Gy ionizing radiation showed clear additive effects on caspase 3 cleavage and apoptotic DNA fragmentation in IEC-6 cells, and auranofin administration significantly aggravated the radiation-induced intestinal injury in mice. Auranofin treatment also resulted in the activation of the unfolded protein response and in the inhibition of thioredoxin reductase, which is a key component of the cellular antioxidant system. Pre-treatment with N-acetyl cysteine, a well-known scavenger of reactive oxygen species, but not with a chemical chaperone, which inhibits endoplasmic reticulum stress and the ensuing unfolded protein response, significantly reduced the radiosensitizing effects of auranofin in the IEC-6 cells. In addition, transfection of IEC-6 cells with a small interfering RNA targeted against thioredoxin reductase significantly enhanced the radiosensitivity of these cells. These results suggest that auranofin-induced radiosensitization of intestinal epithelial cells is mediated through oxidative stress caused by the deregulation of thioredoxin redox system, and auranofin treatment can be an independent risk factor for the development of acute pelvic radiation disease.
Collapse
Affiliation(s)
- Eun Sang Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Joong Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Hyounji Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jee-Yeon Ryu
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jong Kyung Sonn
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea
| | - Young-Bin Lim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| |
Collapse
|
90
|
Preliminary Studies of the Impact of CXCL12 on the Foreign Body Reaction to Pancreatic Islets Microencapsulated in Alginate in Nonhuman Primates. Transplant Direct 2019; 5:e447. [PMID: 31165082 PMCID: PMC6511446 DOI: 10.1097/txd.0000000000000890] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022] Open
Abstract
Supplemental Digital Content is available in the text. Background. We previously demonstrated that the incorporation of the chemokine CXCL12 into alginate microbeads supported long-term survival of microencapsulated auto-, allo-, and xenogeneic islets in murine models of diabetes without systemic immune suppression. The purpose of this study was to test whether CXCL12 could abrogate foreign body responses (FBRs) against alginate microbeads which were empty or contained autologous islets in healthy nonhuman primates (NHPs; n = 4). Methods. Two NHPs received intraperitoneal implants of 400 000 alginate microbeads with or without CXCL12, and postimplantation immunological and histopathological changes were evaluated up to 6 months postimplantation. A similar evaluation of autologous islets in CXCL12-containing alginate microbeads was performed in NHPs (n = 2). Results. CXCL12-containing alginate microbeads were associated with a markedly reduced FBR to microbeads. Host responses to microbead implants were minimal, as assessed by clinical observations, blood counts, and chemistry. Evaluation of encapsulated islets was limited by the development of necrotizing pancreatitis after hemipancreatectomy in 1 NHP. A limited number of functioning islets were detectable at 6 months posttransplantation in the second NHP. In general, empty microbeads or islet-containing beads were found to be evenly distributed through the intraperitoneal cavity and did not accumulate in the Pouch of Douglas. Conclusions. Inclusion of CXCL12 in alginate microbeads minimized localized FBR. The NHP autologous islet implant model had limited utility for excluding inflammatory/immune responses to implanted islets because of the complexity of pancreatic surgery (hemipancreatectomy) before transplantation and the need to microencapsulate and transplant encapsulated autologous islets immediately after pancreatectomy and islet isolation.
Collapse
|
91
|
Karunakaran U, Elumalai S, Moon JS, Jeon JH, Kim ND, Park KG, Won KC, Leem J, Lee IK. Myricetin Protects Against High Glucose-Induced β-Cell Apoptosis by Attenuating Endoplasmic Reticulum Stress via Inactivation of Cyclin-Dependent Kinase 5. Diabetes Metab J 2019; 43:192-205. [PMID: 30688049 PMCID: PMC6470101 DOI: 10.4093/dmj.2018.0052] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/16/2018] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Chronic hyperglycemia has deleterious effects on pancreatic β-cell function and turnover. Recent studies support the view that cyclin-dependent kinase 5 (CDK5) plays a role in β-cell failure under hyperglycemic conditions. However, little is known about how CDK5 impair β-cell function. Myricetin, a natural flavonoid, has therapeutic potential for the treatment of type 2 diabetes mellitus. In this study, we examined the effect of myricetin on high glucose (HG)-induced β-cell apoptosis and explored the relationship between myricetin and CDK5. METHODS To address this question, we subjected INS-1 cells and isolated rat islets to HG conditions (30 mM) in the presence or absence of myricetin. Docking studies were conducted to validate the interaction between myricetin and CDK5. Gene expression and protein levels of endoplasmic reticulum (ER) stress markers were measured by real-time reverse transcription polymerase chain reaction and Western blot analysis. RESULTS Activation of CDK5 in response to HG coupled with the induction of ER stress via the down regulation of sarcoendoplasmic reticulum calcium ATPase 2b (SERCA2b) gene expression and reduced the nuclear accumulation of pancreatic duodenal homeobox 1 (PDX1) leads to β-cell apoptosis. Docking study predicts that myricetin inhibit CDK5 activation by direct binding in the ATP-binding pocket. Myricetin counteracted the decrease in the levels of PDX1 and SERCA2b by HG. Moreover, myricetin attenuated HG-induced apoptosis in INS-1 cells and rat islets and reduce the mitochondrial dysfunction by decreasing reactive oxygen species production and mitochondrial membrane potential (ΔΨm) loss. CONCLUSION Myricetin protects the β-cells against HG-induced apoptosis by inhibiting ER stress, possibly through inactivation of CDK5 and consequent upregulation of PDX1 and SERCA2b.
Collapse
Affiliation(s)
- Udayakumar Karunakaran
- Department of Biomedical Science, Graduate School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Suma Elumalai
- Institute of Medical Science, Yeungnam University College of Medicine, Daegu, Korea
| | - Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jae Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea
| | - Nam Doo Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Keun Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea
| | - Kyu Chang Won
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu, Korea.
| | - In Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.
| |
Collapse
|
92
|
Gao Y, Wu X, Zhao S, Zhang Y, Ma H, Yang Z, Yang W, Zhao C, Wang L, Zhang Q. Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells. Cell Mol Biol Lett 2019; 24:21. [PMID: 30915128 PMCID: PMC6416941 DOI: 10.1186/s11658-019-0147-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Melatonin receptors MT1 and MT2 (genes officially named MTNR1A and MTNR1B, respectively) play crucial roles in melatonin-mediated regulation of circadian rhythms, the immune system, and control of reproduction in seasonally breeding animals. In this study, immunolocalization assay showed that MT1 and MT2 are highly expressed in Leydig cell membrane. To understand the biological function of melatonin receptors in hCG-induced testosterone synthesis, we generated melatonin receptor knockdown cells using specific siRNA and performed testosterone detection after hCG treatment. We found that knockdown of melatonin receptors, especially MTNR1A, led to an obvious decrease (> 60%) of testosterone level. Our further study revealed that knockdown of melatonin receptors repressed expression, at both the mRNA level and the protein level, of key steroidogenic genes, such as p450scc, p450c17 and StAR, which are essential for testosterone synthesis. hCG triggered endoplasmic reticulum (ER) stress to regulate steroidogenic genes' expression and apoptosis. To further investigate the potential roles of melatonin receptors in hCG-induced regulation of ER stress and apoptosis, we examined expression of some crucial ER stress markers, including Grp78, Chop, ATF4, Xbp1, and IRE1. We found that inhibition of melatonin receptors increased hCG-induced expression of Grp78, Chop and ATF4, but not Xbp1 and IRE1, suggesting that hCG may modulate IRE1 signaling pathways in a melatonin receptor-dependent manner. In addition, our further data showed that knockdown of MTNR1A and MTNR1B promoted hCG-induced expression of apoptosis markers, including p53, caspase-3 and Bcl-2. These results suggested that the melatonin receptors MTNR1A and MTNR1B are essential to repress hCG-induced ER stress and cell apoptosis. Our studies demonstrated that the mammalian melatonin receptors MT1 and MT2 are involved in testosterone synthesis via mediating multiple cell pathways.
Collapse
Affiliation(s)
- Yuan Gao
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Xiaochun Wu
- 2College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu China
| | - Shuqin Zhao
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Yujun Zhang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Hailong Ma
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Zhen Yang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Wanghao Yang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Chen Zhao
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Li Wang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Quanwei Zhang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China.,2College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu China
| |
Collapse
|
93
|
Hironaka JY, Kitahama S, Sato H, Inoue M, Takahashi T, Tamori Y. Sleeve Gastrectomy Induced Remission of Slowly Progressive Type 1 Diabetes in a Morbidly Obese Japanese Patient. Intern Med 2019; 58:675-678. [PMID: 30333397 PMCID: PMC6443562 DOI: 10.2169/internalmedicine.1217-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The effects of bariatric/metabolic surgery on glycemic control in obese type 1 diabetic patients are controversial. We herein report a case of a morbidly obese 35-year-old woman who completely recovered from slowly progressive type 1 diabetes (SPIDDM) following laparoscopic sleeve gastrectomy. Preoperatively, her body mass index (BMI) was 49.8 kg/m2 and hemoglobin A1c was 5.7% with intensive insulin therapy. Six months after bariatric/metabolic surgery, her BMI decreased to 33.2 kg/m2 and her glycemic control was normal despite the discontinuation of all diabetic medicine. This case demonstrates the usefulness of bariatric/metabolic surgery for achieving glycemic control in morbidly obese patients with SPIDDM in Japan.
Collapse
Affiliation(s)
- Jun-Ya Hironaka
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Chibune General Hospital, Japan
| | - Seiichi Kitahama
- Department of Bariatric and Metabolic Surgery, Chibune General Hospital, Japan
| | - Hiroyuki Sato
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Chibune General Hospital, Japan
| | - Maki Inoue
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Chibune General Hospital, Japan
| | - Tetsuya Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Chibune General Hospital, Japan
| | - Yoshikazu Tamori
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Chibune General Hospital, Japan
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| |
Collapse
|
94
|
Tian H, Li Y, Kang P, Wang Z, Yue F, Jiao P, Yang N, Qin S, Yao S. Endoplasmic reticulum stress-dependent autophagy inhibits glycated high-density lipoprotein-induced macrophage apoptosis by inhibiting CHOP pathway. J Cell Mol Med 2019; 23:2954-2969. [PMID: 30746902 PMCID: PMC6433656 DOI: 10.1111/jcmm.14203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/07/2018] [Accepted: 01/13/2019] [Indexed: 12/15/2022] Open
Abstract
This study was designed to explore the inductive effect of glycated high‐density lipoprotein (gly‐HDL) on endoplasmic reticulum (ER) stress‐C/EBP homologous protein (CHOP)‐mediated macrophage apoptosis and its relationship with autophagy. Our results showed that gly‐HDL caused macrophage apoptosis with concomitant activation of ER stress pathway, including nuclear translocation of activating transcription factor 6, phosphorylation of protein kinase‐like ER kinase (PERK) and eukaryotic translation initiation factor 2α, and CHOP up‐regulation, which were inhibited by 4‐phenylbutyric acid (PBA, an ER stress inhibitor) and the gene silencing of PERK and CHOP. Similar data were obtained from macrophages treated by HDL isolated from diabetic patients. Gly‐HDL induced macrophage autophagy as assessed by up‐regulation of beclin‐1, autophagy‐related gene 5 and microtubule‐associated protein one light chain 3‐II, which were depressed by PBA and PERK siRNA. Gly‐HDL‐induced apoptosis, PERK phosphorylation and CHOP up‐regulation were suppressed by rapamycin (an autophagy inducer), whereas aggravated by 3‐methyladenine (an autophagy inhibitor) and beclin‐1 siRNA. Administration of diabetic apoE−/− mice with rapamycin attenuated MOMA‐2 and CHOP up‐regulation and apoptosis in atherosclerotic lesions. These data indicate that gly‐HDL may induce macrophage apoptosis through activating ER stress‐CHOP pathway and ER stress mediates gly‐HDL‐induced autophagy, which in turn protects macrophages against apoptosis by alleviating CHOP pathway.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Yanyan Li
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Panpan Kang
- Affiliated hospital of Chengde Medical University, Chengde Medical University, Chengde, China
| | - Zhichao Wang
- College of Nursing, Taishan Medical University, Taian, China
| | - Feng Yue
- Department of Endocrinology, Central Hospital of Taian, Taian, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Nana Yang
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China.,College of Basic Medical Sciences, Taishan Medical University, Taian, China
| |
Collapse
|
95
|
Imam S, Prathibha R, Dar P, Almotah K, Al-Khudhair A, Hasan SAM, Salim N, Jilani TN, Mirmira RG, Jaume JC. eIF5A inhibition influences T cell dynamics in the pancreatic microenvironment of the humanized mouse model of Type 1 Diabetes. Sci Rep 2019; 9:1533. [PMID: 30733517 PMCID: PMC6367423 DOI: 10.1038/s41598-018-38341-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022] Open
Abstract
We have developed a transgenic mouse model of Type 1 Diabetes (T1D) in which human GAD65 is expressed in pancreatic β-cells, and human MHC-II is expressed on antigen presenting cells. Induced GAD65 antigen presentation activates T-cells, which initiates the downstream events leading to diabetes. In our humanized mice, we have shown downregulation of eukaryotic translation initiation factor 5 A (elF5A), expressed only in actively dividing mammalian cells. In-vivo inhibition of elF5A hypusination by deoxyhypusine synthase (DHS) inhibitor "GC7" was studied; DHS inhibitor alters the pathophysiology in our mouse model by catalyzing the crucial hypusination and the rate-limiting step of elF5A activation. In our mouse model, we have shown that inhibition of eIF5A resets the pro-inflammatory bias in the pancreatic microenvironment. There was: (a) reduction of Th1/Th17 response, (b) an increase in Treg numbers, (c) debase in IL17 and IL21 cytokines levels in serum, (d) lowering of anti-GAD65 antibodies, and (e) ablation of the ER stress that improved functionality of the β-cells, but minimal effect on the cytotoxic CD8 T-cell (CTL) mediated response. Conclusively, immune modulation, in the case of T1D, may help to manipulate inflammatory responses, decreasing disease severity, and may help manage T1D in early stages of disease. Our study also demonstrates that without manipulating the CTLs mediated response extensively, it is difficult to treat T1D.
Collapse
Affiliation(s)
- Shahnawaz Imam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| | - R Prathibha
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Pervaiz Dar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Shuhama, Srinagar, 190006, Jammu and Kashmir, India
| | - Khalil Almotah
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Ahmed Al-Khudhair
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Syed Abdul-Moiz Hasan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Nancy Salim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Talha Naser Jilani
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Juan Carlos Jaume
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research (CeDER), Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
96
|
Chan KP, Chao SH, Kah JCY. Enhanced Secretion of Functional Insulin with DNA-Functionalized Gold Nanoparticles in Cells. ACS Biomater Sci Eng 2019; 5:1602-1610. [DOI: 10.1021/acsbiomaterials.9b00032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Kian Ping Chan
- National University of Singapore Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, University Hall, Tan Chin Tuan Wing, Level 04, #04-02, 21 Lower Kent Ridge, Singapore 119077
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, #06-01 Centros, Singapore 138668
| | - Sheng-Hao Chao
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, 20 Biopolis Way, #06-01 Centros, Singapore 138668
- Department of Microbiology and Immunology, National University of Singapore, 5 Science Drive 2, Blk MD4, Level 3, Singapore 117597
| | - James Chen Yong Kah
- National University of Singapore Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, University Hall, Tan Chin Tuan Wing, Level 04, #04-02, 21 Lower Kent Ridge, Singapore 119077
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Blk E4, #04-08, Singapore 117583
| |
Collapse
|
97
|
Bugliani M, Mossuto S, Grano F, Suleiman M, Marselli L, Boggi U, De Simone P, Eizirik DL, Cnop M, Marchetti P, De Tata V. Modulation of Autophagy Influences the Function and Survival of Human Pancreatic Beta Cells Under Endoplasmic Reticulum Stress Conditions and in Type 2 Diabetes. Front Endocrinol (Lausanne) 2019; 10:52. [PMID: 30863363 PMCID: PMC6399112 DOI: 10.3389/fendo.2019.00052] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/21/2019] [Indexed: 01/09/2023] Open
Abstract
Autophagy is the major mechanism involved in degradation and recycling of intracellular components, and its alterations have been proposed to cause beta cell dysfunction. In this study, we explored the effects of autophagy modulation in human islets under conditions associated to endoplasmic reticulum (ER) stress. Human pancreatic islets were isolated by enzymatic digestion and density gradient purification from pancreatic samples of non-diabetic (ND; n = 17; age 65 ± 21 years; gender: 5 M/12 F; BMI 23.4 ± 3.3 kg/m2) and T2D (n = 9; age 76 ± 6 years; 4 M/5 F; gender: BMI 25.4 ± 3.7 kg/m2) organ donors. Nine ND organ donors were treated for hypertension and 1 for both hypertension and hypercholesterolemia. T2D organ donors were treated with metformin (1), oral hypoglycemic agents (2), diet + oral hypoglycemic agents (3), insulin (3) or insulin plus metformin (3) as for antidiabetic therapy and, of these, 3 were treated also for hypertension and 6 for both hypertension and hypercholesterolemia. Two days after isolation, they were cultured for 1-5 days with 10 ng/ml rapamycin (autophagy inducer), 5 mM 3-methyladenine or 1.0 nM concanamycin-A (autophagy blockers), either in the presence or not of metabolic (0.5 mM palmitate) or chemical (0.1 ng/ml brefeldin A) ER stressors. In ND islets palmitate exposure induced a 4 to 5-fold increase of beta cell apoptosis, which was significantly prevented by rapamycin and exacerbated by 3-MA. Similar results were observed with brefeldin treatment. Glucose-stimulated insulin secretion from ND islets was reduced by palmitate (-40 to 50%) and brefeldin (-60 to 70%), and rapamycin counteracted palmitate, but not brefeldin, cytotoxic actions. Both palmitate and brefeldin induced PERK, CHOP and BiP gene expression, which was partially, but significantly prevented by rapamycin. With T2D islets, rapamycin alone reduced the amount of p62, an autophagy receptor that accumulates in cells when macroautophagy is inhibited. Compared to untreated T2D cells, rapamycin-exposed diabetic islets showed improved insulin secretion, reduced proportion of beta cells showing signs of apoptosis and better preserved insulin granules, mitochondria and ER ultrastructure; this was associated with significant reduction of PERK, CHOP and BiP gene expression. This study emphasizes the importance of autophagy modulation in human beta cell function and survival, particularly in situations of ER stress. Tuning autophagy could be a tool for beta cell protection.
Collapse
Affiliation(s)
- M. Bugliani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S. Mossuto
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - F. Grano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M. Suleiman
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L. Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - U. Boggi
- Department of Surgical Pathology, Medicine, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - P. De Simone
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - D. L. Eizirik
- Medical Faculty, ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - M. Cnop
- Medical Faculty, ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - P. Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - V. De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- *Correspondence: V. De Tata
| |
Collapse
|
98
|
Rowlands J, Heng J, Newsholme P, Carlessi R. Pleiotropic Effects of GLP-1 and Analogs on Cell Signaling, Metabolism, and Function. Front Endocrinol (Lausanne) 2018; 9:672. [PMID: 30532733 PMCID: PMC6266510 DOI: 10.3389/fendo.2018.00672] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The incretin hormone Glucagon-Like Peptide-1 (GLP-1) is best known for its "incretin effect" in restoring glucose homeostasis in diabetics, however, it is now apparent that it has a broader range of physiological effects in the body. Both in vitro and in vivo studies have demonstrated that GLP-1 mimetics alleviate endoplasmic reticulum stress, regulate autophagy, promote metabolic reprogramming, stimulate anti-inflammatory signaling, alter gene expression, and influence neuroprotective pathways. A substantial body of evidence has accumulated with respect to how GLP-1 and its analogs act to restore and maintain normal cellular functions. These findings have prompted several clinical trials which have reported GLP-1 analogs improve cardiac function, restore lung function and reduce mortality in patients with obstructive lung disease, influence blood pressure and lipid storage, and even prevent synaptic loss and neurodegeneration. Mechanistically, GLP-1 elicits its effects via acute elevation in cAMP levels, and subsequent protein kinase(s) activation, pathways well-defined in pancreatic β-cells which stimulate insulin secretion in conjunction with elevated Ca2+ and ATP. More recently, new studies have shed light on additional downstream pathways stimulated by chronic GLP-1 exposure, findings which have direct relevance to our understanding of the potential therapeutic effects of longer lasting analogs recently developed for clinical use. In this review, we provide a comprehensive description of the diverse roles for GLP-1 across multiple tissues, describe downstream pathways stimulated by acute and chronic exposure, and discuss novel pleiotropic applications of GLP-1 mimetics in the treatment of human disease.
Collapse
Affiliation(s)
| | | | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| | - Rodrigo Carlessi
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| |
Collapse
|
99
|
Gebert M, Bartoszewska S, Janaszak-Jasiecka A, Moszyńska A, Cabaj A, Króliczewski J, Madanecki P, Ochocka RJ, Crossman DK, Collawn JF, Bartoszewski R. PIWI proteins contribute to apoptosis during the UPR in human airway epithelial cells. Sci Rep 2018; 8:16431. [PMID: 30401887 PMCID: PMC6219583 DOI: 10.1038/s41598-018-34861-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/27/2018] [Indexed: 01/13/2023] Open
Abstract
Small noncoding microRNAs (miRNAs) post-transcriptionally regulate a large portion of the human transcriptome. miRNAs have been shown to play an important role in the unfolded protein response (UPR), a cellular adaptive mechanism that is important in alleviating endoplasmic reticulum (ER) stress and promoting cell recovery. Another class of small noncoding RNAs, the Piwi-interacting RNAs (piRNAs) together with PIWI proteins, was originally shown to play a role as repressors of germline transposable elements. More recent studies, however, indicate that P-element induced WImpy proteins (PIWI proteins) and piRNAs also regulate mRNA levels in somatic tissues. Using genome-wide small RNA next generation sequencing, cell viability assays, and caspase activity assays in human airway epithelial cells, we demonstrate that ER stress specifically up-regulates total piRNA expression profiles, and these changes correlate with UPR-induced apoptosis as shown by up-regulation of two pro-apoptotic factor mRNAs, CHOP and NOXA. Furthermore, siRNA knockdown of PIWIL2 and PIWIL4, two proteins involved in piRNA function, attenuates UPR-related cell death, inhibits piRNA expression, and inhibits the up-regulation of CHOP and NOXA mRNA expression. Hence, we provide evidence that PIWIL2 and PIWIL4 proteins, and potentially the up-regulated piRNAs, constitute a novel epigenetic mechanism that control cellular fate during the UPR.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Anna Janaszak-Jasiecka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Adrianna Moszyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Aleksandra Cabaj
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Jarosław Króliczewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Madanecki
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Renata J Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - David K Crossman
- Department of Genetics, Heflin Center for Genomic Science, University of Alabama at Birmingham, Birmingham, USA
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
100
|
Danilova T, Lindahl M. Emerging Roles for Mesencephalic Astrocyte-Derived Neurotrophic Factor (MANF) in Pancreatic Beta Cells and Diabetes. Front Physiol 2018; 9:1457. [PMID: 30386256 PMCID: PMC6198132 DOI: 10.3389/fphys.2018.01457] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/26/2018] [Indexed: 12/31/2022] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) was originally identified as a secreted trophic factor for dopamine neurons in vitro. It protects and restores damaged cells in rodent models of Parkinson's disease, brain and heart ischemia, spinocerebellar ataxia and retina in vivo. However, its exact mechanism of action is not known. MANF is widely expressed in most human and mouse organs with high levels in secretory tissues. Intracellularly, MANF localizes to the endoplasmic reticulum (ER) and ER stress increases it's expression in cells and tissues. Furthermore, increased MANF levels has been detected in the sera of young children with newly diagnosed Type 1 (T1D) diabetes and Type 2 (T2D) diabetic patients. ER stress is caused by the accumulation of misfolded and aggregated proteins in the ER. It activates a cellular defense mechanism, the unfolded protein response (UPR), a signaling cascade trying to restore ER homeostasis. However, if prolonged, unresolved ER stress leads to apoptosis. Unresolved ER stress contributes to the progressive death of pancreatic insulin-producing beta cells in both T1D and T2D. Diabetes mellitus is characterized by hyperglycemia, caused by the inability of the beta cells to maintain sufficient levels of circulating insulin. The current medications, insulin and antidiabetic drugs, alleviate diabetic symptoms but cannot reconstitute physiological insulin secretion which increases the risk of devastating vascular complications of the disease. Thus, one of the main strategies in improving current diabetes therapy is to define and validate novel approaches to protect beta cells from stress as well as activate their regeneration. Embryonic deletion of the Manf gene in mice led to gradual postnatal development of insulin-deficient diabetes caused by reduced beta cell proliferation and increased beta cell death due to increased and sustained ER stress. In vitro, recombinant MANF partly protected mouse and human beta cells from ER stress-induced beta cell death and potentiated mouse and human beta cell proliferation. Importantly, in vivo overexpression of MANF in the pancreas of T1D mice led to increased beta cell proliferation and decreased beta cell death, suggesting that MANF could be a new therapeutic candidate for beta cell protection and regeneration in diabetes.
Collapse
Affiliation(s)
- Tatiana Danilova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|