1
|
Karampelias C, Liu KC, Tengholm A, Andersson O. Mechanistic insights and approaches for beta cell regeneration. Nat Chem Biol 2025:10.1038/s41589-024-01822-y. [PMID: 39881214 DOI: 10.1038/s41589-024-01822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Diabetes is characterized by variable loss of insulin-producing beta cells, and new regenerative approaches to increasing the functional beta cell mass of patients hold promise for reversing disease progression. In this Review, we summarize recent chemical biology breakthroughs advancing our knowledge of beta cell regeneration. We present current chemical-based tools, sensors and mechanistic insights into pathways that can be targeted to enhance beta cell regeneration in model organisms. We group the pathways according to the cellular processes they affect, that is, proliferation, conversion of other mature cell types to beta cells and beta cell differentiation from progenitor-like populations. We also suggest assays for assessing the functionality of the regenerated beta cells. Although regeneration processes differ between animal models, such as zebrafish, mice and pigs, regenerative mechanisms identified in any one animal model may be translatable to humans. Overall, chemical biology-based approaches in beta cell regeneration give hope that specific molecular pathways can be targeted to enhance beta cell regeneration.
Collapse
Affiliation(s)
- Christos Karampelias
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Ka-Cheuk Liu
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Olov Andersson
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden.
| |
Collapse
|
2
|
Hecht JT, Veerisetty AC, Hossain MG, Patra D, Carrer M, Chiu F, Relic D, Jafar-nejad P, Posey KL. Loss of CHOP Prevents Joint Degeneration and Pain in a Mouse Model of Pseudoachondroplasia. Int J Mol Sci 2024; 26:16. [PMID: 39795874 PMCID: PMC11720453 DOI: 10.3390/ijms26010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025] Open
Abstract
Pseudoachondroplasia (PSACH), a severe dwarfing condition characterized by impaired skeletal growth and early joint degeneration, results from mutations in cartilage oligomeric matrix protein (COMP). These mutations disrupt normal protein folding, leading to the accumulation of misfolded COMP in chondrocytes. The MT-COMP mouse is a murine model of PSACH that expresses D469del human COMP in response to doxycycline and replicates the PSACH chondrocyte and clinical pathology. The basis for the mutant-COMP pathology involves endoplasmic reticulum (ER) stress signaling through the PERK/eIF2α/CHOP pathway. C/EBP homologous protein (CHOP), in conjunction with a TNFα inflammatory process, upregulates mTORC1, hindering autophagy clearance of mutant COMP protein. Life-long joint pain/degeneration diminishes quality of life, and treatments other than joint replacements are urgently needed. To assess whether molecules that reduce CHOP activity should be considered as a potential treatment for PSACH, we evaluated MT-COMP mice with 50% CHOP (MT-COMP/CHOP+/-), antisense oligonucleotide (ASO)-mediated CHOP knockdown, and complete CHOP ablation (MT-COMP/CHOP-/-). While earlier studies demonstrated that loss of CHOP in MT-COMP mice reduced intracellular retention, inflammation, and growth plate chondrocyte death, we now show that it did not normalize limb growth. ASO treatment reduced CHOP mRNA by approximately 60%, as measured by RT-qPCR, but did not improve limb length similar to MT-COMP/CHOP+/-. Interestingly, both 50% genetic reduction and complete loss of CHOP alleviated pain, while total ablation of CHOP in MT-COMP mice was necessary to preserve joint health. These results indicate that (1) CHOP reduction therapy is not an effective strategy for improving limb length and (2) pain and chondrocyte pathology are more responsive to intervention than the prevention of joint damage.
Collapse
Affiliation(s)
- Jacqueline T. Hecht
- Department of Pediatrics, McGovern Medical School UTHealth, Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
- School of Dentistry, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Alka C. Veerisetty
- Department of Pediatrics, McGovern Medical School UTHealth, Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Mohammad G. Hossain
- Department of Pediatrics, McGovern Medical School UTHealth, Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Debabrata Patra
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Michele Carrer
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA; (M.C.); (D.R.); (P.J.)
| | - Frankie Chiu
- Department of Pediatrics, McGovern Medical School UTHealth, Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Dorde Relic
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA; (M.C.); (D.R.); (P.J.)
| | - Paymaan Jafar-nejad
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA; (M.C.); (D.R.); (P.J.)
| | - Karen L. Posey
- Department of Pediatrics, McGovern Medical School UTHealth, Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| |
Collapse
|
3
|
Anwar AA, Jalan-Sakrikar N, Huebert RC. LncRNAs, RNA Therapeutics, and Emerging Technologies in Liver Pathobiology. Semin Liver Dis 2024. [PMID: 39603269 DOI: 10.1055/a-2490-1921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The field of ribonucleic acid (RNA) biology has revealed an array of noncoding RNA species, particularly long noncoding RNAs (lncRNAs), which play crucial roles in liver disease pathogenesis. This review explores the diverse functions of lncRNAs in liver pathology, including metabolic-associated steatotic liver disease, hepatocellular carcinoma, alcohol-related liver disease, and cholangiopathies such as primary sclerosing cholangitis and cholangiocarcinoma. We highlight key lncRNAs that regulate lipid metabolism, inflammation, fibrosis, and oncogenesis in the liver, demonstrating their diagnostic and therapeutic potential. Emerging RNA-based therapies, such as mRNA therapy, RNA interference, and antisense oligonucleotides, offer approaches to modulate lncRNA activity and address liver disease at a molecular level. Advances in sequencing technologies and bioinformatics pipelines are simultaneously enabling the identification and functional characterization of novel lncRNAs, driving innovation in personalized medicine. In conclusion, this review highlights the potential of lncRNAs as biomarkers and therapeutic targets in liver disease and emphasizes the need for further research into their regulatory mechanisms and clinical applications.
Collapse
Affiliation(s)
- Abid A Anwar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
- Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Robert C Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
- Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| |
Collapse
|
4
|
Xie H, Liu X, Li S, Wang M, Li Y, Chen T, Li L, Wang F, Xiao X. Tissue adaptation to metabolic stress: insights from SUMOylation. Front Endocrinol (Lausanne) 2024; 15:1434338. [PMID: 39588331 PMCID: PMC11586182 DOI: 10.3389/fendo.2024.1434338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Post-translational modification (PTM) plays a crucial role in adaptation of mammals to environmental changes, enabling them to survive in stressful situations. One such PTM is SUMO modification, which is evolutionarily conserved. It involves the covalent and reversible attachment of a small ubiquitin-like modifier (SUMO) to lysine (Lys) residues in the target protein. SUMOylation regulates various functions, including cell proliferation, differentiation, apoptosis, senescence, and maintenance of specific cellular activities. It achieves this by influencing protein-protein interactions, subcellular localization, protein stability, and DNA binding activity. Mounting evidence suggests that SUMOylation is implicated in the pathogenesis of metabolic disorders such as obesity, insulin resistance, and fatty liver. This review aims to provide an overview of the role of SUMOylation in regulating tissue adaptation to metabolic stress. Recent advancements in spectroscopic techniques have shed light on potential targets of SUMOylation and the underlying regulatory mechanisms have been elucidated, laying the theoretical foundation for the development of targeted SUMOylation interventions for metabolic syndrome while minimizing side effects.
Collapse
Affiliation(s)
- Hao Xie
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin Liu
- Department of Interventional Radiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuo Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ming Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying Li
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ting Chen
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Linwei Li
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Faxi Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
5
|
Guo X, Feng H, Cai L, Zheng J, Li Y. DPP-IV as a potential candidate in anti-obesity and obesity-related diseases treatment. Biomed Pharmacother 2024; 180:117464. [PMID: 39326107 DOI: 10.1016/j.biopha.2024.117464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Along with social development and lifestyle changes, the number of overweight and obese patients worldwide is rising annually. Obesity is a chronic metabolic disease with complex etiology. Dipeptidyl peptidase IV (DPP-IV) is a novel adipokine with significantly elevated expression in the visceral fat of obese patients. DPP-IV is a molecule that regulates metabolic homeostasis and inflammatory processes. Through its enzymatic activity, it plays a significant part in achieving hypoglycemic and weight loss effects through various pathways. DPP-IV and DPP-IV inhibitors also have pleiotropic effects in modulating obesity-related diseases by reducing obesity-related inflammation, ameliorating inflammatory bowel disease (IBD), improving hepatic steatosis and lowering cardiovascular risk, and even decreasing the risk of novel coronavirus disease-19 (COVID-19). This paper reviews the mechanisms of action based on DPP-IV targets in obesity and metabolic homeostasis, as well as their active role in the treatment of chronic diseases associated with obesity.
Collapse
Affiliation(s)
- Xin Guo
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| | - Huolun Feng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China.
| | - Liyang Cai
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| | - Jiabin Zheng
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| | - Yong Li
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
6
|
Tong X, Yagan M, Hu R, Nevills S, Doss TD, Stein RW, Balamurugan AN, Gu G. Metabolic Stress Levels Influence the Ability of Myelin Transcription Factors to Regulate β-Cell Identity and Survival. Diabetes 2024; 73:1662-1672. [PMID: 39058602 PMCID: PMC11417441 DOI: 10.2337/db23-0528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
A hallmark of type 2 diabetes (T2D) is endocrine islet β-cell failure, which can occur via cell dysfunction, loss of identity, and/or death. How each is induced remains largely unknown. We used mouse β-cells deficient for myelin transcription factors (Myt TFs; including Myt1, -2, and -3) to address this question. We previously reported that inactivating all three Myt genes in pancreatic progenitor cells (MytPancΔ) caused β-cell failure and late-onset diabetes in mice. Their lower expression in human β-cells is correlated with β-cell dysfunction, and single nucleotide polymorphisms in MYT2 and MYT3 are associated with a higher risk of T2D. We now show that these Myt TF-deficient postnatal β-cells also dedifferentiate by reactivating several progenitor markers. Intriguingly, mosaic Myt TF inactivation in only a portion of islet β-cells did not result in overt diabetes, but this created a condition where Myt TF-deficient β-cells remained alive while activating several markers of Ppy-expressing islet cells. By transplanting MytPancΔ islets into the anterior eye chambers of immune-compromised mice, we directly show that glycemic and obesity-related conditions influence cell fate, with euglycemia inducing several Ppy+ cell markers and hyperglycemia and insulin resistance inducing additional cell death. These findings suggest that the observed β-cell defects in T2D depend not only on their inherent genetic/epigenetic defects but also on the metabolic load. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xin Tong
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Mahircan Yagan
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Ruiying Hu
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Simone Nevills
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Teri D. Doss
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Roland W. Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Appakalai N. Balamurugan
- Center for Clinical and Translational Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH
| | - Guoqiang Gu
- Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
7
|
Li TL, Zhu NN, Yin Z, Sun J, Guo JP, Yuan HT, Shi XM, Guo HY, Li SX, Shan ZL. Transcriptomic analysis of epicardial adipose tissue reveals the potential crosstalk genes and immune relationship between type 2 diabetes mellitus and atrial fibrillation. Gene 2024; 920:148528. [PMID: 38703871 DOI: 10.1016/j.gene.2024.148528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/27/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The complex relationship between atrial fibrillation (AF) and type 2 diabetes mellitus (T2DM) suggests a potential role for epicardial adipose tissue (EAT) that requires further investigation. This study employs bioinformatics and experimental approaches to clarify EAT's role in linking T2DM and AF, aiming to unravel the biological mechanisms involved. METHOD Bioinformatics analysis initially identified common differentially expressed genes (DEGs) in EAT from T2DM and AF datasets. Pathway enrichment and network analyses were then performed to determine the biological significance and network connections of these DEGs. Hub genes were identified through six CytoHubba algorithms and subsequently validated biologically, with further in-depth analyses confirming their roles and interactions. Experimentally, db/db mice were utilized to establish a T2DM model. AF induction was executed via programmed transesophageal electrical stimulation and burst pacing, focusing on comparing the incidence and duration of AF. Frozen sections and Hematoxylin and Eosin (H&E) staining illuminated the structures of the heart and EAT. Moreover, quantitative PCR (qPCR) measured the expression of hub genes. RESULTS The study identified 106 DEGs in EAT from T2DM and AF datasets, underscoring significant pathways in energy metabolism and immune regulation. Three hub genes, CEBPZ, PAK1IP1, and BCCIP, emerged as pivotal in this context. In db/db mice, a marked predisposition towards AF induction and extended duration was observed, with HE staining verifying the presence of EAT. Additionally, qPCR validated significant changes in hub genes expression in db/db mice EAT. In-depth analysis identified 299 miRNAs and 33 TFs as potential regulators, notably GRHL1 and MYC. GeneMANIA analysis highlighted the hub genes' critical roles in stress responses and leukocyte differentiation, while immune profile correlations highlighted their impact on mast cells and neutrophils, emphasizing the genes' significant influence on immune regulation within the context of T2DM and AF. CONCLUSION This investigation reveals the molecular links between T2DM and AF with a focus on EAT. Targeting these pathways, especially EAT-related ones, may enable personalized treatments and improved outcomes.
Collapse
Affiliation(s)
- Tian-Lun Li
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Na-Na Zhu
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhao Yin
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiao Sun
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jian-Pin Guo
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Tao Yuan
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiang-Min Shi
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Yang Guo
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shi-Xing Li
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhao-Liang Shan
- Postgraduate School, Medical School of Chinese PLA, Beijing, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
8
|
de Oliveira EF, Garg P, Hjerling-Leffler J, Batista-Brito R, Sjulson L. Identifying patterns differing between high-dimensional datasets with generalized contrastive PCA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607264. [PMID: 39149388 PMCID: PMC11326262 DOI: 10.1101/2024.08.08.607264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
High-dimensional data have become ubiquitous in the biological sciences, and it is often desirable to compare two datasets collected under different experimental conditions to extract low-dimensional patterns enriched in one condition. However, traditional dimensionality reduction techniques cannot accomplish this because they operate on only one dataset. Contrastive principal component analysis (cPCA) has been proposed to address this problem, but it has seen little adoption because it requires tuning a hyperparameter resulting in multiple solutions, with no way of knowing which is correct. Moreover, cPCA uses foreground and background conditions that are treated differently, making it ill-suited to compare two experimental conditions symmetrically. Here we describe the development of generalized contrastive PCA (gcPCA), a flexible hyperparameter-free approach that solves these problems. We first provide analyses explaining why cPCA requires a hyperparameter and how gcPCA avoids this requirement. We then describe an open-source gcPCA toolbox containing Python and MATLAB implementations of several variants of gcPCA tailored for different scenarios. Finally, we demonstrate the utility of gcPCA in analyzing diverse high-dimensional biological data, revealing unsupervised detection of hippocampal replay in neurophysiological recordings and heterogeneity of type II diabetes in single-cell RNA sequencing data. As a fast, robust, and easy-to-use comparison method, gcPCA provides a valuable resource facilitating the analysis of diverse high-dimensional datasets to gain new insights into complex biological phenomena.
Collapse
Affiliation(s)
| | - Pranjal Garg
- All India Institute of Medical Sciences, Rishikesh, India
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Renata Batista-Brito
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| | - Lucas Sjulson
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
9
|
Chen Q, Zhao X, Xu Z, Liu Y. Endoplasmic reticulum stress mechanisms and exercise intervention in type 2 diabetes mellitus. Biomed Pharmacother 2024; 177:117122. [PMID: 38991302 DOI: 10.1016/j.biopha.2024.117122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease primarily characterized by insulin resistance (IR) and insufficient insulin secretion. The unfolded protein response (UPR) overactivation induced by endoplasmic reticulum stress (ERS) appears to play a key role in this process, although the exact pathogenesis of T2DM is not fully understood. Studies have demonstrated that appropriate exercise can regulate ERS in the heart, liver, pancreas, skeletal muscle, and other body tissues leading to an improvement in diabetes and its complications. However, the exact mechanism remains unclear. By analyzing the relationship between ERS, T2DM pathology, and exercise intervention, this review concludes that exercise can increase insulin sensitivity, inhibit IR, promote insulin secretion and alleviate T2DM by regulating ERS. This paper specifically reviews the signaling pathways by which ERS induces diabetes, the mechanisms of exercise regulation of ERS in diabetes, and the varying effects of different types of exercise on diabetes improvement through ERS mechanisms. Physical exercise is an effective non-pharmacological intervention for T2DM. Thus, further exploration of how exercise regulates ERS in diabetes could refine "precision exercise medicine" for diabetes and identify new drug targets.
Collapse
Affiliation(s)
- Qianyu Chen
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Xiaoqin Zhao
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Zujie Xu
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Yiyao Liu
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| |
Collapse
|
10
|
Cheng D, Zhang M, Zheng Y, Wang M, Gao Y, Wang X, Liu X, Lv W, Zeng X, Belosludtsev KN, Su J, Zhao L, Liu J. α-Ketoglutarate prevents hyperlipidemia-induced fatty liver mitochondrial dysfunction and oxidative stress by activating the AMPK-pgc-1α/Nrf2 pathway. Redox Biol 2024; 74:103230. [PMID: 38875959 PMCID: PMC11226981 DOI: 10.1016/j.redox.2024.103230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
α-Ketoglutarate (AKG), a crucial intermediate in the tricarboxylic acid cycle, has been demonstrated to mitigate hyperlipidemia-induced dyslipidemia and endothelial damage. While hyperlipidemia stands as a major trigger for non-alcoholic fatty liver disease, the protection of AKG on hyperlipidemia-induced hepatic metabolic disorders remains underexplored. This study aims to investigate the potential protective effects and mechanisms of AKG against hepatic lipid metabolic disorders caused by acute hyperlipidemia. Our observations indicate that AKG effectively alleviates hepatic lipid accumulation, mitochondrial dysfunction, and loss of redox homeostasis in P407-induced hyperlipidemia mice, as well as in palmitate-injured HepG2 cells and primary hepatocytes. Mechanistic insights reveal that the preventive effects are mediated by activating the AMPK-PGC-1α/Nrf2 pathway. In conclusion, our findings shed light on the role and mechanism of AKG in ameliorating abnormal lipid metabolic disorders in hyperlipidemia-induced fatty liver, suggesting that AKG, an endogenous mitochondrial nutrient, holds promising potential for addressing hyperlipidemia-induced fatty liver conditions.
Collapse
Affiliation(s)
- Danyu Cheng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Mo Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Yezi Zheng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Min Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Yilin Gao
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710018, China
| | - Xudong Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Xuyun Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Xin Zeng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Konstantin N Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, Pl. Lenina 1, Yoshkar-Ola, 424001, Russia; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290, Russia
| | - Jiacan Su
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Lin Zhao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China.
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, and Cardiometabolic Innovation Center of Ministry of Education, Department of Cardiology, and Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266071, China.
| |
Collapse
|
11
|
Jaume G, Peeters T, Song AH, Pettit R, Williamson DFK, Oldenburg L, Vaidya A, de Brot S, Chen RJ, Thiran JP, Le LP, Gerber G, Mahmood F. AI-driven Discovery of Morphomolecular Signatures in Toxicology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604355. [PMID: 39091765 PMCID: PMC11291055 DOI: 10.1101/2024.07.19.604355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Early identification of drug toxicity is essential yet challenging in drug development. At the preclinical stage, toxicity is assessed with histopathological examination of tissue sections from animal models to detect morphological lesions. To complement this analysis, toxicogenomics is increasingly employed to understand the mechanism of action of the compound and ultimately identify lesion-specific safety biomarkers for which in vitro assays can be designed. However, existing works that aim to identify morphological correlates of expression changes rely on qualitative or semi-quantitative morphological characterization and remain limited in scale or morphological diversity. Artificial intelligence (AI) offers a promising approach for quantitatively modeling this relationship at an unprecedented scale. Here, we introduce GEESE, an AI model designed to impute morphomolecular signatures in toxicology data. Our model was trained to predict 1,536 gene targets on a cohort of 8,231 hematoxylin and eosin-stained liver sections from Rattus norvegicus across 127 preclinical toxicity studies. The model, evaluated on 2,002 tissue sections from 29 held-out studies, can yield pseudo-spatially resolved gene expression maps, which we correlate with six key drug-induced liver injuries (DILI). From the resulting 25 million lesion-expression pairs, we established quantitative relations between up and downregulated genes and lesions. Validation of these signatures against toxicogenomic databases, pathway enrichment analyses, and human hepatocyte cell lines asserted their relevance. Overall, our study introduces new methods for characterizing toxicity at an unprecedented scale and granularity, paving the way for AI-driven discovery of toxicity biomarkers.
Collapse
Affiliation(s)
- Guillaume Jaume
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas Peeters
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Signal Processing Laboratory, EPFL, Lausanne, Switzerland
| | - Andrew H. Song
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Rowland Pettit
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Drew F. K. Williamson
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Lukas Oldenburg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Anurag Vaidya
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA
| | - Simone de Brot
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Switzerland
| | - Richard J. Chen
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | | | - Long Phi Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| | - Georg Gerber
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| |
Collapse
|
12
|
Shi C, Wang Y, Guo J, Zhang D, Zhang Y, Gong Z. Deacetylated MDH1 and IDH1 aggravates PANoptosis in acute liver failure through endoplasmic reticulum stress signaling. Cell Death Discov 2024; 10:275. [PMID: 38851781 PMCID: PMC11162427 DOI: 10.1038/s41420-024-02054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024] Open
Abstract
Acute liver failure (ALF) is a disease with a high mortality rate and poor prognosis, whose pathogenesis is not fully understood. PANoptosis is a recently proposed mode of cell death characterized by pyroptosis, apoptosis, and necroptosis, but it cannot be explained by any of them alone. This study aims to explore the role of PANoptosis in ALF and the impact and mechanism of deacetylated malate dehydrogenase 1 (MDH1) and isocitrate dehydrogenase 1 (IDH1) on PANoptosis. Our results found that, compared with the control group, the cell viability in the lipopolysaccharide (LPS)/D-galactosamine (D-Gal) group decreased, lactate dehydrogenase (LDH) release increased, cell death increased, and the levels of PANoptosis-related molecules RIPK1, GSDMD, caspase-3, MLKL, IL-18, IL-1β increased, indicating that PANoptosis increased during ALF. Deacetylated MDH1 at K118 and IDH1 at K93 increased the expression of PANoptosis-related molecules RIPK1, GSDMD, caspase-3, MLKL, IL-18, and IL-1β in vivo and in vitro. The deacetylation weakened the inhibitory effect of histone deacetylase (HDAC) inhibitor ACY1215 on PANoptosis-related molecules, suggesting that deacetylated MDH1 at K118 and IDH1 at K93 aggravated PANoptosis during ALF. Deacetylated MDH1 at K118 and IDH1 at K93 also promoted the expression of endoplasmic reticulum stress-related molecules BIP, ATF6, XBP1, and CHOP in vivo and in vitro. The use of endoplasmic reticulum stress inhibitor 4-PBA weakened the promotion effect of deacetylated MDH1 K118 and IDH1 K93 on PANoptosis. The results suggested that deacetylated MDH1 at K118 and IDH1 at K93 may aggravate PANoptosis in ALF through endoplasmic reticulum stress signaling. In conclusion, deacetylated MDH1 and IDH1 may aggravate PANoptosis in ALF, and the mechanism may act through endoplasmic reticulum stress signaling.
Collapse
Affiliation(s)
- Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanqiong Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Zhu SL, Qi M, Chen MT, Lin JP, Huang HF, Deng LJ, Zhou XW. A novel DDIT3 activator dehydroevodiamine effectively inhibits tumor growth and tumor cell stemness in pancreatic cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155377. [PMID: 38503154 DOI: 10.1016/j.phymed.2024.155377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The existence of pancreatic cancer stem cells (PCSCs) results in limited survival benefits from current treatment options. There is a scarcity of effective agents for treating pancreatic cancer patients. Dehydroevodiamine (DeHE), a quinazoline alkaloid isolated from the traditional Chinese herb Evodiae fructus, exhibited potent inhibition of pancreatic ductal adenocarcinoma (PDAC) cell proliferation and tumor growth both in vitro and in vivo. METHODS The cytotoxic effect of DeHE on PDAC cells was assessed using CCK-8 and colony formation assays. The antitumor efficacy of DeHE were appraised in human PANC-1 xenograft mouse model. Sphere formation assay and flow cytometry were employed to quantify the tumor stemness. RNA-Seq analysis, drug affinity responsive target stability assay (DARTS), and RNA interference transfection were conducted to elucidate potential signaling pathways. Western blotting and immunohistochemistry were utilized to assess protein expression levels. RESULTS DeHE effectively inhibited PDAC cell proliferation and tumor growth in vitro and in vivo, and exhibited a better safety profile compared to the clinical drug gemcitabine (GEM). DeHE inhibited PCSCs, as evidenced by its suppression of self-renewal capabilities of PCSCs, reduced the proportion of ALDH+ cells and downregulated stemness-associated proteins (Nanog, Sox-2, and Oct-4) both in vitro and in vivo. Furthermore, there is potential involvement of DDIT3 and its downstream DDIT3/TRIB3/AKT/mTOR pathway in the suppression of stemness characteristics within DeHE-treated PDAC cells. Additionally, results from the DARTS assay indicated that DeHE interacts with DDIT3, safeguarding it against degradation mediated by pronase. Notably, the inhibitory capabilities of DeHE on PDAC cell proliferation and tumor stemness were partially restored by siDDIT3 or the AKT activator SC-79. CONCLUSION In summary, our study has identified DeHE, a novel antitumor natural product, as an activator of DDIT3 with the ability to suppress the AKT/mTOR pathway. This pathway is intricately linked to tumor cell proliferation and stemness characteristics in PDAC. These findings suggest that DeHE holds potential as a promising candidate for the development of innovative anticancer therapeutics.
Collapse
Affiliation(s)
- Su-Li Zhu
- Department of Biochemistry and Pharmacology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Ming Qi
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Mei-Ting Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, PR China
| | - Jia-Peng Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Hai-Fu Huang
- Internal Medicine-Oncology, Shenzhen Hospital of Guangzhou University of Traditional Chinese Medicine, PR China
| | - Li-Juan Deng
- Guangzhou Key Laboratory of Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, PR China.
| | - Xing-Wang Zhou
- Department of Biochemistry and Pharmacology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China.
| |
Collapse
|
14
|
Wang R, Huang S, Wang P, Tang X, Xu H, Zhang W, Shi L, Zhong X, Lü M, Zhou X, Shi X. Research status and hotspots in the field of endoplasmic reticulum stress and liver disease: A bibliometric study. Medicine (Baltimore) 2024; 103:e38450. [PMID: 39259055 PMCID: PMC11142769 DOI: 10.1097/md.0000000000038450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/10/2024] [Accepted: 05/10/2024] [Indexed: 09/12/2024] Open
Abstract
Recently, the study of endoplasmic reticulum stress (ERS) and liver disease has attracted much attention, but bibliometric analysis on this field is scarce. Therefore, to address this gap, we conducted a bibliometric analysis to explore the research status, hotspots, and trends in this field. We searched the Web of Science Core Collection database for publications on ERS and liver disease from 2007 to 2022. Bibliometric online analysis platform, VOSviewer, and CiteSpace were used to perform bibliometric analysis. Two thousand seven hundred fifty-one publications were retrieved form the Web of Science Core Collection database. The USA was the most productive and influential country. Seoul National University, International Journal of Molecular Sciences, and Kaufman RJ were the most productive institution, journal, and author. "Endoplasmic reticulum stress," "nonalcoholic fatty liver disease," "inflammation," "oxidative stress" and "insulin resistance" were the high-frequency keywords, "necrosis factor alpha" was the keywords with the strongest citation bursts, and "nonalcoholic fatty liver," "fibrosis" and "lipid droplet" were the keywords that were still bursting in 2022. The number of publications on ERS and liver disease has increased over the past years. The USA was the most productive and influential country. China has become the country with the largest number of annual publications, but it still needs to work on the quality. ERS and nonalcoholic fatty liver disease, especially the insulin resistance and lipotoxicity in hepatocytes may be the research hotspots and trends in this field of ERS and liver disease.
Collapse
Affiliation(s)
- Ruiyu Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui County People’ Hospital, Huaian, China
- Department of Gastroenterology, Lianshui People’ Hospital of Kangda College, Affiliated to Nanjing Medical University, Huaian, China
| | - Ping Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Xiaowei Tang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Huan Xu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Wei Zhang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Lei Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Muhan Lü
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Xian Zhou
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Xiaomin Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| |
Collapse
|
15
|
Kapuy O. Mechanism of Decision Making between Autophagy and Apoptosis Induction upon Endoplasmic Reticulum Stress. Int J Mol Sci 2024; 25:4368. [PMID: 38673953 PMCID: PMC11050573 DOI: 10.3390/ijms25084368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Dynamic regulation of the cellular proteome is mainly controlled in the endoplasmic reticulum (ER). Accumulation of misfolded proteins due to ER stress leads to the activation of unfolded protein response (UPR). The primary role of UPR is to reduce the bulk of damages and try to drive back the system to the former or a new homeostatic state by autophagy, while an excessive level of stress results in apoptosis. It has already been proven that the proper order and characteristic features of both surviving and self-killing mechanisms are controlled by negative and positive feedback loops, respectively. The new results suggest that these feedback loops are found not only within but also between branches of the UPR, fine-tuning the response to ER stress. In this review, we summarize the recent knowledge of the dynamical characteristic of endoplasmic reticulum stress response mechanism by using both theoretical and molecular biological techniques. In addition, this review pays special attention to describing the mechanism of action of the dynamical features of the feedback loops controlling cellular life-and-death decision upon ER stress. Since ER stress appears in diseases that are common worldwide, a more detailed understanding of the behaviour of the stress response is of medical importance.
Collapse
Affiliation(s)
- Orsolya Kapuy
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
16
|
Diaz-Perez JA, Kerr DA. Gene of the month: DDIT3. J Clin Pathol 2024; 77:211-216. [PMID: 38053287 DOI: 10.1136/jcp-2023-208963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 12/07/2023]
Abstract
DNA damage-inducible transcript 3 (DDIT3) gene, mapped to the human chromosome 12q13.3, encodes a protein that belongs to the CCAAT/enhancer-binding protein family of transcription factors. DDIT3 is involved in the proliferative control that responds to endoplasmic reticulum stress in normal conditions, dimerising other transcription factors with basic leucine zipper (bZIP) structural motifs. DDIT3 plays a significant role during cell differentiation, especially adipogenesis, arresting the maturation of adipoblasts. In disease, FUS/EWSR1::DDIT3 fusion is the pathogenic event that drives the development of myxoid liposarcoma. The amplification of DDIT3 in other adipocytic neoplasms mediates the presence of adipoblast-like elements. Another fusion, GLI1::DDIT3, has rarely been documented in other tumours. This paper reviews the structure and function of DDIT3, its role in disease-particularly cancer-and its use and pitfalls in diagnostic testing, including immunohistochemistry as a tissue-based marker.
Collapse
Affiliation(s)
- Julio A Diaz-Perez
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Darcy A Kerr
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
17
|
Yang Y, Liu J, Kousteni S. Lipocalin 2-A bone-derived anorexigenic and β-cell promoting signal: From mice to humans. J Diabetes 2024; 16:e13504. [PMID: 38035773 PMCID: PMC10940901 DOI: 10.1111/1753-0407.13504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The skeleton is traditionally known for its structural support, organ protection, movement, and maintenance of mineral homeostasis. Over the last 10 years, bone has emerged as an endocrine organ with diverse physiological functions. The two key molecules in this context are fibroblast growth factor 23 (FGF23), secreted by osteocytes, and osteocalcin, a hormone produced by osteoblasts. FGF23 affects mineral homeostasis through its actions on the kidneys, and osteocalcin has beneficial effects in improving glucose homeostasis, muscle function, brain development, cognition, and male fertility. In addition, another osteoblast-derived hormone, lipocalin 2 (LCN2) has emerged into the researchers' field of vision. In this review, we mainly focus on LCN2's role in appetite regulation and glucose metabolism and also briefly introduce its effects in other pathophysiological conditions, such as nonalcoholic fatty liver disease, sarcopenic obesity, and cancer-induced cachexia.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Stavroula Kousteni
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
18
|
Wortham M, Ramms B, Zeng C, Benthuysen JR, Sai S, Pollow DP, Liu F, Schlichting M, Harrington AR, Liu B, Prakash TP, Pirie EC, Zhu H, Baghdasarian S, Auwerx J, Shirihai OS, Sander M. Metabolic control of adaptive β-cell proliferation by the protein deacetylase SIRT2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.24.581864. [PMID: 38464227 PMCID: PMC10925077 DOI: 10.1101/2024.02.24.581864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Selective and controlled expansion of endogenous β-cells has been pursued as a potential therapy for diabetes. Ideally, such therapies would preserve feedback control of β-cell proliferation to avoid excessive β-cell expansion and an increased risk of hypoglycemia. Here, we identified a regulator of β-cell proliferation whose inactivation results in controlled β-cell expansion: the protein deacetylase Sirtuin 2 (SIRT2). Sirt2 deletion in β-cells of mice increased β-cell proliferation during hyperglycemia with little effect in homeostatic conditions, indicating preservation of feedback control of β-cell mass. SIRT2 restrains proliferation of human islet β-cells cultured in glucose concentrations above the glycemic set point, demonstrating conserved SIRT2 function. Analysis of acetylated proteins in islets treated with a SIRT2 inhibitor revealed that SIRT2 deacetylates enzymes involved in oxidative phosphorylation, dampening the adaptive increase in oxygen consumption during hyperglycemia. At the transcriptomic level, Sirt2 inactivation has context-dependent effects on β-cells, with Sirt2 controlling how β-cells interpret hyperglycemia as a stress. Finally, we provide proof-of-principle that systemic administration of a GLP1-coupled Sirt2-targeting antisense oligonucleotide achieves β-cell selective Sirt2 inactivation and stimulates β-cell proliferation under hyperglycemic conditions. Overall, these studies identify a therapeutic strategy for increasing β-cell mass in diabetes without circumventing feedback control of β-cell proliferation.
Collapse
Affiliation(s)
- Matthew Wortham
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Bastian Ramms
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Chun Zeng
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Jacqueline R Benthuysen
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Somesh Sai
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Dennis P Pollow
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Fenfen Liu
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Michael Schlichting
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Austin R Harrington
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Bradley Liu
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Thazha P Prakash
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Elaine C Pirie
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Han Zhu
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
| | - Siyouneh Baghdasarian
- Departments of Medicine and Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Johan Auwerx
- Laboratory of Integrated Systems Physiology, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Orian S Shirihai
- Departments of Medicine and Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
19
|
Doğanyiğit Z, Okan A, Taheri S, Yılmaz Z, Akyüz E, Demir N. Evaluation of linagliptin and insulin combined therapy on unfolded protein response in type 1 diabetic mouse heart. Chem Biol Drug Des 2023; 102:1085-1096. [PMID: 37532256 DOI: 10.1111/cbdd.14308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/07/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
The aim of this study is to reveal the effects of the use of linagliptin, a DPP-4 inhibitor due to its beneficial cardiovascular effects, on endoplasmic reticulum stress (ERS) signaling, which is involved in the pathogenesis of cardiovascular complications related to type 1 diabetes. BALB/c female mice (n = 72) were divided into six groups: control, diabetes+insulin, diabetes+linagliptin, diabetes+linagliptin+insulin, diabetes+TUDCA, and diabetes+TUDCA+insulin. Immunohistochemistry and western blot method, qRT-PCR, ELISA method, and malondialdehyde (MDA) measurements were performed. Linagliptin administered to the type 1 diabetic mouse heart significantly reduced the expression levels of the total and cleaved forms of ATF6, ATF4, and p-JNK, caspase 3. Immunohistochemical and western blot analyses revealed that cleaved caspase 3 protein expression was significantly increased in the diabetes+insulin group compared to the other groups. According to ELISA findings, TUDCA was more effective in reducing NOX 1 and MDA levels than linagliptin. While linagliptin decreased the Chop mRNA level, no change was observed in the Grp78 mRNA level. Our findings showed that there was not much difference between the administration of linagliptin alone or in combination with insulin. Our study reveals that linagliptin is an effective therapeutic agent on ERS and apoptotic UPR in type 1 diabetic hearts.
Collapse
Affiliation(s)
- Züleyha Doğanyiğit
- Faculty of Medicine, Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| | - Aslı Okan
- Faculty of Medicine, Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| | - Serpil Taheri
- Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey
| | - Zeynep Yılmaz
- Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey
| | - Enes Akyüz
- Faculty of International Medicine, Department of Biophysics, University of Health Sciences, Istanbul, Turkey
| | - Necdet Demir
- Faculty of Medicine, Department of Histology and Embryology, Akdeniz University, Antalya, Turkey
| |
Collapse
|
20
|
Lee JH, Ryu H, Lee H, Yu HR, Gao Y, Lee KM, Kim YJ, Lee J. Endoplasmic reticulum stress in pancreatic β cells induces incretin desensitization and β-cell dysfunction via ATF4-mediated PDE4D expression. Am J Physiol Endocrinol Metab 2023; 325:E448-E465. [PMID: 37729023 DOI: 10.1152/ajpendo.00156.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/18/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
Pancreatic β-cell dysfunction and eventual loss are key steps in the progression of type 2 diabetes (T2D). Endoplasmic reticulum (ER) stress responses, especially those mediated by the protein kinase RNA-like ER kinase and activating transcription factor 4 (PERK-ATF4) pathway, have been implicated in promoting these β-cell pathologies. However, the exact molecular events surrounding the role of the PERK-ATF4 pathway in β-cell dysfunction remain unknown. Here, we report our discovery that ATF4 promotes the expression of PDE4D, which disrupts β-cell function via a downregulation of cAMP signaling. We found that β-cell-specific transgenic expression of ATF4 led to early β-cell dysfunction and loss, a phenotype that resembles accelerated T2D. Expression of ATF4, rather than C/EBP homologous protein (CHOP), promoted PDE4D expression, reduced cAMP signaling, and attenuated responses to incretins and elevated glucose. Furthermore, we found that β-cells of leptin receptor-deficient diabetic (db/db) mice had elevated nuclear localization of ATF4 and PDE4D expression, accompanied by impaired β-cell function. Accordingly, pharmacological inhibition of the ATF4 pathway attenuated PDE4D expression in the islets and promoted incretin-simulated glucose tolerance and insulin secretion in db/db mice. Finally, we found that inhibiting PDE4 activity with selective pharmacological inhibitors improved β-cell function in both db/db mice and β-cell-specific ATF4 transgenic mice. In summary, our results indicate that ER stress causes β-cell failure via ATF4-mediated PDE4D production, suggesting the ATF4-PDE4D pathway could be a therapeutic target for protecting β-cell function during the progression of T2D.NEW & NOTEWORTHY Endoplasmic reticulum stress has been implied to cause multiple β-cell pathologies during the progression of type 2 diabetes (T2D). However, the precise molecular events underlying this remain unknown. Here, we discovered that elevated ATF4 activity, which was seen in T2D β cells, attenuated β-cell proliferation and impaired insulin secretion via PDE4D-mediated downregulation of cAMP signaling. Additionally, we demonstrated that pharmacological inhibition of the ATF4 pathway or PDE4D activity alleviated β-cell dysfunction, suggesting its therapeutic usefulness against T2D.
Collapse
Affiliation(s)
- Ji-Hye Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Hanguk Ryu
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Hyejin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Hye Ram Yu
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Yurong Gao
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Kyeong-Min Lee
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Young-Joon Kim
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| |
Collapse
|
21
|
Zhang Y, Zhao T, Hu L, Xue J. Integrative Analysis of Core Genes and Biological Process Involved in Polycystic Ovary Syndrome. Reprod Sci 2023; 30:3055-3070. [PMID: 37171773 DOI: 10.1007/s43032-023-01259-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 04/29/2023] [Indexed: 05/13/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common gynecological endocrine disordered disease, affecting the function of the ovaries in women of reproductive age. However, there are limited curative therapies for PCOS due to lack of reliable candidates. Hence, this study aimed to identify hub pathogenic genes and potential therapeutic targets for PCOS using bioinformatics tools. We obtained the expression profiles of 29 PCOS samples and 24 normal samples from three Gene Expression Omnibus (GEO) datasets. Then, the differentially expressed genes (DEGs) were screened, which were subjected to functional enrichment analyses. Moreover, we found 30 ferroptosis-related genes out of the 89 DEGs. Among the top 10 significant ferroptosis-related DEGs, 8 genes showed good predictive performance. We constructed interaction network of top three ferroptosis-related DEGs (SLC38A1, ACO1, DDIT3). Finally, real-time PCR was performed to test the relative expression of these genes. In conclusions, we have identified ferroptosis-related DEGs as core genes and potential therapeutic targets of PCOS based on comprehensive bioinformatics analysis. The findings are conducive to understanding of the pathogenesis of PCOS and paving the way towards curative therapies.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Obstetrics and Gynecology, Yan'an University Affiliated Hospital, No. 43 North Street, Baota District, Yan'an, 716000, Shaanxi, People's Republic of China
| | - Tianyi Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Lishuang Hu
- Department of Obstetrics and Gynecology, Yan'an University Affiliated Hospital, No. 43 North Street, Baota District, Yan'an, 716000, Shaanxi, People's Republic of China
| | - Juan Xue
- Department of Obstetrics and Gynecology, Yan'an University Affiliated Hospital, No. 43 North Street, Baota District, Yan'an, 716000, Shaanxi, People's Republic of China.
| |
Collapse
|
22
|
Klyosova E, Azarova I, Buikin S, Polonikov A. Differentially Expressed Genes Regulating Glutathione Metabolism, Protein-Folding, and Unfolded Protein Response in Pancreatic β-Cells in Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:12059. [PMID: 37569434 PMCID: PMC10418503 DOI: 10.3390/ijms241512059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/12/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Impaired redox homeostasis in the endoplasmic reticulum (ER) may contribute to proinsulin misfolding and thus to activate the unfolded protein response (UPR) and apoptotic pathways, culminating in pancreatic β-cell loss and type 2 diabetes (T2D). The present study was designed to identify differentially expressed genes (DEGs) encoding enzymes for glutathione metabolism and their impact on the expression levels of genes regulating protein folding and UPR in β-cells of T2D patients. The GEO transcriptome datasets of β-cells of diabetics and non-diabetics, GSE20966 and GSE81608, were analyzed for 142 genes of interest using limma and GREIN software, respectively. Diabetic β-cells showed dataset-specific patterns of DEGs (FDR ≤ 0.05) implicated in the regulation of glutathione metabolism (ANPEP, PGD, IDH2, and CTH), protein-folding (HSP90AB1, HSP90AA1, HSPA1B, HSPA8, BAG3, NDC1, NUP160, RLN1, and RPS19BP1), and unfolded protein response (CREB3L4, ERP27, and BID). The GCLC gene, encoding the catalytic subunit of glutamate-cysteine ligase, the first rate-limiting enzyme of glutathione biosynthesis, was moderately down-regulated in diabetic β-cells from both datasets (p ≤ 0.05). Regression analysis established that genes involved in the de novo synthesis of glutathione, GCLC, GCLM, and GSS affect the expression levels of genes encoding molecular chaperones and those involved in the UPR pathway. This study showed for the first time that diabetic β-cells exhibit alterations in the expression of genes regulating glutathione metabolism, protein-folding, and UPR and provided evidence for the molecular crosstalk between impaired redox homeostasis and abnormal protein folding, underlying ER stress in type 2 diabetes.
Collapse
Affiliation(s)
- Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia; (E.K.); (I.A.)
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
| | - Iuliia Azarova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia; (E.K.); (I.A.)
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
| | - Stepan Buikin
- Centre of Omics Technology, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, 119991 Moscow, Russia;
- Department of Internal Diseases, Yaroslav the Wise Novgorod State University, 41 Bolshaya St. Petersburg Street, 173003 Veliky Novgorod, Russia
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| |
Collapse
|
23
|
Lacombe J, Guo K, Bonneau J, Faubert D, Gioanni F, Vivoli A, Muir SM, Hezzaz S, Poitout V, Ferron M. Vitamin K-dependent carboxylation regulates Ca 2+ flux and adaptation to metabolic stress in β cells. Cell Rep 2023; 42:112500. [PMID: 37171959 DOI: 10.1016/j.celrep.2023.112500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 02/24/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
Vitamin K is a micronutrient necessary for γ-carboxylation of glutamic acids. This post-translational modification occurs in the endoplasmic reticulum (ER) and affects secreted proteins. Recent clinical studies implicate vitamin K in the pathophysiology of diabetes, but the underlying molecular mechanism remains unknown. Here, we show that mouse β cells lacking γ-carboxylation fail to adapt their insulin secretion in the context of age-related insulin resistance or diet-induced β cell stress. In human islets, γ-carboxylase expression positively correlates with improved insulin secretion in response to glucose. We identify endoplasmic reticulum Gla protein (ERGP) as a γ-carboxylated ER-resident Ca2+-binding protein expressed in β cells. Mechanistically, γ-carboxylation of ERGP protects cells against Ca2+ overfilling by diminishing STIM1 and Orai1 interaction and restraining store-operated Ca2+ entry. These results reveal a critical role of vitamin K-dependent carboxylation in regulation of Ca2+ flux in β cells and in their capacity to adapt to metabolic stress.
Collapse
Affiliation(s)
- Julie Lacombe
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada.
| | - Kevin Guo
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Jessica Bonneau
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Denis Faubert
- Mass Spectrometry and Proteomics Platform, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Florian Gioanni
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Alexis Vivoli
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Sarah M Muir
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Soraya Hezzaz
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Mathieu Ferron
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
24
|
Tang H, Ling J, Meng H, Wu L, Zhu L, Zhu S. Temporal Relationship Between Insulin Resistance and Lipid Accumulation After Bariatric Surgery: a Multicenter Cohort Study. Obes Surg 2023:10.1007/s11695-023-06508-3. [PMID: 37060490 DOI: 10.1007/s11695-023-06508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/12/2022] [Accepted: 06/15/2022] [Indexed: 04/16/2023]
Abstract
PURPOSE Insulin resistance (IR) is closely associated with lipid accumulation. Here, we investigated the temporal relationship between the two conditions after bariatric surgery. MATERIALS AND METHODS In total, 409 participants were enrolled from three bariatric centers in China from 2009 to 2018. We evaluated whether baseline IR (proxied by homeostasis model assessment of insulin resistance (HOMA-IR)) and lipid accumulation (proxied by visceral adiposity index (VAI) and lipid accumulation product (LAP)) were associated with follow-up IR and lipid accumulation (3 months postoperatively) using linear regression models. We then conducted a cross-lagged panel analysis model to simultaneously examine the bidirectional relationship between IR and lipid accumulation. RESULTS Multivariable linear regression analyses showed that baseline HOMA-IR was associated with follow-up VAI (β = 0.430, 95% CI: 0.082-0.778, p = 0.016) and LAP (β = 0.070, 95% CI: 0.010-0.130, p = 0.022). There was no relationship between baseline lipid accumulation and follow-up IR. Further cross-lagged panel analyses indicated that the path coefficient from baseline HOMA-IR to follow-up VAI (β2 = 0.145, p = 0.003) was significantly greater than the coefficient from baseline VAI to follow-up HOMA-IR (β1 = - 0.013, p = 0.777). Similarly, the path coefficient from baseline HOMA-IR to follow-up LAP (β2 = 0.141, p = 0.003) was significantly greater than the coefficient from baseline LAP to follow-up HOMA-IR (β1 = 0.041, p = 0.391). CONCLUSION Our study demonstrated a unidirectional relationship from HOMA-IR to VAI and LAP, suggesting that the change in IR may precede lipid accumulation after surgery.
Collapse
Affiliation(s)
- Haibo Tang
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jiapu Ling
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hua Meng
- Department of General Surgery, The China-Japan Friendship Hospital, Beijing, China
| | - Liangping Wu
- Department of Metabolic Surgery, The Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liyong Zhu
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shaihong Zhu
- Department of Metabolic and Bariatric Surgery, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
25
|
Mastracci TL, Apte M, Amundadottir LT, Alvarsson A, Artandi S, Bellin MD, Bernal-Mizrachi E, Caicedo A, Campbell-Thompson M, Cruz-Monserrate Z, El Ouaamari A, Gaulton KJ, Geisz A, Goodarzi MO, Hara M, Hull-Meichle RL, Kleger A, Klein AP, Kopp JL, Kulkarni RN, Muzumdar MD, Naren AP, Oakes SA, Olesen SS, Phelps EA, Powers AC, Stabler CL, Tirkes T, Whitcomb DC, Yadav D, Yong J, Zaghloul NA, Pandol SJ, Sander M. Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases: Workshop Proceedings. Diabetes 2023; 72:433-448. [PMID: 36940317 PMCID: PMC10033248 DOI: 10.2337/db22-0942] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/29/2022] [Indexed: 03/22/2023]
Abstract
The Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases workshop was a 1.5-day scientific conference at the National Institutes of Health (Bethesda, MD) that engaged clinical and basic science investigators interested in diseases of the pancreas. This report provides a summary of the proceedings from the workshop. The goals of the workshop were to forge connections and identify gaps in knowledge that could guide future research directions. Presentations were segregated into six major theme areas, including 1) pancreas anatomy and physiology, 2) diabetes in the setting of exocrine disease, 3) metabolic influences on the exocrine pancreas, 4) genetic drivers of pancreatic diseases, 5) tools for integrated pancreatic analysis, and 6) implications of exocrine-endocrine cross talk. For each theme, multiple presentations were followed by panel discussions on specific topics relevant to each area of research; these are summarized here. Significantly, the discussions resulted in the identification of research gaps and opportunities for the field to address. In general, it was concluded that as a pancreas research community, we must more thoughtfully integrate our current knowledge of normal physiology as well as the disease mechanisms that underlie endocrine and exocrine disorders so that there is a better understanding of the interplay between these compartments.
Collapse
Affiliation(s)
- Teresa L. Mastracci
- Department of Biology, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Minoti Apte
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | | | - Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Mount Sinai Hospital, New York, NY
| | - Steven Artandi
- Department of Internal Medicine, Stanford University, Stanford, CA
| | - Melena D. Bellin
- Departments of Pediatrics and Surgery, University of Minnesota Medical School, Minneapolis, MN
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Andrea Geisz
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Rebecca L. Hull-Meichle
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University, Ulm, Germany
| | - Alison P. Klein
- Department of Pathology and Medicine, Johns Hopkins School of Medicine, Baltimore MD
| | - Janel L. Kopp
- Department of Cellular & Physiological Sciences, The University of British Columbia, Vancouver, Canada
| | | | - Mandar D. Muzumdar
- Departments of Genetics and Internal Medicine (Oncology), Yale University School of Medicine, New Haven, CT
| | | | - Scott A. Oakes
- Department of Pathology, The University of Chicago, Chicago, IL
| | - Søren S. Olesen
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Alvin C. Powers
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Temel Tirkes
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN
| | | | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Jing Yong
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Norann A. Zaghloul
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Stephen J. Pandol
- Department of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Maike Sander
- Department of Pediatrics and Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
26
|
Gurlo T, Prakash TP, Wang Z, Archang M, Pei L, Rosenberger M, Pirie E, Lee RG, Butler PC. Efficacy of IAPP suppression in mouse and human islets by GLP-1 analogue conjugated antisense oligonucleotide. Front Mol Biosci 2023; 10:1096286. [PMID: 36814640 PMCID: PMC9939749 DOI: 10.3389/fmolb.2023.1096286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
Insulin resistance is the major risk factor for Type 2 diabetes (T2D). In vulnerable individuals, insulin resistance induces a progressive loss of insulin secretion with islet pathology revealing a partial deficit of beta cells and islet amyloid derived from islet amyloid polypeptide (IAPP). IAPP is co-expressed and secreted with insulin by beta cells, expression of both proteins being upregulated in response to insulin resistance. If IAPP expression exceeds the threshold for clearance of misfolded proteins, beta cell failure occurs exacerbated by the action of IAPP toxicity to compromise the autophagy lysosomal pathway. We postulated that suppression of IAPP expression by an IAPP antisense oligonucleotide delivered to beta cells by the GLP-1 agonist exenatide (eGLP1-IAPP-ASO) is a potential disease modifying therapy for T2D. While eGLP1-IAPP-ASO suppressed mouse IAPP and transgenic human IAPP expression in mouse islets, it had no discernable effects on IAPP expression in human islets under the conditions studied. Suppression of transgenic human IAPP expression in mouse islets attenuated disruption of the autophagy lysosomal pathway in beta cells, supporting the potential of this strategy.
Collapse
Affiliation(s)
- Tatyana Gurlo
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States,*Correspondence: Tatyana Gurlo, ; Peter C. Butler,
| | | | - Zhongying Wang
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maani Archang
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lina Pei
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Madeline Rosenberger
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine Pirie
- IONIS Pharmaceuticals, Carlsbad, CA, United States
| | | | - Peter C. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States,*Correspondence: Tatyana Gurlo, ; Peter C. Butler,
| |
Collapse
|
27
|
Ajoolabady A, Kaplowitz N, Lebeaupin C, Kroemer G, Kaufman RJ, Malhi H, Ren J. Endoplasmic reticulum stress in liver diseases. Hepatology 2023; 77:619-639. [PMID: 35524448 PMCID: PMC9637239 DOI: 10.1002/hep.32562] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 02/02/2023]
Abstract
The endoplasmic reticulum (ER) is an intracellular organelle that fosters the correct folding of linear polypeptides and proteins, a process tightly governed by the ER-resident enzymes and chaperones. Failure to shape the proper 3-dimensional architecture of proteins culminates in the accumulation of misfolded or unfolded proteins within the ER, disturbs ER homeostasis, and leads to canonically defined ER stress. Recent studies have elucidated that cellular perturbations, such as lipotoxicity, can also lead to ER stress. In response to ER stress, the unfolded protein response (UPR) is activated to reestablish ER homeostasis ("adaptive UPR"), or, conversely, to provoke cell death when ER stress is overwhelmed and sustained ("maladaptive UPR"). It is well documented that ER stress contributes to the onset and progression of multiple hepatic pathologies including NAFLD, alcohol-associated liver disease, viral hepatitis, liver ischemia, drug toxicity, and liver cancers. Here, we review key studies dealing with the emerging role of ER stress and the UPR in the pathophysiology of liver diseases from cellular, murine, and human models. Specifically, we will summarize current available knowledge on pharmacological and non-pharmacological interventions that may be used to target maladaptive UPR for the treatment of nonmalignant liver diseases.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Neil Kaplowitz
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cynthia Lebeaupin
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Randal J. Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jun Ren
- Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
28
|
Wong A, Pritchard S, Moore M, Akhaphong B, Avula N, Beetch M, Fujitani Y, Alejandro EU. Overexpression of Pdx1, reduction of p53, or deletion of CHOP attenuates pancreas hypoplasia in mice with pancreas-specific O-GlcNAc transferase deletion. J Biol Chem 2023; 299:102878. [PMID: 36623733 PMCID: PMC9932656 DOI: 10.1016/j.jbc.2023.102878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 01/09/2023] Open
Abstract
Deletion of O-GlcNAc transferase (Ogt) in pancreatic epithelial progenitor cells results in pancreatic hypoplasia at birth, partly due to increased apoptosis during embryonic development. Constitutive loss of Ogt in β-cells results in increased ER stress and apoptosis, and in the Ogt-deficient pancreas, transcriptomic data previously revealed both tumor suppressor protein p53 and pancreatic duodenal homeobox 1 (Pdx1), key cell survival proteins in the developing pancreas, as upstream regulators of differentially expressed genes. However, the specific roles of these genes in pancreatic hypoplasia are unclear. In this study, we explored the independent roles of p53, ER stress protein CHOP, and Pdx1 in pancreas development and their use in the functional rescue of pancreatic hypoplasia in the context of Ogt loss. Using in vivo genetic manipulation and morphometric analysis, we show that Ogt plays a key regulatory role in pancreas development. Heterozygous, but not homozygous, loss of pancreatic p53 afforded a partial rescue of β-cell, α-cell, and exocrine cell masses, while whole body loss of CHOP afforded a partial rescue in pancreas weight and a full rescue in exocrine cell mass. However, neither was sufficient to fully mitigate pancreatic hypoplasia at birth in the Ogt-deficient pancreas. Furthermore, overexpression of Pdx1 in the pancreatic epithelium resulted in partial rescues in pancreas weight and β-cell mass in the Ogt loss background. These findings highlight the requirement of Ogt in pancreas development by targeting multiple proteins such as transcription factor Pdx1 and p53 in the developing pancreas.
Collapse
Affiliation(s)
- Alicia Wong
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA; Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Samantha Pritchard
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mackenzie Moore
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian Akhaphong
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nandini Avula
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Megan Beetch
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yoshio Fujitani
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Emilyn U Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
29
|
Kulkarni A, Muralidharan C, May SC, Tersey SA, Mirmira RG. Inside the β Cell: Molecular Stress Response Pathways in Diabetes Pathogenesis. Endocrinology 2022; 164:bqac184. [PMID: 36317483 PMCID: PMC9667558 DOI: 10.1210/endocr/bqac184] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Indexed: 11/05/2022]
Abstract
The pathogeneses of the 2 major forms of diabetes, type 1 and type 2, differ with respect to their major molecular insults (loss of immune tolerance and onset of tissue insulin resistance, respectively). However, evidence suggests that dysfunction and/or death of insulin-producing β-cells is common to virtually all forms of diabetes. Although the mechanisms underlying β-cell dysfunction remain incompletely characterized, recent years have witnessed major advances in our understanding of the molecular pathways that contribute to the demise of the β-cell. Cellular and environmental factors contribute to β-cell dysfunction/loss through the activation of molecular pathways that exacerbate endoplasmic reticulum stress, the integrated stress response, oxidative stress, and impaired autophagy. Whereas many of these stress responsive pathways are interconnected, their individual contributions to glucose homeostasis and β-cell health have been elucidated through the development and interrogation of animal models. In these studies, genetic models and pharmacological compounds have enabled the identification of genes and proteins specifically involved in β-cell dysfunction during diabetes pathogenesis. Here, we review the critical stress response pathways that are activated in β cells in the context of the animal models.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Charanya Muralidharan
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Sarah C May
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
30
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|
31
|
Xu S, Zhang P, Heing-Becker I, Zhang J, Tang P, Bej R, Bhatia S, Zhong Y, Haag R. Dual tumor- and subcellular-targeted photodynamic therapy using glucose-functionalized MoS 2 nanoflakes for multidrug-resistant tumor ablation. Biomaterials 2022; 290:121844. [PMID: 36302305 DOI: 10.1016/j.biomaterials.2022.121844] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2022]
Abstract
Photodynamic therapy (PDT) is emerging as an efficient strategy to combat multidrug-resistant (MDR) cancer. However, the short half-life and limited diffusion of reactive oxygen species (ROS) undermine the therapeutic outcomes of this therapy. To address this issue, a tumor-targeting nanoplatform was developed to precisely deliver mitochondria- and endoplasmic reticulum (ER)-targeting PDT agents to desired sites for dual organelle-targeted PDT. The nanoplatform is constructed by functionalizing molybdenum disulfide (MoS2) nanoflakes with glucose-modified hyperbranched polyglycerol (hPG), and then loading the organelle-targeting PDT agents. The resultant nanoplatform Cy7.5-TG@GPM is demonstrated to mediate both greatly enhanced internalization within MDR cells and precise subcellular localization of PDT agents, facilitating in situ near-infrared (NIR)-triggered ROS generation for augmented PDT and reversal of MDR, causing impressive tumor shrinkage in a HeLa multidrug-resistant tumor mouse model. As revealed by mechanistic studies of the synergistic mitochondria- and ER-targeted PDT, ROS-induced ER stress not only activates the cytosine-cytosine-adenosine-adenosine thymidine/enhancer-binding protein homologous protein (CHOP) pro-apoptotic signaling pathway, but also cooperates with ROS-induced mitochondrial dysfunction to trigger cytochrome C release from the mitochondria and induce subsequent cell death. Furthermore, the mitochondrial dysfunction reduces ATP production and thereby contributes to the reversal of MDR. This nanoplatform, with its NIR-responsive properties and ability to target tumors and subcellular organelles, offers a promising strategy for effective MDR cancer therapy.
Collapse
Affiliation(s)
- Shaohui Xu
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany
| | - Pan Zhang
- School of Engineering, China Pharmaceutical University, 639 Longmian Avenue, 211198, Nanjing, China
| | - Isabelle Heing-Becker
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany
| | - Junmei Zhang
- School of Engineering, China Pharmaceutical University, 639 Longmian Avenue, 211198, Nanjing, China
| | - Peng Tang
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany
| | - Raju Bej
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany
| | - Sumati Bhatia
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany
| | - Yinan Zhong
- School of Engineering, China Pharmaceutical University, 639 Longmian Avenue, 211198, Nanjing, China.
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany.
| |
Collapse
|
32
|
Kang Z, Chen F, Wu W, Liu R, Chen T, Xu F. UPRmt and coordinated UPRER in type 2 diabetes. Front Cell Dev Biol 2022; 10:974083. [PMID: 36187475 PMCID: PMC9523447 DOI: 10.3389/fcell.2022.974083] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is a molecular mechanism that maintains mitochondrial proteostasis under stress and is closely related to various metabolic diseases, such as type 2 diabetes (T2D). Similarly, the unfolded protein response of the endoplasmic reticulum (UPRER) is responsible for maintaining proteomic stability in the endoplasmic reticulum (ER). Since the mitochondria and endoplasmic reticulum are the primary centers of energy metabolism and protein synthesis in cells, respectively, a synergistic mechanism must exist between UPRmt and UPRER to cooperatively resist stresses such as hyperglycemia in T2D. Increasing evidence suggests that the protein kinase RNA (PKR)-like endoplasmic reticulum kinase (PERK) signaling pathway is likely an important node for coordinating UPRmt and UPRER. The PERK pathway is activated in both UPRmt and UPRER, and its downstream molecules perform important functions. In this review, we discuss the mechanisms of UPRmt, UPRER and their crosstalk in T2D.
Collapse
Affiliation(s)
- Zhanfang Kang
- Department of Basic Medical Research, Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Feng Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wanhui Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Rui Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tianda Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fang Xu
- Department of Basic Medical Research, Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Fang Xu,
| |
Collapse
|
33
|
Guevara-Olaya L, Chimal-Vega B, Castañeda-Sánchez CY, López-Cossio LY, Pulido-Capiz A, Galindo-Hernández O, Díaz-Molina R, Ruiz Esparza-Cisneros J, García-González V. LDL Promotes Disorders in β-Cell Cholesterol Metabolism, Implications on Insulin Cellular Communication Mediated by EVs. Metabolites 2022; 12:754. [PMID: 36005626 PMCID: PMC9415214 DOI: 10.3390/metabo12080754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/01/2022] Open
Abstract
Dyslipidemia is described as a hallmark of metabolic syndrome, promoting a stage of metabolic inflammation (metainflammation) that could lead to misbalances in energetic metabolism, contributing to insulin resistance, and modifying intracellular cholesterol pathways and the renin-angiotensin system (RAS) in pancreatic islets. Low-density lipoprotein (LDL) hypercholesterolemia could disrupt the tissue communication between Langerhans β-cells and hepatocytes, wherein extracellular vesicles (EVs) are secreted by β-cells, and exposition to LDL can impair these phenomena. β-cells activate compensatory mechanisms to maintain insulin and metabolic homeostasis; therefore, the work aimed to characterize the impact of LDL on β-cell cholesterol metabolism and the implication on insulin secretion, connected with the regulation of cellular communication mediated by EVs on hepatocytes. Our results suggest that β-cells can endocytose LDL, promoting an increase in de novo cholesterol synthesis targets. Notably, LDL treatment increased mRNA levels and insulin secretion; this hyperinsulinism condition was associated with the transcription factor PDX-1. However, a compensatory response that maintains basal levels of intracellular calcium was described, mediated by the overexpression of calcium targets PMCA1/4, SERCA2, and NCX1, together with the upregulation of the unfolded protein response (UPR) through the activation of IRE1 and PERK arms to maintain protein homeostasis. The LDL treatment induced metainflammation by IL-6, NF-κB, and COX-2 overexpression. Furthermore, LDL endocytosis triggered an imbalance of the RAS components. LDL treatment increased the intracellular levels of cholesterol on lipid droplets; the adaptive β-cell response was portrayed by the overexpression of cholesterol transporters ABCA1 and ABCG1. Therefore, lipotoxicity and hyperinsulinism induced by LDL were regulated by the natural compound auraptene, a geranyloxyn coumarin modulator of cholesterol-esterification by ACAT1 enzyme inhibition. EVs isolated from β-cells impaired insulin signaling via mTOR/p70S6Kα in hepatocytes, a phenomenon regulated by auraptene. Our results show that LDL overload plays a novel role in hyperinsulinism, mechanisms associated with a dysregulation of intracellular cholesterol, lipotoxicity, and the adaptive UPR, which may be regulated by coumarin-auraptene; these conditions explain the affectations that occur during the initial stages of insulin resistance.
Collapse
Affiliation(s)
- Lizbeth Guevara-Olaya
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| | - Brenda Chimal-Vega
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| | - César Yahel Castañeda-Sánchez
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| | - Leslie Y. López-Cossio
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| | - Angel Pulido-Capiz
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio de Biología Molecular, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
| | - Octavio Galindo-Hernández
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| | - Raúl Díaz-Molina
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| | | | - Victor García-González
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, BC, Mexico
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de BC, Mexicali 21000, BC, Mexico
| |
Collapse
|
34
|
Lv C, Sun Y, Zhang ZY, Aboelela Z, Qiu X, Meng ZX. β-cell dynamics in type 2 diabetes and in dietary and exercise interventions. J Mol Cell Biol 2022; 14:6656373. [PMID: 35929791 PMCID: PMC9710517 DOI: 10.1093/jmcb/mjac046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/07/2022] [Accepted: 08/03/2022] [Indexed: 01/14/2023] Open
Abstract
Pancreatic β-cell dysfunction and insulin resistance are two of the major causes of type 2 diabetes (T2D). Recent clinical and experimental studies have suggested that the functional capacity of β-cells, particularly in the first phase of insulin secretion, is a primary contributor to the progression of T2D and its associated complications. Pancreatic β-cells undergo dynamic compensation and decompensation processes during the development of T2D, in which metabolic stresses such as endoplasmic reticulum stress, oxidative stress, and inflammatory signals are key regulators of β-cell dynamics. Dietary and exercise interventions have been shown to be effective approaches for the treatment of obesity and T2D, especially in the early stages. Whilst the targeted tissues and underlying mechanisms of dietary and exercise interventions remain somewhat vague, accumulating evidence has implicated the improvement of β-cell functional capacity. In this review, we summarize recent advances in the understanding of the dynamic adaptations of β-cell function in T2D progression and clarify the effects and mechanisms of dietary and exercise interventions on β-cell dysfunction in T2D. This review provides molecular insights into the therapeutic effects of dietary and exercise interventions on T2D, and more importantly, it paves the way for future research on the related underlying mechanisms for developing precision prevention and treatment of T2D.
Collapse
Affiliation(s)
- Chengan Lv
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuchen Sun
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China,Zhejiang University–University of Edinburgh Institute (ZJE), Zhejiang University, Haining 314400, China
| | - Zhe Yu Zhang
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zeyad Aboelela
- Department of Pathology and Pathophysiology and Metabolic Research Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China,Bachelors of Surgery, Bachelors of Medicine (MBBS), Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | | |
Collapse
|
35
|
Sahin E, Saglam N, Erdem S, Alvuroglu E, Abidin I, Yulug E, Alver A. 7,8-Dihydroxyflavone alleviates Endoplasmic Reticulum Stress in cafeteria diet-induced metabolic syndrome. Life Sci 2022; 306:120781. [PMID: 35835252 DOI: 10.1016/j.lfs.2022.120781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 10/17/2022]
Abstract
AIMS Prolonged Endoplasmic Reticulum Stress (ERS) is involved in the pathogenesis of metabolic syndrome, including type-2 diabetes mellitus, cardiovascular diseases, atherosclerosis, obesity, and fatty liver disease. There have been significant efforts to discover molecules to treat ERS and/or to ameliorate associate symptoms. In this study, we investigated the effect of 7,8-Dihydroxyflavone (7,8-DHF) on ERS in liver and pancreas tissues in a cafeteria (CAF) diet induced metabolic syndrome model. MAIN METHODS Male C57BL/6 mice were fed CAF diet for 16 weeks and 7,8-DHF was administered intraperitoneally (5 mg/kg/day) for last four weeks. 78-kDa glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) in liver and pancreas tissues, insulin and interleukin-1β (IL-1β) in serum were analyzed by ELISA method and serum biochemistry parameters were analyzed with autoanalyzer. GRP78 and CHOP gene expression levels were determined by qRT-PCR. In addition, histopathological analyzes were performed on liver and pancreas tissues. KEY FINDINGS Findings revealed that CAF diet caused metabolic abnormalities, insulin resistance and inflammation in serum and triggered ERS in pancreas and liver tissues. 7,8-DHF treatment significantly reduced metabolic abnormalities by reducing serum biochemical parameters, HOMO-IR and IL-1β levels. qRT-PCR and ELISA results indicated that 7,8-DHF treatment down-regulated GRP78 and CHOP expression and protein levels in the liver and GRP78 expression in pancreas. Efficiency of 7,8-DHF in these tissues was also demonstrated by histopathological tests. SIGNIFICANCE In conclusion, CAF diet-induced metabolic syndrome model, 7,8-DHF suppressed ERS and ERS-induced metabolic disorders in both liver and pancreas. Therefore, 7,8-DHF may potentially be a novel therapeutic compound to ameliorate ERS and related metabolic symptoms.
Collapse
Affiliation(s)
- Elif Sahin
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon, Turkiye.
| | - Neslihan Saglam
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon, Turkiye
| | - Seniz Erdem
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon, Turkiye
| | - Elif Alvuroglu
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Ismail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Esin Yulug
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Ahmet Alver
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| |
Collapse
|
36
|
Wang N, Gao Q, Shi J, Yulan C, Ji W, Sheng X, Zhang R. Acacetin antagonized lipotoxicity in pancreatic β-cells via ameliorating oxidative stress and endoplasmic reticulum stress. Mol Biol Rep 2022; 49:8727-8740. [PMID: 35780455 DOI: 10.1007/s11033-022-07717-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE During the pathogenesis and progression of diabetes, lipotoxicity is a major threat to the function and survival of pancreatic β-cells. To battle against the lipotoxicity induced cellular damages, the present study investigated the beneficial effects of acacetin, a natural antioxidant, on free fatty acid (FFA) stressed RINm5F cells and the potential mechanism involved. MATERIALS AND METHODS RINm5F cells with or without 1 h pretreatment of acacetin were treated with 0.35 mM sodium palmitate for 24 h. Cell viability, intracellular reactive oxygen species (ROS) level, antioxidant capacity, cellular apoptosis, and endoplasmic reticulum (ER) stress biomarker expression were investigated. RESULTS Our experiments demonstrated that acacetin treatment significantly scavenged the intracellular ROS, upregulated the endogenous antioxidant enzymes, and diminished the sub-G1 DNA fraction in the cells exposed to FFA, suggesting its efficacy against oxidative stress. Meanwhile, acacetin treatment significantly mitigated the overload of intracellular Ca2+ and reduced the pro-apoptotic protein expression in the FFA stimulated cells, and thereby attenuated the ER stress-mediated cell apoptosis. Furthermore, siRNA interference results confirmed that the suppressing of C/EBP-homologous protein (CHOP) was critical to improve FFA-induced reduction in cell viability and ameliorated the ER stress caused by FFA stimulation. CONCLUSIONS Acacetin may antagonize lipotoxicity in pancreatic cells by attenuating the oxidative stress and ER stress.
Collapse
Affiliation(s)
- Ning Wang
- Department of Biotechnology, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| | - Qing Gao
- Department of Biotechnology, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jie Shi
- Department of Biochemistry, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Chen Yulan
- Department of Biotechnology, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Weimeng Ji
- Department of Biochemistry, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiumei Sheng
- Department of Biochemistry, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Rui Zhang
- Department of Biochemistry, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
37
|
Truong E, Pandol S, Jeon C. Uniting epidemiology and experimental models: pancreatic steatosis and pancreatic cancer. EBioMedicine 2022; 79:103996. [PMID: 35405390 PMCID: PMC9010750 DOI: 10.1016/j.ebiom.2022.103996] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/07/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023] Open
Abstract
Research from epidemiologic studies and experimental animal models provide insights into the role of pancreatic steatosis in the development of pancreatic cancer. Epidemiologic data demonstrate that pancreatic steatosis is widely prevalent and significantly associated with both development and progression of pancreatic cancer. By focusing on current experimental models, this review elucidates potential cellular mechanisms underlying not only the pathophysiology of pancreatic steatosis itself, but also the pathogenesis behind pancreatic steatosis's role in changing the tumour microenvironment and accelerating the development of pancreatic cancer. This review further explores the impact of bariatric surgery on pancreatic steatosis and pancreatic cancer. Synthesizing knowledge from both epidemiologic studies and experimental animal models, this review identifies gaps in current knowledge regarding pancreatic steatosis and its role in carcinogenesis and proposes future research directions to elucidate the possible mechanisms underlying other obesity-associated cancers.
Collapse
Affiliation(s)
- Emily Truong
- Department of Medicine; Cedars-Sinai Medical Center, Los Angeles, California.
| | - Stephen Pandol
- Department of Medicine; Cedars-Sinai Medical Center, Los Angeles, California
| | - Christie Jeon
- Department of Medicine; Cedars-Sinai Medical Center, Los Angeles, California; UCLA Fielding School of Public Health, Los Angeles, CA
| |
Collapse
|
38
|
Lee JH, Lee J. Endoplasmic Reticulum (ER) Stress and Its Role in Pancreatic β-Cell Dysfunction and Senescence in Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23094843. [PMID: 35563231 PMCID: PMC9104816 DOI: 10.3390/ijms23094843] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023] Open
Abstract
An increased life span and accompanying nutritional affluency have led to a rapid increase in diseases associated with aging, such as obesity and type 2 diabetes, imposing a tremendous economic and health burden on society. Pancreatic β-cells are crucial for controlling glucose homeostasis by properly producing and secreting the glucose-lowering hormone insulin, and the dysfunction of β-cells determines the outcomes for both type 1 and type 2 diabetes. As the native structure of insulin is formed within the endoplasmic reticulum (ER), ER homeostasis should be appropriately maintained to allow for the proper metabolic homeostasis and functioning of β-cells. Recent studies have found that cellular senescence is critically linked with cellular stresses, including ER stress, oxidative stress, and mitochondrial stress. These studies implied that β-cell senescence is caused by ER stress and other cellular stresses and contributes to β-cells’ dysfunction and the impairment of glucose homeostasis. This review documents and discusses the current understanding of cellular senescence, β-cell function, ER stress, its associated signaling mechanism (unfolded protein response), and the effect of ER stress on β-cell senescence and dysfunction.
Collapse
Affiliation(s)
- Ji-Hye Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Correspondence:
| |
Collapse
|
39
|
Misfolding-induced chronic pancreatitis in CPA1 N256K mutant mice is unaffected by global deletion of Ddit3/Chop. Sci Rep 2022; 12:6357. [PMID: 35428786 PMCID: PMC9012826 DOI: 10.1038/s41598-022-09595-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic mutations in pancreatic digestive enzymes may cause protein misfolding, endoplasmic reticulum (ER) stress and chronic pancreatitis. The CPA1 N256K mouse model carries the human p.N256K carboxypeptidase A1 (CPA1) mutation, a classic example of a pancreatitis-associated misfolding variant. CPA1 N256K mice develop spontaneous, progressive chronic pancreatitis with moderate acinar atrophy, acinar-to-ductal metaplasia, fibrosis, and macrophage infiltration. Upregulation of the ER-stress associated pro-apoptotic transcription factor Ddit3/Chop mRNA was observed in the pancreas of CPA1 N256K mice suggesting that acinar cell death might be mediated through this mechanism. Here, we crossed the CPA1 N256K strain with mice containing a global deletion of the Ddit3/Chop gene (Ddit3-KO mice) and evaluated the effect of DDIT3/CHOP deficiency on the course of chronic pancreatitis. Surprisingly, CPA1 N256K x Ddit3-KO mice developed chronic pancreatitis with a similar time course and features as the CPA1 N256K parent strain. In contrast, Ddit3-KO mice showed no pancreas pathology. The observations indicate that DDIT3/CHOP plays no significant role in the development of misfolding-induced chronic pancreatitis in CPA1 N256K mice and this transcription factor is not a viable target for therapeutic intervention in this disease.
Collapse
|
40
|
Mugabo Y, Zhao C, Tan JJ, Ghosh A, Campbell SA, Fadzeyeva E, Paré F, Pan SS, Galipeau M, Ast J, Broichhagen J, Hodson DJ, Mulvihill EE, Petropoulos S, Lim GE. 14-3-3ζ constrains insulin secretion by regulating mitochondrial function in pancreatic β-cells. JCI Insight 2022; 7:156378. [PMID: 35298439 PMCID: PMC9089799 DOI: 10.1172/jci.insight.156378] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
While critical for neurotransmitter synthesis, 14-3-3 proteins are often assumed to have redundant functions due to their ubiquitous expression, but despite this assumption, various 14-3-3 isoforms have been implicated in regulating metabolism. We previously reported contributions of 14-3-3ζ in β cell function, but these studies were performed in tumor-derived MIN6 cells and systemic KO mice. To further characterize the regulatory roles of 14-3-3ζ in β cell function, we generated β cell–specific 14-3-3ζ–KO mice. Although no effects on β cell mass were detected, potentiated glucose-stimulated insulin secretion (GSIS), mitochondrial function, and ATP synthesis were observed. Deletion of 14-3-3ζ also altered the β cell transcriptome, as genes associated with mitochondrial respiration and oxidative phosphorylation were upregulated. Acute 14-3-3 protein inhibition in mouse and human islets recapitulated the enhancements in GSIS and mitochondrial function, suggesting that 14-3-3ζ is the critical isoform in β cells. In dysfunctional db/db islets and human islets from type 2 diabetic donors, expression of Ywhaz/YWHAZ, the gene encoding 14-3-3ζ, was inversely associated with insulin secretion, and pan–14-3-3 protein inhibition led to enhanced GSIS and mitochondrial function. Taken together, this study demonstrates important regulatory functions of 14-3-3ζ in the regulation of β cell function and provides a deeper understanding of how insulin secretion is controlled in β cells.
Collapse
Affiliation(s)
- Yves Mugabo
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Cheng Zhao
- Division of Obstetrics and Gynecology, Department of Clinical Science, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Ju Jing Tan
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, Canada
| | - Anindya Ghosh
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Scott A Campbell
- Cardiometabolic Axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, Canada
| | - Evgenia Fadzeyeva
- Energy Substrate Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Frédéric Paré
- Cardiometabolic Axis, Centre de recherche du CHUM (CRCHUM), Montreal, Canada
| | - Siew Siew Pan
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Maria Galipeau
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Julia Ast
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Pro, University of Birmingham, Birmingham, United Kingdom
| | - Johannes Broichhagen
- Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Pro, University of Birmingham, Birmingham, United Kingdom
| | - Erin E Mulvihill
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | | | - Gareth E Lim
- Department of Medicine, Université de Montréal, Montreal, Canada
| |
Collapse
|
41
|
Yan B, Chen L, Wang Y, Zhang J, Zhao H, Hua Q, Pei S, Yue Z, Liang H, Zhang H. Preventive Effect of Apple Polyphenol Extract on High-Fat Diet-Induced Hepatic Steatosis in Mice through Alleviating Endoplasmic Reticulum Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3172-3180. [PMID: 35227062 DOI: 10.1021/acs.jafc.1c07733] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In this work, the protective effect of apple polyphenol extract (APE) on hepatic steatosis was investigated. Thirty-two C57BL/6J mice were assigned randomly to control group, hepatic steatosis group, lovastatin group, and APE group. After 8 weeks of intervention, APE supplementation markedly decreased the body weight gain, liver weight, liver index, epididymal adipose weight, epididymal adipose index, serum, and hepatic lipid levels. Hematoxylin and eosin staining revealed that APE supplementation alleviated histopathological changes of hepatic steatosis. Western blot revealed that APE downregulated the protein levels of GRP78, IRE1α, p-IRE1α, XBP1, PERK, p-PERK, p-eIF2α, ATF6, PPAR-γ, SREBP-1c, FAS, and ACC1. In conclusion, this study found that APE inhibited IRE1α-XBP1, PERK-eIF2α, and ATF6 signaling pathways to alleviate endoplasmic reticulum stress, thereby improving HFD-induced hepatic steatosis.
Collapse
Affiliation(s)
- Bei Yan
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Lei Chen
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yanhui Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Jiacheng Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hui Zhao
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Qinglian Hua
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Shengjie Pei
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Zihang Yue
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
42
|
Zheng X, Xu L, Ye M, Gu T, Yao YL, Lv LB, Yu D, Yao YG. Characterizing the role of Tupaia DNA damage inducible transcript 3 (DDIT3) gene in viral infections. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104307. [PMID: 34748795 DOI: 10.1016/j.dci.2021.104307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 06/13/2023]
Abstract
DNA damage inducible transcript 3 (DDIT3, also known as CHOP) belongs to the CCAAT/enhancer-binding protein (C/EBP) family and plays an essential role in endoplasmic reticulum stress. Here, we characterized the potential role of the Chinese tree shrew (Tupaia belangeri chinensis) DDIT3 (tDDIT3) in viral infections. The tDDIT3 protein is highly conserved and has a species-specific insertion of the SQSS repeat upstream of the C-terminal basic-leucine zipper (bZIP) domain. Phylogenetic analysis of DDIT3 protein sequences of tree shrew and related mammals indicated a closer genetic affinity between tree shrew and primates than between tree shrew and rodents. Three positively selected sites (PSSs: Glu83, Pro93, and Ser172) were identified in tDDIT3 based on the branch-site model. Expression analysis of tDDIT3 showed a constitutively expressed level in different tissues and a significantly increased level in tree shrew cells upon herpes simplex virus type 1 (HSV-1) and Newcastle disease virus (NDV) infections. Overexpression of tDDIT3 significantly increased the production of HSV-1 and vesicular stomatitis virus (VSV) in tree shrew primary renal cells (TSPRCs), whereas tDDIT3 knockout in tree shrew stable cell line (TSR6 cells) had an inhibitory effect on virus production. The enhanced effect on viral infection by tDDIT3 was not associated with the three PSSs. Mechanistically, tDDIT3 overexpression inhibited type I IFN signaling. tDDIT3 interacted with tMAVS through CARD and PRR domains, but not with other immune-related factors such as tMDA5, tSTING and tTBK1. Collectively, our results revealed tDDIT3 as a negative regulator for virus infection.
Collapse
Affiliation(s)
- Xiao Zheng
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Maosen Ye
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Tianle Gu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yu-Lin Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Long-Bao Lv
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| |
Collapse
|
43
|
Radwan E, Belmadani S, Matrougui K. Disrupting Interleukin 12 Improves Microvascular Endothelial Function in Type 2 Diabetes Through ER Stress CHOP and Oxidative Stress Mechanisms. Diabetes Metab Syndr Obes 2022; 15:2633-2642. [PMID: 36065460 PMCID: PMC9440700 DOI: 10.2147/dmso.s369488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/08/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Vascular endothelial dysfunction is well established in type 2 diabetes. Interleukin-12 (IL-12) and endoplasmic reticulum (ER) stress are up-regulated in type 2 diabetic patients and animal models of type 2 diabetes. However, the role and underlying mechanisms of IL-12 and the ER stress CHOP in endothelial dysfunction are not fully understood. METHODS We generated double knockout mice between db-/db- and p40IL-12-/- mice (db-/db-p40-IL-12-/-) and endoplasmic (ER) stress-CHOP-/- mice (db-/db-CHOP-/-). We performed a glucose tolerance test (GTT) to determine the effect of IL-12 and ER stress CHOP on glucose metabolism. We assessed the endothelial function and determined the phosphorylation level of eNOS, Akt, AMPK, and the expression of ER stress (CHOP, BIP), and oxidative stress (Nox2 and Nox4 and NADPH oxidase activity). RESULTS The results showed that GTT was improved in db-/db-p40-IL-12-/- and db-/db-CHOP-/- suggesting IL-12 and CHOP as parts of a mechanism involved in the development of type 2 diabetes. The microvascular endothelial dysfunction in db-/db- mouse is associated with decreased phosphorylated eNOS, Akt, AMPK, and increased CHOP, BIP, Nox2, and Nox4 expressions. Interestingly, disrupting IL-12 and ER stress CHOP in db-/db- mice significantly improved endothelial function, increased survival markers expression and decreased ER and oxidative stress. CONCLUSION Using a genetic approach, these findings provide evidence that IL-12 and ER stress CHOP play a significant role in microvascular endothelial dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Eman Radwan
- Department of Physiological Sciences, EVMS, Norfolk, VA, 23501, USA
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Asyut, Egypt
| | - Souad Belmadani
- Department of Physiological Sciences, EVMS, Norfolk, VA, 23501, USA
| | - Khalid Matrougui
- Department of Physiological Sciences, EVMS, Norfolk, VA, 23501, USA
- Correspondence: Khalid Matrougui, Department of Physiological Sciences, EVMS, Norfolk, VA, 23501, USA, Tel +1 757-446-5278, Email
| |
Collapse
|
44
|
Sahin GS, Lee H, Engin F. An accomplice more than a mere victim: The impact of β-cell ER stress on type 1 diabetes pathogenesis. Mol Metab 2021; 54:101365. [PMID: 34728341 PMCID: PMC8606542 DOI: 10.1016/j.molmet.2021.101365] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pancreatic β-cells are the insulin factory of an organism with a mission to regulate glucose homeostasis in the body. Due to their high secretory activity, β-cells rely on a functional and intact endoplasmic reticulum (ER). Perturbations to ER homeostasis and unmitigated stress lead to β-cell dysfunction and death. Type 1 diabetes (T1D) is a chronic inflammatory disease caused by the autoimmune-mediated destruction of β-cells. Although autoimmunity is an essential component of T1D pathogenesis, accumulating evidence suggests an important role of β-cell ER stress and aberrant unfolded protein response (UPR) in disease initiation and progression. SCOPE OF REVIEW In this article, we introduce ER stress and the UPR, review β-cell ER stress in various mouse models, evaluate its involvement in inflammation, and discuss the effects of ER stress on β-cell plasticity and demise, and islet autoimmunity in T1D. We also highlight the relationship of ER stress with other stress response pathways and provide insight into ongoing clinical studies targeting ER stress and the UPR for the prevention or treatment of T1D. MAJOR CONCLUSIONS Evidence from ex vivo studies, in vivo mouse models, and tissue samples from patients suggest that β-cell ER stress and a defective UPR contribute to T1D pathogenesis. Thus, restoration of β-cell ER homeostasis at various stages of disease presents a plausible therapeutic strategy for T1D. Identifying the specific functions and regulation of each UPR sensor in β-cells and uncovering the crosstalk between stressed β-cells and immune cells during T1D progression would provide a better understanding of the molecular mechanisms of disease process, and may reveal novel targets for development of effective therapies for T1D.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA; Department of Cell & Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
45
|
Kalwat MA, Scheuner D, Rodrigues-dos-Santos K, Eizirik DL, Cobb MH. The Pancreatic ß-cell Response to Secretory Demands and Adaption to Stress. Endocrinology 2021; 162:bqab173. [PMID: 34407177 PMCID: PMC8459449 DOI: 10.1210/endocr/bqab173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic β cells dedicate much of their protein translation capacity to producing insulin to maintain glucose homeostasis. In response to increased secretory demand, β cells can compensate by increasing insulin production capability even in the face of protracted peripheral insulin resistance. The ability to amplify insulin secretion in response to hyperglycemia is a critical facet of β-cell function, and the exact mechanisms by which this occurs have been studied for decades. To adapt to the constant and fast-changing demands for insulin production, β cells use the unfolded protein response of the endoplasmic reticulum. Failure of these compensatory mechanisms contributes to both type 1 and 2 diabetes. Additionally, studies in which β cells are "rested" by reducing endogenous insulin demand have shown promise as a therapeutic strategy that could be applied more broadly. Here, we review recent findings in β cells pertaining to the metabolic amplifying pathway, the unfolded protein response, and potential advances in therapeutics based on β-cell rest.
Collapse
Affiliation(s)
- Michael A Kalwat
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Donalyn Scheuner
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | | | - Decio L Eizirik
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|