1
|
Parmar T, Parmar V, Malek G. Potential Role of NUR77 in the Aging Retinal Pigment Epithelium and Age-Related Macular Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:165-169. [PMID: 39930190 DOI: 10.1007/978-3-031-76550-6_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
The underlying mechanisms associated with age-related changes in the morphology and function of retinal pigmented epithelial (RPE) cells are poorly understood. The aging RPE progresses through several structural changes including loss of melanin granules, accumulation of lipofuscin, and cytoskeletal changes, among others. Extracellular to it, there is also thickening of Bruch's membrane and changes in the integrity of the choroid. Recent studies have revealed that aging also affects the metabolic ecosystem of the RPE. Aged mitochondria exhibit decreased rates of oxidative phosphorylation, increased reactive oxygen species generation, and increased number of mitochondrial mutations relative to baseline. These changes are also found in age-related macular degeneration (AMD), a late-onset vision-impairing disease, in which the RPE is particularly vulnerable. The orphan nuclear receptor NR4A1/NUR77 is an early response gene and regulator of various cellular processes during development, aging, and disease. Previously we observed decreased levels of Nur77/NUR77 in both mouse and human RPE as a function of age. Current knowledge of the function of this receptor in the RPE is limited. Herein, we discuss the putative roles of NUR77 in the RPE during aging and disease.
Collapse
Affiliation(s)
- Tanu Parmar
- Departments of Ophthalmology, Duke University School of Medicine, Durham, NC, USA.
- Albert Eye Research Institute, Durham, NC, USA.
| | - Vipul Parmar
- Departments of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Goldis Malek
- Departments of Ophthalmology, Duke University School of Medicine, Durham, NC, USA.
- Albert Eye Research Institute, Durham, NC, USA.
- Departments of Pathology, Duke University School of Medicine, Durham, NC, USA.
- Departments of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
2
|
Lan Z, Yang Y, Sun R, Lin X, Yan J, Chen X, Tian K, Wu G, Saad M, Wu Z, Xue D, Jin Q. Characterization of PANoptosis-related genes with immunoregulatory features in osteoarthritis. Int Immunopharmacol 2024; 140:112889. [PMID: 39128418 DOI: 10.1016/j.intimp.2024.112889] [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: 05/13/2024] [Revised: 08/04/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
This study aimed to characterize PANoptosis-related genes with immunoregulatory features in osteoarthritis (OA) and investigate their potential diagnostic and therapeutic implications. Gene expression data from OA patients and healthy controls were obtained from the Gene Expression Omnibus (GEO) database. Differential expression analysis and functional enrichment analysis were conducted to identify PANoptosis-related genes (PRGs) associated with OA pathogenesis. A diagnostic model was developed using LASSO regression, and the diagnostic value of key PRGs was evaluated using Receiver Operating Characteristic Curve (ROC) analysis. The infiltration of immune cells and potential small molecule agents were also examined. A total of 39 differentially expressed PANoptosis-related genes (DE-PRGs) were identified, with functional enrichment analysis revealing their involvement in inflammatory response regulation and immune modulation pathways. Seven key PRGs, including CDKN1A, EZH2, MEG3, NR4A1, PIK3R2, S100A8, and SYVN1, were selected for diagnostic model construction, demonstrating high predictive performance in both training and validation datasets. The correlation between key PRGs and immune cell infiltration was explored. Additionally, molecular docking analysis identified APHA-compound-8 as a potential therapeutic agent targeting key PRGs. This study identified and analyzed PRGs in OA, uncovering their roles in immune regulation. Seven key PRGs were used to construct a diagnostic model with high predictive performance. The identified PRGs' correlation with immune cell infiltration was elucidated, and APHA-compound-8 was highlighted as a potential therapeutic agent. These findings offer novel diagnostic markers and therapeutic targets for OA, warranting further in vivo validation and exploration of clinical applications.
Collapse
Affiliation(s)
- Zhibin Lan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yang Yang
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Rui Sun
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xue Lin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Jiangbo Yan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xiaolei Chen
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Kuanmin Tian
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Gang Wu
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Muhammad Saad
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Zhiqiang Wu
- Quanzhou Orthopedic-Traumatological Hospital, Quanzhou, China
| | - Di Xue
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| | - Qunhua Jin
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China; Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
3
|
Sheethal G, Verma A, Mall R, Parsa KV, Tokala RK, Bynigeri R, Pondugala PK, Vemula K, Sai Latha S, Sowpati DT, Singh SS, Rao GV, Talukdar R, Kanneganti TD, Reddy DN, Sasikala M. Modulation of Nuclear Receptor 4A1 Expression Improves Insulin Secretion in a Mouse Model of Chronic Pancreatitis. Pancreas 2024; 53:e760-e773. [PMID: 38710022 DOI: 10.1097/mpa.0000000000002370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
OBJECTIVES Diabetes secondary to chronic pancreatitis (CP) presents clinical challenges due to lack of understanding on factor(s) triggering insulin secretory defects. Therefore, we aimed to delineate the molecular mechanism of β-cell dysfunction in CP. MATERIALS AND METHODS Transcriptomic analysis was conducted to identify endocrine-specific receptor expression in mice and human CP on microarray. The identified receptor (NR4A1) was overexpressed in MIN6 cells using PEI linear transfection. RNA-Seq analysis of NR4A1-overexpressed (OE) MIN6 cells on NovaSeq6000 identified aberrant metabolic pathways. Upstream trigger for NR4A1OE was studied by InBio Discover and cytokine exposure, whereas downstream effect was examined by Fura2 AM-based fluorimetric and imaging studies. Mice with CP were treated with IFN-γ-neutralizing monoclonal antibodies to assess NR4A1 expression and insulin secretion. RESULTS Increased expression of NR4A1 associated with decreased insulin secretion in islets (humans: controls 9 ± 0.2, CP 3.7 ± 0.2, mice: controls 8.5 ± 0.2, CP 2.1 ± 0.1 μg/L). NR4A1OE in MIN6 cells (13.2 ± 0.1) showed reduction in insulin secretion (13 ± 5 to 0.2 ± 0.1 μg/mg protein per minute, P = 0.001) and downregulation of calcium and cAMP signaling pathways. IFN-γ was identified as upstream signal for NR4A1OE in MIN6. Mice treated with IFN-γ-neutralizing antibodies showed decreased NR4A1 expression 3.4 ± 0.11-fold ( P = 0.03), showed improved insulin secretion (4.4 ± 0.2-fold, P = 0.01), and associated with increased Ca 2+ levels (2.39 ± 0.06-fold, P = 0.009). CONCLUSIONS Modulating NR4A1 expression can be a promising therapeutic strategy to improve insulin secretion in CP.
Collapse
Affiliation(s)
| | - Archana Verma
- CSIR-CCMB, Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Raghvendra Mall
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN
| | - Kishore Vl Parsa
- Centre for Innovation in Molecular and Pharmaceutical Sciences, Dr Reddy's Institute of Life Sciences
| | | | - Ratnakar Bynigeri
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN
| | | | | | - S Sai Latha
- Centre for Innovation in Molecular and Pharmaceutical Sciences, Dr Reddy's Institute of Life Sciences
| | - Divya Tej Sowpati
- CSIR-CCMB, Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - G V Rao
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Rupjyoti Talukdar
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | | | - D Nageshwar Reddy
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | | |
Collapse
|
4
|
Sharma R, Maity SK, Chakrabarti P, Katika MR, Kapettu S, Parsa KVL, Misra P. PIMT Controls Insulin Synthesis and Secretion through PDX1. Int J Mol Sci 2023; 24:ijms24098084. [PMID: 37175791 PMCID: PMC10179560 DOI: 10.3390/ijms24098084] [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: 01/06/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic beta cell function is an important component of glucose homeostasis. Here, we investigated the function of PIMT (PRIP-interacting protein with methyl transferase domain), a transcriptional co-activator binding protein, in the pancreatic beta cells. We observed that the protein levels of PIMT, along with key beta cell markers such as PDX1 (pancreatic and duodenal homeobox 1) and MafA (MAF bZIP transcription factor A), were reduced in the beta cells exposed to hyperglycemic and hyperlipidemic conditions. Consistently, PIMT levels were reduced in the pancreatic islets isolated from high fat diet (HFD)-fed mice. The RNA sequencing analysis of PIMT knockdown beta cells identified that the expression of key genes involved in insulin secretory pathway, Ins1 (insulin 1), Ins2 (insulin 2), Kcnj11 (potassium inwardly-rectifying channel, subfamily J, member 11), Kcnn1 (potassium calcium-activated channel subfamily N member 1), Rab3a (member RAS oncogene family), Gnas (GNAS complex locus), Syt13 (synaptotagmin 13), Pax6 (paired box 6), Klf11 (Kruppel-Like Factor 11), and Nr4a1 (nuclear receptor subfamily 4, group A, member 1) was attenuated due to PIMT depletion. PIMT ablation in the pancreatic beta cells and in the rat pancreatic islets led to decreased protein levels of PDX1 and MafA, resulting in the reduction in glucose-stimulated insulin secretion (GSIS). The results from the immunoprecipitation and ChIP experiments revealed the interaction of PIMT with PDX1 and MafA, and its recruitment to the insulin promoter, respectively. Importantly, PIMT ablation in beta cells resulted in the nuclear translocation of insulin. Surprisingly, forced expression of PIMT in beta cells abrogated GSIS, while Ins1 and Ins2 transcript levels were subtly enhanced. On the other hand, the expression of genes, PRIP/Asc2/Ncoa6 (nuclear receptor coactivator 6), Pax6, Kcnj11, Syt13, Stxbp1 (syntaxin binding protein 1), and Snap25 (synaptosome associated protein 25) associated with insulin secretion, was significantly reduced, providing an explanation for the decreased GSIS upon PIMT overexpression. Our findings highlight the importance of PIMT in the regulation of insulin synthesis and secretion in beta cells.
Collapse
Affiliation(s)
- Rahul Sharma
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, India
| | - Sujay K Maity
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Madhumohan R Katika
- Central Research Lab Mobile Virology Research & Diagnostics BSL3 Lab, ESIC Medical College and Hospital, Hyderabad 500038, India
| | - Satyamoorthy Kapettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Kishore V L Parsa
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, India
| | - Parimal Misra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, India
| |
Collapse
|
5
|
Chung JY, Ma Y, Zhang D, Bickerton HH, Stokes E, Patel SB, Tse HM, Feduska J, Welner RS, Banerjee RR. Pancreatic islet cell type-specific transcriptomic changes during pregnancy and postpartum. iScience 2023; 26:106439. [PMID: 37020962 PMCID: PMC10068570 DOI: 10.1016/j.isci.2023.106439] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/11/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Pancreatic β-cell mass expands during pregnancy and regresses in the postpartum period in conjunction with dynamic metabolic demands on maternal glucose homeostasis. To understand transcriptional changes driving these adaptations in β-cells and other islet cell types, we performed single-cell RNA sequencing on islets from virgin, late gestation, and early postpartum mice. We identified transcriptional signatures unique to gestation and the postpartum in β-cells, including induction of the AP-1 transcription factor subunits and other genes involved in the immediate-early response (IEGs). In addition, we found pregnancy and postpartum-induced changes differed within each endocrine cell type, and in endothelial cells and antigen-presenting cells within islets. Together, our data reveal insights into cell type-specific transcriptional changes responsible for adaptations by islet cells to pregnancy and their resolution postpartum.
Collapse
Affiliation(s)
- Jin-Yong Chung
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Yongjie Ma
- Department of Pharmacology, the University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Dingguo Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Hayden H. Bickerton
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Eric Stokes
- Department of Pharmacology, University of Colorado Denver/Anschutz, Aurora, CO 80045, USA
| | - Sweta B. Patel
- Division of Hematology and Oncology, Department of Medicine, The University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Hubert M. Tse
- Department of Microbiology, the University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Joseph Feduska
- Department of Microbiology, the University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Rob S. Welner
- Division of Hematology and Oncology, Department of Medicine, The University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Ronadip R. Banerjee
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| |
Collapse
|
6
|
Pei Y, Liu C, Feng M, Li L, Zhou C, Chen L, Hu X, Song S, Cao Y, Gao Y. The clinical application of 68Ga-PSMA PET/CT and regulating mechanism of PSMA expression in patients with brain metastases of lung cancer. Transl Oncol 2023; 28:101616. [PMID: 36621073 PMCID: PMC9850174 DOI: 10.1016/j.tranon.2023.101616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023] Open
Abstract
Brain metastases (BMs) of lung cancer are common malignant intracranial tumours associated with severe neurological symptoms and an abysmal prognosis. Prostate-specific membrane antigen (PSMA) has been reported to express significantly in a variety of solid tumours. However, the clinical applications of 68Ga-PSMA PET/CT and the mechanism of PSMA expression in patients with BMs of lung cancer have rarely been reported. Experiments with 68Ga-PSMA PET/CT and immunohistochemical staining were conducted to evaluate the expression of PSMA from seven patients with BMs of lung cancer who accepted surgical treatment in Fudan University Shanghai Cancer Center between October 2020 and October 2021. The mechanism of PSMA expression in BMs of lung cancer was explored by using single-cell RNA sequencing. The median maximum standardized uptake value (SUVmax) in BMs was higher than that in primary lung cancer (8.6 ± 2.8 vs. 3.6 ± 1.3, P < 0.01). The mean SUVmax in BMs was 1.76-fold higher than that in the liver, which indicated the potential of PSMA radioligand therapy (PSMA-RLT) for BMs. BMs showed intense PSMA staining, while normal lung tissue had no PSMA staining and there was only faint primary lung cancer staining by immunohistochemistry (IHC). Single-cell RNA sequencing (scRNA-seq) analysis found that PSMA was mainly expressed in oligodendrocytes of BMs, whereas it was expressed at lower levels in solid cells of lung cancer. PSMA expression in oligodendrocytes might be regulated by the factors ATF3 and NR4A1, which were associated with ER stress.
Collapse
Affiliation(s)
- Yuchen Pei
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mingtao Feng
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Changshuai Zhou
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Chen
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin Hu
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yiqun Cao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yang Gao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China,Corresponding author.
| |
Collapse
|
7
|
Zandi Shafagh R, Youhanna S, Keulen J, Shen JX, Taebnia N, Preiss LC, Klein K, Büttner FA, Bergqvist M, van der Wijngaart W, Lauschke VM. Bioengineered Pancreas-Liver Crosstalk in a Microfluidic Coculture Chip Identifies Human Metabolic Response Signatures in Prediabetic Hyperglycemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203368. [PMID: 36285680 PMCID: PMC9731722 DOI: 10.1002/advs.202203368] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/05/2022] [Indexed: 05/19/2023]
Abstract
Aberrant glucose homeostasis is the most common metabolic disturbance affecting one in ten adults worldwide. Prediabetic hyperglycemia due to dysfunctional interactions between different human tissues, including pancreas and liver, constitutes the largest risk factor for the development of type 2 diabetes. However, this early stage of metabolic disease has received relatively little attention. Microphysiological tissue models that emulate tissue crosstalk offer emerging opportunities to study metabolic interactions. Here, a novel modular multitissue organ-on-a-chip device is presented that allows for integrated and reciprocal communication between different 3D primary human tissue cultures. Precisely controlled heterologous perfusion of each tissue chamber is achieved through a microfluidic single "synthetic heart" pneumatic actuation unit connected to multiple tissue chambers via specific configuration of microchannel resistances. On-chip coculture experiments of organotypic primary human liver spheroids and intact primary human islets demonstrate insulin secretion and hepatic insulin response dynamics at physiological timescales upon glucose challenge. Integration of transcriptomic analyses with promoter motif activity data of 503 transcription factors reveals tissue-specific interacting molecular networks that underlie β-cell stress in prediabetic hyperglycemia. Interestingly, liver and islet cultures show surprising counter-regulation of transcriptional programs, emphasizing the power of microphysiological coculture to elucidate the systems biology of metabolic crosstalk.
Collapse
Affiliation(s)
- Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Jibbe Keulen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Lena C Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, 64293, Darmstadt, Germany
| | - Kathrin Klein
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Florian A Büttner
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Mikael Bergqvist
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| |
Collapse
|
8
|
Yoshihara E. Adapting Physiology in Functional Human Islet Organogenesis. Front Cell Dev Biol 2022; 10:854604. [PMID: 35557947 PMCID: PMC9086403 DOI: 10.3389/fcell.2022.854604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of three-dimensional (3D)-structured functional human islets is expected to be an alternative cell source for cadaveric human islet transplantation for the treatment of insulin-dependent diabetes. Human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer infinite resources for newly synthesized human islets. Recent advancements in hPSCs technology have enabled direct differentiation to human islet-like clusters, which can sense glucose and secrete insulin, and those islet clusters can ameliorate diabetes when transplanted into rodents or non-human primates (NHPs). However, the generated hPSC-derived human islet-like clusters are functionally immature compared with primary human islets. There remains a challenge to establish a technology to create fully functional human islets in vitro, which are functionally and transcriptionally indistinguishable from cadaveric human islets. Understanding the complex differentiation and maturation pathway is necessary to generate fully functional human islets for a tremendous supply of high-quality human islets with less batch-to-batch difference for millions of patients. In this review, I summarized the current progress in the generation of 3D-structured human islets from pluripotent stem cells and discussed the importance of adapting physiology for in vitro functional human islet organogenesis and possible improvements with environmental cues.
Collapse
Affiliation(s)
- Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
9
|
Zhang S, Peng X, Yang S, Li X, Huang M, Wei S, Liu J, He G, Zheng H, Yang L, Li H, Fan Q. The regulation, function, and role of lipophagy, a form of selective autophagy, in metabolic disorders. Cell Death Dis 2022; 13:132. [PMID: 35136038 PMCID: PMC8825858 DOI: 10.1038/s41419-022-04593-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/07/2022] [Accepted: 01/27/2022] [Indexed: 12/15/2022]
Abstract
Autophagy is a conserved method of quality control in which cytoplasmic contents are degraded via lysosomes. Lipophagy, a form of selective autophagy and a novel type of lipid metabolism, has recently received much attention. Lipophagy is defined as the autophagic degradation of intracellular lipid droplets (LDs). Although much remains unknown, lipophagy appears to play a significant role in many organisms, cell types, metabolic states, and diseases. It participates in the regulation of intracellular lipid storage, intracellular free lipid levels (e.g., fatty acids), and energy balance. However, it remains unclear how intracellular lipids regulate autophagy. Impaired lipophagy can cause cells to become sensitive to death stimuli and may be responsible for the onset of a variety of diseases, including nonalcoholic fatty liver disease and metabolic syndrome. Like autophagy, the role of lipophagy in cancer is poorly understood, although analysis of specific autophagy receptors has helped to expand the diversity of chemotherapeutic targets. These studies have stimulated increasing interest in the role of lipophagy in the pathogenesis and treatment of cancer and other human diseases.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Mingyao Huang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyu Zheng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Qing Fan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
10
|
Xu Y, Tian J, Kang Q, Yuan H, Liu C, Li Z, Liu J, Li M. Knockout of Nur77 Leads to Amino Acid, Lipid, and Glucose Metabolism Disorders in Zebrafish. Front Endocrinol (Lausanne) 2022; 13:864631. [PMID: 35547009 PMCID: PMC9084189 DOI: 10.3389/fendo.2022.864631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/14/2022] [Indexed: 12/25/2022] Open
Abstract
Orphan nuclear receptor Nur77 has been reported to be implicated in a diverse range of metabolic processes, including carbohydrate metabolism and lipid metabolism. However, the detailed mechanism of Nur77 in the regulation of metabolic pathway still needs to be further investigated. In this study, we created a global nur77 knockout zebrafish model by CRISPR/Cas9 technique, and then performed whole-organism RNA sequencing analysis in wildtype and nur77-deficient zebrafish to dissect the genetic changes in metabolic-related pathways. We found that many genes involved in amino acid, lipid, and carbohydrate metabolism changed by more than twofold. Furthermore, we revealed that nur77-/- mutant displayed increased total cholesterol (TC) and triglyceride (TG), alteration in total amino acids, as well as elevated glucose. We also demonstrated that the elevated glucose was not due to the change of glucose uptake but was likely caused by the disorder of glycolysis/gluconeogenesis and the impaired β-cell function, including downregulated insb expression, reduced β-cell mass, and suppressed insulin secretion. Importantly, we also verified that targeted expression of Nur77 in the β cells is sufficient to rescue the β-cell defects in global nur77-/- larvae zebrafish. These results provide new information about the global metabolic network that Nur77 signaling regulates, as well as the role of Nur77 in β-cell function.
Collapse
Affiliation(s)
- Yang Xu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Juanjuan Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qi Kang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hang Yuan
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chengdong Liu
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Zhehui Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- *Correspondence: Mingyu Li, ; Jie Liu,
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- *Correspondence: Mingyu Li, ; Jie Liu,
| |
Collapse
|
11
|
Nuclear Receptors in Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:61-82. [DOI: 10.1007/978-3-031-11836-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Zhang J, Ni Y, Qian L, Fang Q, Zheng T, Zhang M, Gao Q, Zhang Y, Ni J, Hou X, Bao Y, Kovatcheva‐Datchary P, Xu A, Li H, Panagiotou G, Jia W. Decreased Abundance of Akkermansia muciniphila Leads to the Impairment of Insulin Secretion and Glucose Homeostasis in Lean Type 2 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100536. [PMID: 34085773 PMCID: PMC8373164 DOI: 10.1002/advs.202100536] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/05/2021] [Indexed: 05/23/2023]
Abstract
Although obesity occurs in most of the patients with type 2 diabetes (T2D), a fraction of patients with T2D are underweight or have normal weight. Several studies have linked the gut microbiome to obesity and T2D, but the role of gut microbiota in lean individuals with T2D having unique clinical characteristics remains unclear. A metagenomic and targeted metabolomic analysis is conducted in 182 lean and abdominally obese individuals with and without newly diagnosed T2D. The abundance of Akkermansia muciniphila (A. muciniphila) significantly decreases in lean individuals with T2D than without T2D, but not in the comparison of obese individuals with and without T2D. Its abundance correlates inversely with serum 3β-chenodeoxycholic acid (βCDCA) levels and positively with insulin secretion and fibroblast growth factor 15/19 (FGF15/19) concentrations. The supplementation with A. muciniphila is sufficient to protect mice against high sucrose-induced impairment of glucose intolerance by decreasing βCDCA and increasing insulin secretion and FGF15/19. Furthermore, βCDCA inhibits insulin secretion and FGF15/19 expression. These findings suggest that decreased abundance of A. muciniphila is linked to the impairment of insulin secretion and glucose homeostasis in lean T2D, paving the way for new therapeutic options for the prevention or treatment of diabetes.
Collapse
Affiliation(s)
- Jing Zhang
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yueqiong Ni
- Systems Biology and Bioinformatics UnitLeibniz Institute for Natural Product Research and Infection Biology–Hans Knöll InstituteBeutenbergstrasse 11aJena07745Germany
- Systems Biology & Bioinformatics GroupSchool of Biological SciencesThe University of Hong KongHong Kong SARChina
| | - Lingling Qian
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Qichen Fang
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Tingting Zheng
- Systems Biology and Bioinformatics UnitLeibniz Institute for Natural Product Research and Infection Biology–Hans Knöll InstituteBeutenbergstrasse 11aJena07745Germany
- Systems Biology & Bioinformatics GroupSchool of Biological SciencesThe University of Hong KongHong Kong SARChina
| | - Mingliang Zhang
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Qiongmei Gao
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Ying Zhang
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Jiacheng Ni
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xuhong Hou
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yuqian Bao
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | | | - Aimin Xu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of MedicineThe University of Hong KongHong Kong SARChina
| | - Huating Li
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Gianni Panagiotou
- Systems Biology and Bioinformatics UnitLeibniz Institute for Natural Product Research and Infection Biology–Hans Knöll InstituteBeutenbergstrasse 11aJena07745Germany
- Systems Biology & Bioinformatics GroupSchool of Biological SciencesThe University of Hong KongHong Kong SARChina
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of MedicineThe University of Hong KongHong Kong SARChina
| | - Weiping Jia
- Shanghai Key Laboratory of Diabetes MellitusDepartment of Endocrinology and MetabolismShanghai Diabetes InstituteShanghai Clinical Center for DiabetesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
13
|
NR4A1 enhances MKP7 expression to diminish JNK activation induced by ROS or ER-stress in pancreatic β cells for surviving. Cell Death Discov 2021; 7:133. [PMID: 34088892 PMCID: PMC8178316 DOI: 10.1038/s41420-021-00521-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/18/2021] [Accepted: 05/13/2021] [Indexed: 12/03/2022] Open
Abstract
Under adverse conditions, such as sustained or chronic hyperglycemia or hyperlipidemia, ROS (reactive oxygen species) or/and ER-stress (endoplasmic reticulum stress) will be induced in pancreatic β cells. ROS or ER-stress damages β-cells even leads to apoptosis. Previously we found ROS or ER-stress resulted in JNK activation in β cells and overexpressing NR4A1 in MIN6 cells reduced JNK activation via modulating cbl-b expression and subsequent degrading the upstream JNK kinase (MKK4). To search other possible mechanisms, we found the mRNA level and protein level of MKP7 (a phosphatase for phospho-JNK) were dramatic reduced in pancreatic β cells in the islets from NR4A1 KO mice compared with that from wild type mice. To confirm what we found in animals, we applied pancreatic β cells (MIN6 cells) and found that the expression of MKP7 was increased in NR4A1-overexpression MIN6 cells. We further found that knocking down the expression of MKP7 increased the p-JNK level in pancreatic β cells upon treatment with TG or H2O2. After that, we figured out that NR4A1 did enhance the transactivation of the MKP7 promoter by physical association with two putative binding sites. In sum, NR4A1 attenuates JNK phosphorylation incurred by ER-stress or ROS partially via enhancing MKP7 expression, potentially decreases pancreatic β cell apoptosis induced by ROS or ER-stress. Our finding provides a clue for diabetes prevention.
Collapse
|
14
|
Pu ZQ, Liu D, Lobo Mouguegue HPP, Jin CW, Sadiq E, Qin DD, Yu TF, Zong C, Chen JC, Zhao RX, Lin JY, Cheng J, Yu X, Li X, Zhang YC, Liu YT, Guan QB, Wang XD. NR4A1 counteracts JNK activation incurred by ER stress or ROS in pancreatic β-cells for protection. J Cell Mol Med 2020; 24:14171-14183. [PMID: 33124187 PMCID: PMC7754045 DOI: 10.1111/jcmm.16028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Sustained hyperglycaemia and hyperlipidaemia incur endoplasmic reticulum stress (ER stress) and reactive oxygen species (ROS) overproduction in pancreatic β‐cells. ER stress or ROS causes c‐Jun N‐terminal kinase (JNK) activation, and the activated JNK triggers apoptosis in different cells. Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an inducible multi‐stress response factor. The aim of this study was to explore the role of NR4A1 in counteracting JNK activation induced by ER stress or ROS and the related mechanism. qPCR, Western blotting, dual‐luciferase reporter and ChIP assays were applied to detect gene expression or regulation by NR4A1. Immunofluorescence was used to detect a specific protein expression in β‐cells. Our data showed that NR4A1 reduced the phosphorylated JNK (p‐JNK) in MIN6 cells encountering ER stress or ROS and reduced MKK4 protein in a proteasome‐dependent manner. We found that NR4A1 increased the expression of cbl‐b (an E3 ligase); knocking down cbl‐b expression increased MKK4 and p‐JNK levels under ER stress or ROS conditions. We elucidated that NR4A1 enhanced the transactivation of cbl‐b promoter by physical association. We further confirmed that cbl‐b expression in β‐cells was reduced in NR4A1‐knockout mice compared with WT mice. NR4A1 down‐regulates JNK activation by ER stress or ROS in β‐cells via enhancing cbl‐b expression.
Collapse
Affiliation(s)
- Ze-Qing Pu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Dong Liu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | | | - Cheng-Wen Jin
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Esha Sadiq
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Dan-Dan Qin
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Tian-Fu Yu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Chen Zong
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Ji-Cui Chen
- Blood Transfusion Department, Qilu Hospital of Shandong University, Jinan, China
| | - Ru-Xing Zhao
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing-Yu Lin
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Jie Cheng
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Xiao Yu
- Department of Physiology, Shandong University School of Medicine, Jinan, China.,Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan, China
| | - Xia Li
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Yu-Chao Zhang
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China
| | - Yuan-Tao Liu
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China
| | - Qing-Bo Guan
- Department of Endocrinology, Shandong Provincial Hospital, Affiliated to Shandong University, Jinan, China
| | - Xiang-Dong Wang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China.,Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan, China
| |
Collapse
|
15
|
Zhao T, Ma J, Li L, Teng W, Tian Y, Ma Y, Wang W, Yan W, Jiao P. MKP-5 Relieves Lipotoxicity-Induced Islet β-Cell Dysfunction and Apoptosis via Regulation of Autophagy. Int J Mol Sci 2020; 21:ijms21197161. [PMID: 32998359 PMCID: PMC7582937 DOI: 10.3390/ijms21197161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 01/28/2023] Open
Abstract
Mitogen-activated protein kinase phosphatase-5 (MKP-5) is a regulator of extracellular signaling that is known to regulate lipid metabolism. In this study, we found that obesity caused by a high-fat diet (HFD) decreased the expression of MKP-5 in the pancreas and primary islet cells derived from mice. Then, we further investigated the role of MKP-5 in the protection of islet cells from lipotoxicity by modulating MKP-5 expression. As a critical inducer of lipotoxicity, palmitic acid (PA) was used to treat islet β-cells. We found that MKP-5 overexpression restored PA-mediated autophagy inhibition in Rin-m5f cells and protected these cells from PA-induced apoptosis and dysfunction. Consistently, a lack of MKP-5 aggravated the adverse effects of lipotoxicity. Islet cells from HFD-fed mice were infected using recombinant adenovirus expressing MKP-5 (Ad-MKP-5), and we found that Ad-MKP-5 was able to alleviate HFD-induced apoptotic protein activation and relieve the HFD-mediated inhibition of functional proteins. Notably, HFD-mediated impairments in autophagic flux were restored by Ad-MKP-5 transduction. Furthermore, the autophagy inhibitor 3-methyladenine (3-MA) was used to treat Rin-m5f cells, confirming that the MKP-5 overexpression suppressed apoptosis, dysfunction, inflammatory response, and oxidative stress induced by PA via improving autophagic signaling. Lastly, employing c-Jun amino-terminal kinas (JNK), P38, or extracellular-regulated kinase (ERK) inhibitors, we established that the JNK and P38 MAPK pathways were involved in the MKP-5-mediated apoptosis, dysfunction, and autophagic inhibition observed in islet β cells in response to lipotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weiqun Yan
- Correspondence: (W.Y.); (P.J.); Tel.: +86-431-8561-9289 (P.J.)
| | - Ping Jiao
- Correspondence: (W.Y.); (P.J.); Tel.: +86-431-8561-9289 (P.J.)
| |
Collapse
|
16
|
Lu J, Shen H, Li Q, Xiong F, Xie R, Yuan M, Yang JK. KCNH6 protects pancreatic β-cells from endoplasmic reticulum stress and apoptosis. FASEB J 2020; 34:15015-15028. [PMID: 32918525 DOI: 10.1096/fj.202001218r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/15/2020] [Accepted: 08/31/2020] [Indexed: 01/08/2023]
Abstract
Adult patients with dysfunction in human ether-a-go-go 2 (hERG2) protein, encoded by KCNH6, present with hypoinsulinemia and hyperglycemia. However, the mechanism of KCNH6 action in glucose disorders has not been clearly defined. Previous studies identified that sustained endoplasmic reticulum (ER) stress-mediated apoptosis of pancreatic β-cells and directly contributed to diabetes. In the present study, we showed that Kcnh6 knockout (KO) mice had impaired glucose tolerance mediated by high ER stress levels, and showed increased apoptosis and elevated intracellular calcium levels in pancreatic β-cells. In contrast, KCNH6 overexpression in islets isolated from C57BL/6J mice attenuated ER stress induced by thapsigargin or palmitic acid. This effect contributed to better preservation of β-cells, as reflected in increased β cell survival and enhanced glucose-stimulated insulin secretion. These results were further corroborated by studies evaluating KCNH6 overexpression in KO islets. Similarly, induction of Kcnh6 in KO mice by lentivirus injection improved glucose tolerance by reducing pancreatic ER stress and apoptosis. Our data provide new insights into how Kcnh6 deficiency causes ER calcium depletion and β cell dysfunction.
Collapse
Affiliation(s)
- Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Han Shen
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Sijiqing Hospital, Beijing, China
| | - Qi Li
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Fengran Xiong
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rongrong Xie
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Mingxia Yuan
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Close AF, Dadheech N, Lemieux H, Wang Q, Buteau J. Disruption of Beta-Cell Mitochondrial Networks by the Orphan Nuclear Receptor Nor1/Nr4a3. Cells 2020; 9:cells9010168. [PMID: 31936632 PMCID: PMC7017372 DOI: 10.3390/cells9010168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/14/2022] Open
Abstract
Nor1, the third member of the Nr4a subfamily of nuclear receptor, is garnering increased interest in view of its role in the regulation of glucose homeostasis. Our previous study highlighted a proapoptotic role of Nor1 in pancreatic beta cells and showed that Nor1 expression was increased in islets isolated from type 2 diabetic individuals, suggesting that Nor1 could mediate the deterioration of islet function in type 2 diabetes. However, the mechanism remains incompletely understood. We herein investigated the subcellular localization of Nor1 in INS832/13 cells and dispersed human beta cells. We also examined the consequences of Nor1 overexpression on mitochondrial function and morphology. Our results show that, surprisingly, Nor1 is mostly cytoplasmic in beta cells and undergoes mitochondrial translocation upon activation by proinflammatory cytokines. Mitochondrial localization of Nor1 reduced glucose oxidation, lowered ATP production rates, and inhibited glucose-stimulated insulin secretion. Western blot and microscopy images revealed that Nor1 could provoke mitochondrial fragmentation via mitophagy. Our study unveils a new mode of action for Nor1, which affects beta-cell viability and function by disrupting mitochondrial networks.
Collapse
Affiliation(s)
- Anne-Françoise Close
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Nidheesh Dadheech
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Hélène Lemieux
- Faculty Saint-Jean, Department of Medicine, University of Alberta, Edmonton, AB T6C 4G9, Canada
| | - Qian Wang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jean Buteau
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Correspondence: ; Tel.: +1-780-492-8386
| |
Collapse
|
18
|
Peng T, Liu X, Wang J, Liu Y, Fu Z, Ma X, Li J, Sun G, Ji Y, Lu J, Wan W, Lu H. Fluoxetine-mediated inhibition of endoplasmic reticulum stress is involved in the neuroprotective effects of Parkinson's disease. Aging (Albany NY) 2019; 10:4188-4196. [PMID: 30585175 PMCID: PMC6326670 DOI: 10.18632/aging.101716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/06/2018] [Indexed: 12/25/2022]
Abstract
Background: Accumulating evidence suggests that Fluoxetine (FLX), an anti-depressant drug, has broad neurobiological functions and neuroprotective effects in central nervous system injury, but its roles in Parkinson's disease (PD) remain unclear. In this study, we aimed to evaluate whether fluoxetine attenuates rotenone-induced neurodegeneration in PD. Methods: Male Sprague-Dawley rats were randomly allocated to control, rotenone-treated, rotenone + FLX-treated and FLX-treated groups. Behavioral tests including open field behavioral test and catalepsy measurement were taken to evaluate neurological behavioral measurements. Apoptosis was detected by TUNEL assay. Endoplasmic reticulum (ER)-related gene expressions were detected by qRT-PCR and western blot. Immunohistochemistry was performed to assess dopaminergic neuronal degeneration. Results: We demonstrated that pretreatment with FLX (10.0 mg/kg, i.p.) significantly ameliorated the catalepsy symptom and increased locomotor activity. In addition, FLX markedly reversed the loss of dopaminergic neurons and suppressed the X‑box‑binding protein 1 (XBP1)/caspase-3-activated ER stress. Furthermore, FLX inhibited rotenone-mediated neurodegeneration through caspase-3-mediated neuronal apoptosis. Conclusion: Taken together, our findings indicate that FLX has beneficial neuroprotective effects in PD and FLX might be a potential therapeutic agent for the treatment of PD. In light of its favorable properties, FLX should be evaluated in the treatment of PD as well as related neurologic disorders.
Collapse
Affiliation(s)
- Tao Peng
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xiaoyan Liu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jingtao Wang
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu Liu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhenqiang Fu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xingrong Ma
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Junmin Li
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Guifang Sun
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yangfei Ji
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jingjing Lu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Wencui Wan
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hong Lu
- Key-Disciplines Laboratory Clinical Medicine Henan, Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
19
|
Function of Nr4a Orphan Nuclear Receptors in Proliferation, Apoptosis and Fuel Utilization Across Tissues. Cells 2019; 8:cells8111373. [PMID: 31683815 PMCID: PMC6912296 DOI: 10.3390/cells8111373] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/21/2022] Open
Abstract
The Nr4a family of nuclear hormone receptors is composed of three members-Nr4a1/Nur77, Nr4a2/Nurr1 and Nr4a3/Nor1. While currently defined as ligandless, these transcription factors have been shown to regulate varied processes across a host of tissues. Of particular interest, the Nr4a family impinge, in a tissue dependent fashion, on cellular proliferation, apoptosis and fuel utilization. The regulation of these processes occurs through both nuclear and non-genomic pathways. The purpose of this review is to provide a balanced perspective of the tissue specific and Nr4a family member specific, effects on cellular proliferation, apoptosis and fuel utilization.
Collapse
|
20
|
Wang L, Ning N, Wang C, Hou X, Yuan Y, Ren Y, Sun C, Yan Z, Wang X, Liu H. Endoplasmic reticulum stress contributed to β1-adrenoceptor autoantibody-induced reduction of autophagy in cardiomyocytes. Acta Biochim Biophys Sin (Shanghai) 2019; 51:1016-1025. [PMID: 31553425 DOI: 10.1093/abbs/gmz089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/22/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022] Open
Abstract
Autophagy reduction has been confirmed as an important mechanism in apoptosis induction. Our previous study showed that decreased autophagy induced by β1-adrenoceptor autoantibodies (β1-AAs) enhanced cardiomyocyte apoptosis and contributed to heart failure progression. Endoplasmic reticulum stress (ERS) is known to be an important mechanism in intracellular homeostasis and is closely related to autophagy. However, ERS in β1-AA-induced autophagy dysfunction of cardiomyocytes remains unclear. In this study, we used an active immunization rat model and H9c2 cardiomyocytes to study the role of ERS in β1-AA-induced autophagy. Results showed that prolonged action of β1-AAs significantly reduced the autophagy of myocardial tissues and H9c2 cardiomyocytes, and ERS and its related apoptotic pathways were significantly activated. Moreover, mRFP-GFP-LC3 double-labeled adenoviruses were used to detect cardiomyocyte autophagic flux to confirm that β1-AAs caused a significant decrease in autophagic flux in H9c2 cardiomyocytes. The ERS inhibitor, 4-phenylbutyrate (4-PBA), partially attenuated the β1-AA-induced reduction of cardiomyocyte autophagy, consistent with the effect of the mammalian target of rapamycin inhibitor rapamycin (Rapa). Compared to the pretreatment with 4-PBA or Rapa alone, pretreatment with the combination of 4-PBA and Rapa had a greater effect on attenuating the β1-AA-induced decrease in autophagy and β1-AA-induced apoptosis in cardiomyocytes. This study provides an experimental basis for the role of β1-AAs in the homeostatic maintenance of cardiomyocytes in patients with heart failure with respect to autophagy and ERS.
Collapse
Affiliation(s)
- Li Wang
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Na Ning
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Changtu Wang
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaohong Hou
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Yuan Yuan
- Laboratory of Morphology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Yanan Ren
- Department of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Cong Sun
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Zi Yan
- Department of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaohui Wang
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
- Laboratory of Morphology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Huirong Liu
- Department of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
21
|
Keller MP, Rabaglia ME, Schueler KL, Stapleton DS, Gatti DM, Vincent M, Mitok KA, Wang Z, Ishimura T, Simonett SP, Emfinger CH, Das R, Beck T, Kendziorski C, Broman KW, Yandell BS, Churchill GA, Attie AD. Gene loci associated with insulin secretion in islets from non-diabetic mice. J Clin Invest 2019; 129:4419-4432. [PMID: 31343992 DOI: 10.1172/jci129143] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Genetic susceptibility to type 2 diabetes is primarily due to β-cell dysfunction. However, a genetic study to directly interrogate β-cell function ex vivo has never been previously performed. We isolated 233,447 islets from 483 Diversity Outbred (DO) mice maintained on a Western-style diet, and measured insulin secretion in response to a variety of secretagogues. Insulin secretion from DO islets ranged >1,000-fold even though none of the mice were diabetic. The insulin secretory response to each secretagogue had a unique genetic architecture; some of the loci were specific for one condition, whereas others overlapped. Human loci that are syntenic to many of the insulin secretion QTL from mouse are associated with diabetes-related SNPs in human genome-wide association studies. We report on three genes, Ptpn18, Hunk and Zfp148, where the phenotype predictions from the genetic screen were fulfilled in our studies of transgenic mouse models. These three genes encode a non-receptor type protein tyrosine phosphatase, a serine/threonine protein kinase, and a Krϋppel-type zinc-finger transcription factor, respectively. Our results demonstrate that genetic variation in insulin secretion that can lead to type 2 diabetes is discoverable in non-diabetic individuals.
Collapse
Affiliation(s)
- Mark P Keller
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Mary E Rabaglia
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Kathryn L Schueler
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Donnie S Stapleton
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | | | | | - Kelly A Mitok
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Ziyue Wang
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | | | - Shane P Simonett
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | | | - Rahul Das
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| | - Tim Beck
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christina Kendziorski
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | - Karl W Broman
- University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | - Brian S Yandell
- University of Wisconsin-Madison, Department of Horticulture, Madison, Wisconsin, USA
| | | | - Alan D Attie
- University of Wisconsin-Madison, Biochemistry Department, Madison, Wisconsin, USA
| |
Collapse
|
22
|
Qin H, Gao F, Wang Y, Huang B, Peng L, Mo B, Wang C. Nur77 promotes cigarette smoke‑induced autophagic cell death by increasing the dissociation of Bcl2 from Beclin-1. Int J Mol Med 2019; 44:25-36. [PMID: 31115481 PMCID: PMC6559304 DOI: 10.3892/ijmm.2019.4184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/08/2019] [Indexed: 01/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by partially reversible airflow limitation and persistent alveolar destruction, and autophagy is involved in the pathogenesis of cigarette smoke (CS)‑induced COPD. Nuclear receptor 77 (Nur77) participates in a number of biological processes, including apoptosis, autophagy and in disease pathogenesis; however, the role of Nur77 in COPD remains unknown. Thus, in this study, we aimed to elucidate the role of Nur77 in COPD. We report that CS promotes Nur77 expression and nuclear export in vivo and in vitro, which increases cigarette smoke extract (CSE)‑induced autophagy. In addition, we found that lung tissues, human bronchial epithelial (HBE) cells and A549 cells exposed to CS or CSE expressed lower levels of LC3 and Beclin‑1 and contained fewer autophagosomes following Nur77 knockdown with siRNA‑Nur77. Moreover, a co‑immunoprecipitation assay demonstrated that CSE promoted autophagy, partly by accelerating the interaction between Nur77 and Bcl2, in turn leading to the increased dissociation of Bcl2 from Beclin‑1; by contrast, leptomycin B (LMB) suppressed the dissociation of Bcl2 from Beclin‑1. Taken together, the findings of this study demonstrate that Nur77 is involved in the CSE‑induced autophagic death of lung cells, and that this process is partially dependent on the increased interaction between Nur77 and Bcl2, and on the dissociation of Bcl2 from Beclin‑1. This study illustrates the role of Nur77 in bronchial and alveolar destruction following exposure to CS.
Collapse
Affiliation(s)
- Huiping Qin
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of The National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Feng Gao
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541002
| | - Yanni Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of The National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Bin Huang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541002
| | - Ling Peng
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of The National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Biwen Mo
- Department of Respiratory Medicine, Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Changming Wang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541002,Correspondence to: Dr Changming Wang, Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, 12 Wenming Road, Guilin, Guangxi 541002, P.R. China, E-mail:
| |
Collapse
|
23
|
Carew NT, Nelson AM, Liang Z, Smith SM, Milcarek C. Linking Endoplasmic Reticular Stress and Alternative Splicing. Int J Mol Sci 2018; 19:ijms19123919. [PMID: 30544499 PMCID: PMC6321306 DOI: 10.3390/ijms19123919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/16/2022] Open
Abstract
RNA splicing patterns in antibody-secreting cells are shaped by endoplasmic reticulum stress, ELL2 (eleven-nineteen lysine-rich leukemia gene 2) induction, and changes in the levels of snRNAs. Endoplasmic reticulum stress induces the unfolded protein response comprising a highly conserved set of genes crucial for cell survival; among these is Ire1, whose auto-phosphorylation drives it to acquire a regulated mRNA decay activity. The mRNA-modifying function of phosphorylated Ire1 non-canonically splices Xbp1 mRNA and yet degrades other cellular mRNAs with related motifs. Naïve splenic B cells will activate Ire1 phosphorylation early on after lipopolysaccharide (LPS) stimulation, within 18 h; large-scale changes in mRNA content and splicing patterns result. Inhibition of the mRNA-degradation function of Ire1 is correlated with further differences in the splicing patterns and a reduction in the mRNA factors for snRNA transcription. Some of the >4000 splicing changes seen at 18 h after LPS stimulation persist into the late stages of antibody secretion, up to 72 h. Meanwhile some early splicing changes are supplanted by new splicing changes introduced by the up-regulation of ELL2, a transcription elongation factor. ELL2 is necessary for immunoglobulin secretion and does this by changing mRNA processing patterns of immunoglobulin heavy chain and >5000 other genes.
Collapse
Affiliation(s)
- Nolan T Carew
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Ashley M Nelson
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Zhitao Liang
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Sage M Smith
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Christine Milcarek
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| |
Collapse
|
24
|
Wei X, Zhu D, Feng C, Chen G, Mao X, Wang Q, Wang J, Liu C. Inhibition of peptidyl-prolyl cis-trans isomerase B mediates cyclosporin A-induced apoptosis of islet β cells. Exp Ther Med 2018; 16:3959-3964. [PMID: 30344674 PMCID: PMC6176207 DOI: 10.3892/etm.2018.6706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 08/29/2018] [Indexed: 12/21/2022] Open
Abstract
Cyclosporin A (CsA) is widely used as an immunosuppressor in the context of organ transplantation or autoimmune disorders. Recent studies have revealed the detrimental effects of CsA on insulin resistance and pancreatic β cell failure; however, the molecular mechanisms are unknown. The present study sought to confirm the associations between CsA and β cell failure, and to investigate the roles of proinsulin folding and endoplasmic reticulum (ER) stress in CsA-induced β cell failure. The viability of MIN6 cells treated with CsA was evaluated with MTT assay. Expression levels of insulin, peptidyl-prolyl cis-trans isomerase B (PPIB), cleaved caspase-3, phospho-protein kinase R (PKR)-like endoplasmic reticulum kinase (p-PERK), PKR-like endoplasmic reticulum kinase (PERK), binding immunoglobulin protein (BIP), and C/EBP homologous protein (CHOP) were detected via reducing western blot assay. Non-reducing western blot analysis was performed to examine the expression of misfolded proinsulin peptides. The proliferation of MIN6 cells was not inhibited by CsA at concentrations <1 µmol/l. CsA treatment resulted in the decreased expression of insulin and PPIB; however, it also increased the phosphorylation of PERK, and upregulated the expression of PERK, BIP, CHOP and cleaved caspase-3. The results indicated that CsA could induce pancreatic β cell dysfunction and the potential mechanism underlying this phenomenon may be PPIB-associated proinsulin misfolding, which in turn induces ER stress in β cells.
Collapse
Affiliation(s)
- Xiao Wei
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Dan Zhu
- Department of Endocrinology, Jiangdu People's Hospital of Yangzhou, Yangzhou, Jiangsu 225200, P.R. China
| | - Chenchen Feng
- Central Laboratory, Jiangsu Province Blood Center, Nanjing, Jiangsu 210042, P.R. China
| | - Guofang Chen
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Xiaodong Mao
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Qifeng Wang
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Jie Wang
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Chao Liu
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| |
Collapse
|
25
|
Qin DD, Yang YF, Pu ZQ, Liu D, Yu C, Gao P, Chen JC, Zong C, Zhang YC, Li X, Wang XD, Liu YT. NR4A1 retards adipocyte differentiation or maturation via enhancing GATA2 and p53 expression. J Cell Mol Med 2018; 22:4709-4720. [PMID: 30044048 PMCID: PMC6156289 DOI: 10.1111/jcmm.13715] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/02/2018] [Indexed: 12/27/2022] Open
Abstract
Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an orphan nuclear receptor with diverse functions. It has been reported that NR4A1, as a transcriptional activator, is implicated in glucose and lipid metabolism. The aim of this study was to investigate the regulatory role of NR4A1 in adipogenesis and explore the underlying mechanisms. Quantitative real‐time PCR and Western blotting were used to analyse the expression of genes involved in synthesis and mobilization of fats in vivo and in vitro. Dual‐luciferase reporter assay was conducted to study the regulatory mechanisms of NR4A1. Our data from in vivo study confirmed that NR4A1 knockout (KO) mice fed with high‐fat diet were more prone to obesity, and gene expression levels of PPARγ and FAS were increased in KO mice compared to controls; our data from in vitro study showed that NR4A1 overexpression in 3T3‐L1 pre‐adipocytes inhibited adipogenesis. Moreover, NR4A1 enhanced GATA binding protein 2 (GATA2) expression, which in turn inhibited peroxisome proliferator‐activated receptor γ (PPARγ); NR4A1 inhibited sterol regulatory element binding transcription factor 1 (SREBP1) and its downstream gene fatty acid synthase (FAS) by up‐regulating p53. NR4A1 inhibits the differentiation and lipid accumulation of adipocytes by enhancing the expression of GATA2 and p53.
Collapse
Affiliation(s)
- Dan-Dan Qin
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Ying-Feng Yang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Ze-Qing Pu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Dong Liu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Cong Yu
- Department of Life Science, Qilu Normal University, Jinan, China
| | - Peng Gao
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Ji-Cui Chen
- Department of Blood Transfusion of Qilu Hospital, Shandong University, Jinan, China
| | - Chen Zong
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Yu-Chao Zhang
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China
| | - Xia Li
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Xiang-Dong Wang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Yuan-Tao Liu
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
26
|
The orphan nuclear receptor NR4A1 attenuates oxidative stress-induced β cells apoptosis via up-regulation of glutathione peroxidase 1. Life Sci 2018; 203:225-232. [DOI: 10.1016/j.lfs.2018.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/07/2018] [Accepted: 04/17/2018] [Indexed: 12/18/2022]
|
27
|
Tecalco-Cruz AC. Molecular pathways involved in the transport of nuclear receptors from the nucleus to cytoplasm. J Steroid Biochem Mol Biol 2018; 178:36-44. [PMID: 29107180 DOI: 10.1016/j.jsbmb.2017.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/18/2017] [Accepted: 10/25/2017] [Indexed: 12/30/2022]
Abstract
Nuclear receptors (NRs) are transcription regulators that direct the expression of many genes linked to cellular processes, such as proliferation, differentiation, and apoptosis. Additionally, some cellular events are also modulated by signaling pathways induced by NRs outside of the nucleus. Hence, the subcellular transport of NRs is dynamic and is modulated by several signals, protein-protein interactions, and posttranslational modifications. Particularly, the exit of NRs from the nucleus to cytoplasm and/or other compartments is transcendental, as it is this export event, which determines their abundance in the cells' compartments, the activation or attenuation of nuclear or extranuclear pathways, and the magnitude and duration of their effects inside or outside of the nucleus. Consequently, an adequate control of the distribution of NRs is critical for homeostasis, because a deregulation in the nucleo-cytoplasmic transport of NRs could be involved in diseases including cancer as well as metabolic and vascular alterations. In this review, we investigated the pathways and molecular and biological aspects that have been described for the nuclear export of NRs so far and their functional relevance in some diseases. This information suggests that the transport of NRs out of the nucleus is a key mechanism for the identification of new therapeutic targets for alterations associated with the deregulation of the function of NRs.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apdo Postal, D.F. 04510, Mexico.
| |
Collapse
|
28
|
Choi J, Kobayashi H, Okuda H, Harada KH, Takeda M, Fujimoto H, Yamane S, Tanaka D, Youssefian S, Inagaki N, Koizumi A. β-cell-specific overexpression of adiponectin receptor 1 does not improve diabetes mellitus in Akita mice. PLoS One 2018; 13:e0190863. [PMID: 29304075 PMCID: PMC5755906 DOI: 10.1371/journal.pone.0190863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 12/21/2017] [Indexed: 01/09/2023] Open
Abstract
Adiponectin, a metabolically-active cytokine secreted from adipose tissue, is reported to have anti-apoptotic effects on β-cells as well as anti-hyperglycemic effects through adiponectin receptor signaling. However, the anti-apoptotic effects of adiponectin on β-cells have not been confirmed in established diabetic models, and the anti-hyperglycemic effects and their associated signal cascades remain controversial. To investigate the effects of adiponectin on β-cell protection and its down-stream signaling events, we have generated β-cell-specific rat insulin promoter (RIP)-AdipoR1 transgenic mice (AdipoR1 mice), in which the adiponectin receptor, AdipoR1, is overexpressed in β-cells in a manner synchronous with insulin demand. AdipoR1 mice were then mated with Akita mice, a diabetes model in which β-cell apoptosis results from endoplasmic reticulum (ER) stress. AdipoR1 protein expression and localization in islets from AdipoR1 mice as well as in an AdipoR1-transfected mouse insulinoma cell line were confirmed, as was the activation of both AMPK and Akt in AdipoR1 mice by adiponectin. Nevertheless, there were no significant differences in Ad lib feed and fasting blood glucose levels, or in glucose tolerance tests, between Akita mice [Ins2Akita (C96Y) +/- mouse model] and AdipoR1/Akita and from 4 weeks to 10 weeks of age. Similarly, pancreatic insulin contents of AdipoR1/Akita mice were not significantly different from those in Akita mice from 15 to 20 weeks of age, but they were significantly lower than in wild-type mice. Immunostaining for insulin and subsequent electron microscopy showed that β-cell destruction in AdipoR1/Akita mice was not markedly improved in comparison with that in Akita mice. Serum adiponectin concentrations were confirmed to be extremely high (> 30 μg / ml) compared with the Kd value (0.06 μg / ml) in all mouse groups at 15 to 20 weeks of age. Therefore, although the physiological levels of adiponectin are sufficient to activate AMPK and Akt when AdipoR1 is overexpressed in β-cells, yet adiponectin cannot protect β-cells in Akita mice from ER stress-induced destruction.
Collapse
Affiliation(s)
- Jungmi Choi
- Department of Health and Environmental Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hatasu Kobayashi
- Department of Health and Environmental Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Hiroko Okuda
- Department of Health and Environmental Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kouji H. Harada
- Department of Health and Environmental Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Midori Takeda
- Department of Health and Environmental Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Shunsuke Yamane
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Tanaka
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shohab Youssefian
- Laboratory of Molecular Biosciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akio Koizumi
- Department of Health and Environmental Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
29
|
Zong C, Qin D, Yu C, Gao P, Chen J, Lu S, Zhang Y, Liu Y, Yang Y, Pu Z, Li X, Fu Y, Guan Q, Wang X. The stress-response molecule NR4A1 resists ROS-induced pancreatic β-cells apoptosis via WT1. Cell Signal 2017; 35:129-139. [PMID: 28342843 DOI: 10.1016/j.cellsig.2017.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/14/2023]
Abstract
Pancreatic β-cells often face endoplasmic reticulum stress and/or ROS-associated oxidative stress under adverse conditions. Our previous work has verified that NR4A1 protects pancreatic β-cells from ER-stress induced apoptosis. However, It remains unknown whether NR4A1 is able to protect pancreatic β-cells against ROS-associated oxidative stress. In the present study, our data showed that NR4A1 protein expression rapidly increased in MIN6 cells upon H2O2 treatment, and overexpression of NR4A1 in MIN6 cells conferred resistance to cell apoptosis induced by H2O2. These results were further substantiated in isolated islets from mice infected with an adenovirus overexpressing NR4A1. 8-hydroxy-2'-deoxyguanosine (8-OHdG) was used as a biomarker for oxidative stress or a marker for ROS damage. We found that the 8-OHdG level in the islets from NR4A1 knockout mice fed with high-fat diet was much higher than that in the islets from parental control mice; and higher apoptotic rate was observed in the islets from NR4A1 KO mice compared to control mice. Further investigation of underlying mechanisms of NR4A1's protective effects showed that NR4A1 overexpression in MIN6 cells reduced Caspase 3 activation caused by H2O2, and increased expression of WT1 and SOD1. There is a putative NR4A1 binding site (-1118bp to -1111bp) in WT1 promoter; our data demonstrated that NR4A1 protein physically associates with the WT1 promoter, and enhanced WT1 promoter transactivation and knockdown of WT1 in MIN6 cells induced apoptosis. These findings suggest that NR4A1 protects pancreatic β-cells against H2O2 mediated apoptosis by up-regulating WT1 expression.
Collapse
Affiliation(s)
- Chen Zong
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China
| | - Dandan Qin
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China
| | - Cong Yu
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China
| | - Peng Gao
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China; Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China
| | - Jicui Chen
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China
| | - Sumei Lu
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong, China
| | - Yuchao Zhang
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao 266071, Shandong, China
| | - Yuantao Liu
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao 266071, Shandong, China
| | - Yingfeng Yang
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China
| | - Zeqing Pu
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China
| | - Xia Li
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China.
| | - Yuchang Fu
- The Department of Nutrition Sciences, University of Alabama at Birmingham, AL 35294, United States.
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Xiangdong Wang
- Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, Shandong, China; Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan 250012, Shandong, China.
| |
Collapse
|
30
|
The role and possible mechanism of lncRNA U90926 in modulating 3T3-L1 preadipocyte differentiation. Int J Obes (Lond) 2016; 41:299-308. [PMID: 27780975 PMCID: PMC5309343 DOI: 10.1038/ijo.2016.189] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/01/2016] [Accepted: 09/25/2016] [Indexed: 12/19/2022]
Abstract
Background: Obesity is a risk factor for metabolic diseases, while preadipocyte differentiation or adipogenesis is closely related to obesity occurrence. Long noncoding RNAs (lncRNAs) are a unique class of transcripts in regulation of a variety of biological processes. Using cDNA microarray, we found lncRNA U90926 is negatively correlated with 3T3-L1 preadipocyte differentiation. Objective: The aim of this study was to explore the role of lncRNA U90926 (lnc-U90926) in adipogenesis and the underlying mechanisms. Methods: Quantitative real-time PCR (qPCR) was performed to determine lnc-U90926 expression in 3T3-L1 preadipocytes, differentiated adipocytes, and in adipose tissues form mice. RNA fluorescent in situ hybridization (FISH) was performed to determine the localization of lnc-U90926 in 3T3-L1 preadipocytes. The effects of lnc-U90926 on 3T3-L1 adipogenesis were analyzed with lentivirus-mediated gain- and loss-of-function experiments. Lipid accumulation was evaluated by oil red O staining; several adipogenesis makers were analyzed by qPCR and western blotting. Dual luciferase assay was applied to explore the transactivation of target genes modulated by lnc-U90926. All measurements were performed at least for three times. Results: Lnc-U90926 expression decreased along the differentiation of 3T3-L1 preadipocytes. In mice, lnc-U90926 is predominantly expressed in adipose tissue. Obese mice have lower lnc-U90926 expression in subcutaneous and visceral adipose tissue than non-obese mice. FISH results showed that lnc-U90926 was mainly located in the cytoplasm. Overexpression lnc-U90926 attenuated 3T3-L1 adipocyte differentiation as evidenced by its ability to inhibit lipid accumulation, to decrease the mRNA levels of peroxisome proliferator-activated receptor gamma 2 (PPARγ2), fatty acid binding protein 4 (FABP4) and adiponectin (AdipoQ) as well as to reduce the protein levels of PPARγ and FABP4 (P<0.05). Knockdown of lnc-U90926 showed opposite effects, which increased mRNA expression of PPARγ2, FABP4, CCAAT/enhancer-binding proteinα (C/EBPα) and AdipoQ. Conclusion: Lnc-U90926 attenuates 3T3-L1 adipocyte differentiation via inhibiting the transactivation of PPARγ2 or PPARγ.
Collapse
|
31
|
Zhang HH, Ma XJ, Wu LN, Zhao YY, Zhang PY, Zhang YH, Shao MW, Liu F, Li F, Qin GJ. Sirtuin-3 (SIRT3) protects pancreatic β-cells from endoplasmic reticulum (ER) stress-induced apoptosis and dysfunction. Mol Cell Biochem 2016; 420:95-106. [PMID: 27449933 DOI: 10.1007/s11010-016-2771-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/09/2016] [Indexed: 01/07/2023]
Abstract
Insufficient insulin produced by pancreatic β-cells in the control of blood sugar is a central feature of the etiology of diabetes. Reports have shown that endoplasmic reticulum (ER) stress is fundamentally involved in β-cell dysfunction. In this study, we hypothesized that NAD-dependent deacetylase sirtuin-3 (SIRT3), an important regulator of cell metabolism, protects pancreatic β-cells from ER stress-mediated apoptosis. To validate our hypothesis, a rat diabetic model was established by a high-fat diet (HFD). We found that SIRT3 expression was markedly decreased in NIT1 and INS1 cells incubated with palmitate. Palmitate treatment significantly decreased β-cell viability and insulin secretion, and promoted malondialdehyde (MDA) formation. However, SIRT3 overexpression in NIT1 and INS1 cells reversed these effects, resulting in higher insulin secretion, decreased β-cell apoptosis, and downregulation of the expression of ER stress-associated genes. Moreover, SIRT3 overexpression also inhibited calcium influx and the hyperacetylation of glucose-regulated protein of 78 kDa (GRP78). SIRT3 knockdown effectively enhanced the upregulation of phospho-extracellular regulated protein kinases (pERK), inositol-requiring enzyme-1 (IRE1), activating transcription factor 6 (ATF6), and C/EBP homologous protein (CHOP) induced by palmitate, and promoted palmitate-induced β-cell apoptosis and dysfunction. Taken together, our results suggest that SIRT3 is an integral regulator of ER function and that its depletion might result in the hyperacetylation of critical ER proteins that protect against islet lipotoxicity under conditions of nutrient excess.
Collapse
Affiliation(s)
- Hao-Hao Zhang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Xiao-Jun Ma
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Li-Na Wu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Yan-Yan Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Peng-Yu Zhang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Ying-Hui Zhang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Ming-Wei Shao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Fei Liu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Fei Li
- Division of Vasculitis, Guancheng Traditional Chinese Medical Hospital, Shangdu Road, Zhengzhou, 450016, China
| | - Gui-Jun Qin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China.
| |
Collapse
|
32
|
Abstract
Type two diabetes (T2D) is a challenging metabolic disorder for which a cure has not yet been found. Its etiology is associated with several phenomena, including significant loss of insulin-producing, beta cell (β cell) mass via progressive programmed cell death and disrupted cellular autophagy. In diabetes, the etiology of β cell death and the role of mitochondria are complex and involve several layers of mechanisms. Understanding the dynamics of those mechanisms could permit researchers to develop an intervention for the progressive loss of β cells. Currently, diabetes research has shifted toward rejuvenation and plasticity technology and away from the simplified approach of hormonal compensation. Diabetes research is currently challenged by questions such as how to enhance cell survival, decrease apoptosis and replenish β cell mass in diabetic patients. In this review, we discuss evidence that β cell development and mass formation are guided by specific signaling systems, particularly hormones, transcription factors, and growth factors, all of which could be manipulated to enhance mass growth. There is also strong evidence that β cells are dynamically active cells, which, under specific conditions such as obesity, can increase in size and subsequently increase insulin secretion. In certain cases of aggressive or advanced forms of T2D, β cells become markedly impaired, and the only alternatives for maintaining glucose homeostasis are through partial or complete cell grafting (the Edmonton protocol). In these cases, the harvesting of an enriched population of viable β cells is required for transplantation. This task necessitates a deep understanding of the pharmacological agents that affect β cell survival, mass, and function. The aim of this review is to initiate discussion about the important signals in pancreatic β cell development and mass formation and to highlight the process by which cell death occurs in diabetes. This review also examines the attempts that have been made to recover or increase cell mass in diabetic patients by using various pharmacological agents.
Collapse
Affiliation(s)
- Husnia I Marrif
- Department of Pharmacology, Faculty of Medicine, University of Benghazi Benghazi, Libya
| | - Salma I Al-Sunousi
- Department of Histology and Anatomy, Faculty of Medicine, University of Benghazi Benghazi, Libya
| |
Collapse
|