1
|
Dekker E, Triñanes J, Muñoz Garcia A, de Graaf N, de Koning E, Carlotti F. Enhanced BMP Signaling Alters Human β-Cell Identity and Function. Adv Biol (Weinh) 2025; 9:e2400470. [PMID: 39499224 PMCID: PMC11760635 DOI: 10.1002/adbi.202400470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Indexed: 11/07/2024]
Abstract
Inflammation contributes to the pathophysiology of diabetes. Identifying signaling pathways involved in pancreatic β-cell failure and identity loss can give insight into novel potential treatment strategies to prevent the loss of functional β-cell mass in diabetes. It is reported earlier that the immunosuppressive drug tacrolimus has a detrimental effect on human β-cell identity and function by activating bone morphogenetic protein (BMP) signaling. Here it is hypothesized that enhanced BMP signaling plays a role in inflammation-induced β-cell failure. Single-cell transcriptomics analyses of primary human islets reveal that IL-1β+IFNγ and IFNα treatment activated BMP signaling in β-cells. These findings are validated by qPCR. Furthermore, enhanced BMP signaling with recombinant BMP2 or 4 triggers a reduced expression of key β-cell maturity genes, associated with increased ER stress, and impaired β-cell function. Altogether, these results indicate that inflammation-activated BMP signaling is detrimental to pancreatic β-cells and that BMP-signaling can be a target to preserve β-cell identity and function in a pro-inflammatory environment.
Collapse
Affiliation(s)
- Esmée Dekker
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333 ZAThe Netherlands
| | - Javier Triñanes
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333 ZAThe Netherlands
| | - Amadeo Muñoz Garcia
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333 ZAThe Netherlands
| | - Natascha de Graaf
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333 ZAThe Netherlands
| | - Eelco de Koning
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333 ZAThe Netherlands
| | - Françoise Carlotti
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333 ZAThe Netherlands
| |
Collapse
|
2
|
Vandenbempt V, Eski SE, Brahma MK, Li A, Negueruela J, Bruggeman Y, Demine S, Xiao P, Cardozo AK, Baeyens N, Martelotto LG, Singh SP, Mariño E, Gysemans C, Gurzov EN. HAMSAB diet ameliorates dysfunctional signaling in pancreatic islets in autoimmune diabetes. iScience 2024; 27:108694. [PMID: 38213620 PMCID: PMC10783594 DOI: 10.1016/j.isci.2023.108694] [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: 04/17/2023] [Revised: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
An altered gut microbiota is associated with type 1 diabetes (T1D), affecting the production of short-chain fatty acids (SCFA) and glucose homeostasis. We previously demonstrated that enhancing serum acetate and butyrate using a dietary supplement (HAMSAB) improved glycemia in non-obese diabetic (NOD) mice and patients with established T1D. The effects of SCFA on immune-infiltrated islet cells remain to be clarified. Here, we performed single-cell RNA sequencing on islet cells from NOD mice fed an HAMSAB or control diet. HAMSAB induced a regulatory gene expression profile in pancreas-infiltrated immune cells. Moreover, HAMSAB maintained the expression of β-cell functional genes and decreased cellular stress. HAMSAB-fed mice showed preserved pancreatic endocrine cell identity, evaluated by decreased numbers of poly-hormonal cells. Finally, SCFA increased insulin levels in human β-like cells and improved transplantation outcome in NOD/SCID mice. Our findings support the use of metabolite-based diet as attractive approach to improve glucose control in T1D.
Collapse
Affiliation(s)
- Valerie Vandenbempt
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Sema Elif Eski
- IRIBHM, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Manoja K. Brahma
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Ao Li
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Javier Negueruela
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Ylke Bruggeman
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Campus Gasthuisberg O&N 1, KU Leuven, 3000 Leuven, Belgium
| | - Stéphane Demine
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Peng Xiao
- Inflammatory and Cell Death Signaling in Diabetes group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Alessandra K. Cardozo
- Inflammatory and Cell Death Signaling in Diabetes group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Nicolas Baeyens
- Laboratoire de Physiologie et de Pharmacologie, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Luciano G. Martelotto
- Single Cell and Spatial-Omics Laboratory, Adelaide Centre of Epigenetics, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Eliana Mariño
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Biochemistry, Monash University, Melbourne, VIC 3800, Australia
- ImmunoBiota Therapeutics Pty Ltd, Melbourne, VIC 3187, Australia
| | - Conny Gysemans
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Campus Gasthuisberg O&N 1, KU Leuven, 3000 Leuven, Belgium
| | - Esteban N. Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
- WELBIO Department, WEL Research Institute, Avenue Pasteur 6, 1300 Wavre, Belgium
| |
Collapse
|
3
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
4
|
Urizar AI, Prause M, Ingerslev LR, Wortham M, Sui Y, Sander M, Williams K, Barrès R, Larsen MR, Christensen GL, Billestrup N. Beta cell dysfunction induced by bone morphogenetic protein (BMP)-2 is associated with histone modifications and decreased NeuroD1 chromatin binding. Cell Death Dis 2023; 14:399. [PMID: 37407581 DOI: 10.1038/s41419-023-05906-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
Insufficient insulin secretion is a hallmark of type 2 diabetes and has been attributed to beta cell identity loss characterized by decreased expression of several key beta cell genes. The pro-inflammatory factor BMP-2 is upregulated in islets of Langerhans from individuals with diabetes and acts as an inhibitor of beta cell function and proliferation. Exposure to BMP-2 induces expression of Id1-4, Hes-1, and Hey-1 which are transcriptional regulators associated with loss of differentiation. The aim of this study was to investigate the mechanism by which BMP-2 induces beta cell dysfunction and loss of cell maturity. Mouse islets exposed to BMP-2 for 10 days showed impaired glucose-stimulated insulin secretion and beta cell proliferation. BMP-2-induced beta cell dysfunction was associated with decreased expression of cell maturity and proliferation markers specific to the beta cell such as Ins1, Ucn3, and Ki67 and increased expression of Id1-4, Hes-1, and Hey-1. The top 30 most regulated proteins significantly correlated with corresponding mRNA expression. BMP-2-induced gene expression changes were associated with a predominant reduction in acetylation of H3K27 and a decrease in NeuroD1 chromatin binding activity. These results show that BMP-2 induces loss of beta cell maturity and suggest that remodeling of H3K27ac and decreased NeuroD1 DNA binding activity participate in the effect of BMP-2 on beta cell dysfunction.
Collapse
Affiliation(s)
| | - Michala Prause
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| | - Lars Roed Ingerslev
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Matthew Wortham
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yinghui Sui
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Kristine Williams
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice Côte d'Azur, Valbonne, France
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | - Nils Billestrup
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
5
|
Ma B, Wang X, Ren H, Li Y, Zhang H, Yang M, Li J. High glucose promotes the progression of colorectal cancer by activating the BMP4 signaling and inhibited by glucagon-like peptide-1 receptor agonist. BMC Cancer 2023; 23:594. [PMID: 37370018 PMCID: PMC10304216 DOI: 10.1186/s12885-023-11077-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The detailed molecular mechanism between type 2 diabetes mellitus (T2DM) and colorectal cancer (CRC) is still uncertain. Bone morphogenetic protein 4 (BMP4) dysregulation is implicated in T2DM and CRC, respectively. This study aims to investigate whether BMP4 can mediate the interaction of CRC with T2DM. METHODS We firstly explored the expression of BMP4 in The Cancer Genome Altas (TCGA) databases and CRC patients with or without DM from the Shanghai Tenth People's Hospital. The diabetic model of CRC cell lines in vitro and the mice model in vivo were developed to explore the BMP4 expression during CRC with or without diabetes. Further inhibition of BMP4 to observe its effects on CRC. Also, glucagon-like peptide-1 receptor agonist (GLP-1RA) was used to verify the underlying mechanism of hypoglycemic drugs on CRC via BMP4. RESULTS BMP4 expression was upregulated in CRC patients, and significantly higher in CRC patients with diabetes (P < 0.05). High glucose-induced insulin resistance (IR)-CRC cells and diabetic mice with metastasis model of CRC had increased BMP4 expression, activated BMP4-Smad1/5/8 pathway, and improved proliferative and metastatic ability mediated by epithelial-mesenchymal transition (EMT). And, treated CRC cells with exogenously BMP inhibitor-Noggin or transfected with lentivirus (sh-BMP4) could block the upregulated metastatic ability of CRC cells induced by IR. Meanwhile, GLP-1R was downregulated by high glucose-induced IR while unregulated by BMP4 inhibitor noggin, and treated GLP-1RA could suppress the proliferation of CRC cells induced by IR through downregulated BMP4. CONCLUSIONS BMP4 increased by high glucose promoted the EMT of CRC. The mechanism of the BMP4/Smad pathway was related to the susceptible metastasis of high glucose-induced IR-CRC. The commonly used hypoglycemic drug, GLP-1RA, inhibited the growth and promoted the apoptosis of CRC through the downregulation of BMP4. The result of our study suggested that BMP4 might serve as a therapeutic target in CRC patients with diabetes.
Collapse
Affiliation(s)
- Bingwei Ma
- Colorectal Cancer Central, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xingchun Wang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
- Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Hui Ren
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yingying Li
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Haijiao Zhang
- Department of Gastrointestinal Surgery, Huadong Hospital affiliated with Fudan University, 221 West Yanan Road, Shanghai, 200040, China
| | - Muqing Yang
- Department of General Surgery, Tenth People's Hospital of Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiyu Li
- Geriatric Cancer Center, Huadong Hospital Affiliated to Fudan University, 221 West Yanan Road, Shanghai, 200040, China.
| |
Collapse
|
6
|
Fetal Programming of the Endocrine Pancreas: Impact of a Maternal Low-Protein Diet on Gene Expression in the Perinatal Rat Pancreas. Int J Mol Sci 2022; 23:ijms231911057. [PMID: 36232358 PMCID: PMC9569808 DOI: 10.3390/ijms231911057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/26/2022] Open
Abstract
In rats, the time of birth is characterized by a transient rise in beta cell replication, as well as beta cell neogenesis and the functional maturation of the endocrine pancreas. However, the knowledge of the gene expression during this period of beta cell expansion is incomplete. The aim was to characterize the perinatal rat pancreas transcriptome and to identify regulatory pathways differentially regulated at the whole organ level in the offspring of mothers fed a regular control diet (CO) and of mothers fed a low-protein diet (LP). We performed mRNA expression profiling via the microarray analysis of total rat pancreas samples at embryonic day (E) 20 and postnatal days (P) 0 and 2. In the CO group, pancreas metabolic pathways related to sterol and lipid metabolism were highly enriched, whereas the LP diet induced changes in transcripts involved in RNA transcription and gene regulation, as well as cell migration and apoptosis. Moreover, a number of individual transcripts were markedly upregulated at P0 in the CO pancreas: growth arrest specific 6 (Gas6), legumain (Lgmn), Ets variant gene 5 (Etv5), alpha-fetoprotein (Afp), dual-specificity phosphatase 6 (Dusp6), and angiopoietin-like 4 (Angptl4). The LP diet induced the downregulation of a large number of transcripts, including neurogenin 3 (Neurog3), Etv5, Gas6, Dusp6, signaling transducer and activator of transcription 3 (Stat3), growth hormone receptor (Ghr), prolactin receptor (Prlr), and Gas6 receptor (AXL receptor tyrosine kinase; Axl), whereas upregulated transcripts were related to inflammatory responses and cell motility. We identified differentially regulated genes and transcriptional networks in the perinatal pancreas. These data revealed marked adaptations of exocrine and endocrine in the pancreas to the low-protein diet, and the data can contribute to identifying novel regulators of beta cell mass expansion and functional maturation and may provide a valuable tool in the generation of fully functional beta cells from stem cells to be used in replacement therapy.
Collapse
|
7
|
Sakhneny L, Mueller L, Schonblum A, Azaria S, Burganova G, Epshtein A, Isaacson A, Wilson H, Spagnoli FM, Landsman L. The postnatal pancreatic microenvironment guides β cell maturation through BMP4 production. Dev Cell 2021; 56:2703-2711.e5. [PMID: 34499867 DOI: 10.1016/j.devcel.2021.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 07/11/2021] [Accepted: 08/16/2021] [Indexed: 12/21/2022]
Abstract
Glucose homeostasis depends on regulated insulin secretion from pancreatic β cells, which acquire their mature phenotype postnatally. The functional maturation of β cells is regulated by a combination of cell-autonomous and exogenous factors; the identity of the latter is mostly unknown. Here, we identify BMP4 as a critical component through which the pancreatic microenvironment regulates β cell function. By combining transgenic mouse models and human iPSCs, we show that BMP4 promotes the expression of core β cell genes and is required for proper insulin production and secretion. We identified pericytes as the primary pancreatic source of BMP4, which start producing this ligand midway through the postnatal period, at the age β cells mature. Overall, our findings show that the islet niche directly promotes β cell functional maturation through the timely production of BMP4. Our study highlights the need to recapitulate the physiological postnatal islet niche for generating fully functional stem-cell-derived β cells for cell replacement therapy for diabetes.
Collapse
Affiliation(s)
- Lina Sakhneny
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Laura Mueller
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Anat Schonblum
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sivan Azaria
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Guzel Burganova
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Alona Epshtein
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Abigail Isaacson
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Heather Wilson
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Francesca M Spagnoli
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Limor Landsman
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
8
|
Butyrate Protects Pancreatic Beta Cells from Cytokine-Induced Dysfunction. Int J Mol Sci 2021; 22:ijms221910427. [PMID: 34638768 PMCID: PMC8508700 DOI: 10.3390/ijms221910427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic beta cell dysfunction caused by metabolic and inflammatory stress contributes to the development of type 2 diabetes (T2D). Butyrate, produced by the gut microbiota, has shown beneficial effects on glucose metabolism in animals and humans and may directly affect beta cell function, but the mechanisms are poorly described. The aim of this study was to investigate the effect of butyrate on cytokine-induced beta cell dysfunction in vitro. Mouse islets, rat INS-1E, and human EndoC-βH1 beta cells were exposed long-term to non-cytotoxic concentrations of cytokines and/or butyrate to resemble the slow onset of inflammation in T2D. Beta cell function was assessed by glucose-stimulated insulin secretion (GSIS), gene expression by qPCR and RNA-sequencing, and proliferation by incorporation of EdU into newly synthesized DNA. Butyrate protected beta cells from cytokine-induced impairment of GSIS and insulin content in the three beta cell models. Beta cell proliferation was reduced by both cytokines and butyrate. Expressions of the beta cell specific genes Ins, MafA, and Ucn3 reduced by the cytokine IL-1β were not affected by butyrate. In contrast, butyrate upregulated the expression of secretion/transport-related genes and downregulated inflammatory genes induced by IL-1β in mouse islets. In summary, butyrate prevents pro-inflammatory cytokine-induced beta cell dysfunction.
Collapse
|
9
|
Song Z, Chen L, Pang S, Yan B. Molecular genetic study on GATA5 gene promoter in acute myocardial infarction. PLoS One 2021; 16:e0248203. [PMID: 33684162 PMCID: PMC7939267 DOI: 10.1371/journal.pone.0248203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/23/2021] [Indexed: 11/23/2022] Open
Abstract
Background Acute myocardial infarction (AMI) is a severe type of coronary artery disease, caused by coronary occlusion and followed by cardiac ischaemia. GATA binding protein 5 (GATA5) is an important member of GATA family and plays an important role in vascular inflammation, endothelial function, oxidative stress and cell metabolism. Previous studies have shown that the DNA sequence variants (DSVs) in GATA4 and GATA6 promoter can increase susceptibility to AMI. In this study, we explored the relationship between GATA5 promoter and AMI for the first time, hoping to provide a new genetic basis for understanding the pathogenesis of AMI. Methods GATA5 promoter was sequenced in 683 individuals (332 AMI patients and 351 controls). The transcriptional activity of the GATA5 promoter with or without DSVs in HEK-293 cells, H9c2 cells and primary neonatal rat cardiomyocytes were examined by Promega Dual-Luciferase® Reporter Assay system. Electrophoretic mobility shift assay (EMSA) was performed to explore whether the DSVs interfered with the binding of transcription factors (TFs). Results Nine mutations have been found in GATA5 promoter, eight of them evidently altered the transcriptional activity of the GATA5 promoter, five of them disrupted the binding of TFs (such as farnesoid X receptor). Furthermore, haplotype AT (across rs80197101 and rs77067995) is a dangerous haplotype of AMI. Genotype GA and allele A of rs80197101 and genotype CT and allele T of rs77067995 are the risk factors of AMI. Conclusions DSVs in GATA5 promoter can increase susceptibility to AMI. But the mechanism remains to be verified in vivo.
Collapse
Affiliation(s)
- Zhipeng Song
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong, China
| | - Lu Chen
- Center for Molecular Medicine, Yanzhou People’s Hospital, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Bo Yan
- Center for Molecular Medicine, Yanzhou People’s Hospital, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- * E-mail:
| |
Collapse
|
10
|
Baboota RK, Blüher M, Smith U. Emerging Role of Bone Morphogenetic Protein 4 in Metabolic Disorders. Diabetes 2021; 70:303-312. [PMID: 33472940 DOI: 10.2337/db20-0884] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022]
Abstract
Bone morphogenetic proteins (BMPs) are a group of signaling molecules that belong to the TGF-β superfamily. Initially discovered for their ability to induce bone formation, BMPs are known to play a diverse and critical array of biological roles. We here focus on recent evidence showing that BMP4 is an important regulator of white/beige adipogenic differentiation with important consequences for thermogenesis, energy homeostasis, and development of obesity in vivo. BMP4 is highly expressed in, and released by, human adipose tissue, and serum levels are increased in obesity. Recent studies have now shown BMP4 to play an important role not only for white/beige/brown adipocyte differentiation and thermogenesis but also in regulating systemic glucose homeostasis and insulin sensitivity. It also has important suppressive effects on hepatic glucose production and lipid metabolism. Cellular BMP4 signaling/action is regulated by both ambient cell/systemic levels and several endogenous and systemic BMP antagonists. Reduced BMP4 signaling/action can contribute to the development of obesity, insulin resistance, and associated metabolic disorders. In this article, we summarize the pleiotropic functions of BMP4 in the pathophysiology of these diseases and also consider the therapeutic implications of targeting BMP4 in the prevention/treatment of obesity and its associated complications.
Collapse
Affiliation(s)
- Ritesh K Baboota
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Yang J, Ueharu H, Mishina Y. Energy metabolism: A newly emerging target of BMP signaling in bone homeostasis. Bone 2020; 138:115467. [PMID: 32512164 PMCID: PMC7423769 DOI: 10.1016/j.bone.2020.115467] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
Abstract
Energy metabolism is the process of generating energy (i.e. ATP) from nutrients. This process is indispensable for cell homeostasis maintenance and responses to varying conditions. Cells require energy for growth and maintenance and have evolved to have multiple pathways to produce energy. Both genetic and functional studies have demonstrated that energy metabolism, such as glucose, fatty acid, and amino acid metabolism, plays important roles in the formation and function of bone cells including osteoblasts, osteocytes, and osteoclasts. Dysregulation of energy metabolism in bone cells consequently disturbs the balance between bone formation and bone resorption. Metabolic diseases have also been reported to affect bone homeostasis. Bone morphogenic protein (BMP) signaling plays critical roles in regulating the formation and function of bone cells, thus affecting bone development and homeostasis. Mutations of BMP signaling-related genes in mice have been reported to show abnormalities in energy metabolism in many tissues, including bone. In addition, BMP signaling correlates with critical signaling pathways such as mTOR, HIF, Wnt, and self-degradative process autophagy to coordinate energy metabolism and bone homeostasis. These findings will provide a newly emerging target of BMP signaling and potential therapeutic strategies and the improved management of bone diseases. This review summarizes the recent advances in our understanding of (1) energy metabolism in regulating the formation and function of bone cells, (2) function of BMP signaling in whole body energy metabolism, and (3) mechanistic interaction of BMP signaling with other signaling pathways and biological processes critical for energy metabolism and bone homeostasis.
Collapse
Affiliation(s)
- Jingwen Yang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
Triñanes J, Ten Dijke P, Groen N, Hanegraaf M, Porrini E, Rodriguez-Rodriguez AE, Drachenberg C, Rabelink TJ, de Koning E, Carlotti F, de Vries APJ. Tacrolimus-Induced BMP/SMAD Signaling Associates With Metabolic Stress-Activated FOXO1 to Trigger β-Cell Failure. Diabetes 2020; 69:193-204. [PMID: 31732500 DOI: 10.2337/db19-0828] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/06/2019] [Indexed: 11/13/2022]
Abstract
Active maintenance of β-cell identity through fine-tuned regulation of key transcription factors ensures β-cell function. Tacrolimus, a widely used immunosuppressant, accelerates onset of diabetes after organ transplantation, but underlying molecular mechanisms are unclear. Here we show that tacrolimus induces loss of human β-cell maturity and β-cell failure through activation of the BMP/SMAD signaling pathway when administered under mild metabolic stress conditions. Tacrolimus-induced phosphorylated SMAD1/5 acts in synergy with metabolic stress-activated FOXO1 through formation of a complex. This interaction is associated with reduced expression of the key β-cell transcription factor MAFA and abolished insulin secretion, both in vitro in primary human islets and in vivo in human islets transplanted into high-fat diet-fed mice. Pharmacological inhibition of BMP signaling protects human β-cells from tacrolimus-induced β-cell dysfunction in vitro. Furthermore, we confirm that BMP/SMAD signaling is activated in protocol pancreas allograft biopsies from recipients on tacrolimus. To conclude, we propose a novel mechanism underlying the diabetogenicity of tacrolimus in primary human β-cells. This insight could lead to new treatment strategies for new-onset diabetes and may have implications for other forms of diabetes.
Collapse
Affiliation(s)
- Javier Triñanes
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Nathalie Groen
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Maaike Hanegraaf
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Esteban Porrini
- Instituto de Tecnología Biomédicas and Hospital Universitario de Canarias, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Ana E Rodriguez-Rodriguez
- Instituto de Tecnología Biomédicas and Hospital Universitario de Canarias, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Cinthia Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Ton J Rabelink
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Eelco de Koning
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Françoise Carlotti
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Aiko P J de Vries
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
13
|
Ibarra Urizar A, Prause M, Wortham M, Sui Y, Thams P, Sander M, Christensen GL, Billestrup N. Beta-cell dysfunction induced by non-cytotoxic concentrations of Interleukin-1β is associated with changes in expression of beta-cell maturity genes and associated histone modifications. Mol Cell Endocrinol 2019; 496:110524. [PMID: 31362031 DOI: 10.1016/j.mce.2019.110524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/12/2019] [Accepted: 07/24/2019] [Indexed: 10/26/2022]
Abstract
Decreased insulin secretory capacity in Type 2 diabetes mellitus is associated with beta-cell dedifferentiation and inflammation. We hypothesize that prolonged exposure of beta-cells to low concentrations of IL-1β induce beta-cell dedifferentiation characterized by impaired glucose-stimulated insulin secretion, reduced expression of key beta-cell genes and changes in histone modifications at gene loci known to affect beta-cell function. Ten days exposure to IL-1β at non-cytotoxic concentrations reduced insulin secretion and beta-cell proliferation and decreased expression of key beta-cell identity genes, including MafA and Ucn3 and decreased H3K27ac at the gene loci, suggesting that inflammatory cytokines directly affects the epigenome. Following removal of IL-1β, beta-cell function was normalized and mRNA expression of beta-cell identity genes, such as insulin and Ucn3 returned to pre-stimulation levels. Our findings indicate that prolonged exposure to low concentrations of IL-1β induces epigenetic changes associated with loss of beta-cell identity as observed in Type 2 diabetes.
Collapse
Affiliation(s)
- Adriana Ibarra Urizar
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Michala Prause
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Matthew Wortham
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yinghui Sui
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Peter Thams
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gitte Lund Christensen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark; Department of Biomedical Laboratory Science, Metropolitan University College, Copenhagen, 2200, Denmark
| | - Nils Billestrup
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark; Lead Contact Nils Billestrup, Department of Biomedical Science, University of Copenhagen, Copenhagen, 2200, Denmark.
| |
Collapse
|
14
|
Meng Y, Cui Y, Zhang W, Fu S, Huang L, Dong H, Du H. Integrative Analysis of Genome and Expression Profile Data Reveals the Genetic Mechanism of the Diabetic Pathogenesis in Goto Kakizaki (GK) Rats. Front Genet 2019; 9:724. [PMID: 30687391 PMCID: PMC6335273 DOI: 10.3389/fgene.2018.00724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/22/2018] [Indexed: 12/18/2022] Open
Abstract
The Goto Kakizaki (GK) rats which can spontaneously develop type 2 diabetes (T2D), are generated by repeated inbreeding of Wistar rats with glucose intolerance. The glucose intolerance in GK rat is mainly attributed to the impairment in glucose-stimulated insulin secretion (GSIS). In addition, GK rat display a decrease in beta cell mass, and a change in insulin action. However, the genetic mechanism of these features remain unclear. In the present study, we analyzed the population variants of GK rats and control Wistar rats by whole genome sequencing and identified 1,839 and 1,333 specific amino acid changed (SAAC) genes in GK and Wistar rats, respectively. We also detected the putative artificial selective sweeps (PASS) regions in GK rat which were enriched with GK fixed variants and were under selected in the initial diabetic-driven derivation by homogeneity test with the fixed and polymorphic sites between GK and Wistar populations. Finally, we integrated the SAAC genes, PASS region genes and differentially expressed genes in GK pancreatic beta cells to reveal the genetic mechanism of the impairment in GSIS, a decrease in beta cell mass, and a change in insulin action in GK rat. The results showed that Slc2a2 gene was related to impaired glucose transport and Adcy3, Cacna1f, Bmp4, Fam3b, and Ptprn2 genes were related to Ca2+ channel dysfunction which may responsible for the impaired GSIS. The genes Hnf4g, Bmp4, and Bad were associated with beta cell development and may be responsible for a decrease in beta cell mass while genes Ide, Ppp1r3c, Hdac9, Ghsr, and Gckr may be responsible for the change in insulin action in GK rats. The overexpression or inhibition of Bmp4, Fam3b, Ptprn2, Ide, Hnf4g, and Bad has been reported to change the glucose tolerance in rodents. However, the genes Bmp4, Fam3b, and Ptprn2 were found to be associated with diabetes in GK rats for the first time in the present study. Our findings provide a comprehensive genetic map of the abnormalities in GK genome which will be helpful in understand the underlying genetic mechanism of pathogenesis of diabetes in GK rats.
Collapse
Affiliation(s)
- Yuhuan Meng
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Ying Cui
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Wenlu Zhang
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Shuying Fu
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Lizhen Huang
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Hua Dong
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering - Department of Biomedical Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
15
|
Polysiphonia japonica Extract Attenuates Palmitate-Induced Toxicity and Enhances Insulin Secretion in Pancreatic Beta-Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4973851. [PMID: 30510621 PMCID: PMC6230388 DOI: 10.1155/2018/4973851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 01/08/2023]
Abstract
Beta-cell loss is a major cause of the pathogenesis of diabetes. Elevated levels of free fatty acids may contribute to the loss of β-cells. Using a transgenic zebrafish, we screened ~50 seaweed crude extracts to identify materials that protect β-cells from free fatty acid damage. We found that an extract of the red seaweed Polysiphonia japonica (PJE) had a β-cell protective effect. We examined the protective effect of PJE on palmitate-induced damage in β-cells. PJE was found to preserve cell viability and glucose-induced insulin secretion in a pancreatic β-cell line, Ins-1, treated with palmitate. Additionally, PJE prevented palmitate-induced insulin secretion dysfunction in zebrafish embryos and mouse primary islets and improved insulin secretion in β-cells against palmitate treatment. These findings suggest that PJE protects pancreatic β-cells from palmitate-induced damage. PJE may be a potential therapeutic functional food for diabetes.
Collapse
|
16
|
Jiang WJ, Peng YC, Yang KM. Cellular signaling pathways regulating β-cell proliferation as a promising therapeutic target in the treatment of diabetes. Exp Ther Med 2018; 16:3275-3285. [PMID: 30233674 PMCID: PMC6143874 DOI: 10.3892/etm.2018.6603] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 07/27/2018] [Indexed: 12/30/2022] Open
Abstract
It is established that a decrease in β-cell number and deficiency in the function of existing β-cells contribute to type 1 and type 2 diabetes mellitus. Therefore, a major focus of current research is to identify novel methods of improving the number and function of β-cells, so as to prevent and/or postpone the development of diabetes mellitus and potentially reverse diabetes mellitus. Based on prior knowledge of the above-mentioned causes, promising therapeutic approaches may include direct transplantation of islets, implantation and subsequent induced differentiation of progenitors/stem cells to β-cells, replication of pre-existing β-cells, or activation of endogenous β-cell progenitors. More recently, with regards to cell replacement and regenerative treatment for diabetes patients, the identification of cellular signaling pathways with related genes or corresponding proteins involved in diabetes has become a topic of interest. However, the majority of pathways and molecules associated with β-cells remain unresolved, and the specialized functions of known pathways remain unclear, particularly in humans. The current article has evaluated the progress of research on pivotal cellular signaling pathways involved with β-cell proliferation and survival, and their validity for therapeutic adult β-cell regeneration in diabetes. More efforts are required to elucidate the cellular events involved in human β-cell proliferation in terms of the underlying mechanisms and functions.
Collapse
Affiliation(s)
- Wen-Juan Jiang
- Institute of Anatomy, Basic Medical College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Yun-Chuan Peng
- Institute of Anatomy, Basic Medical College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Kai-Ming Yang
- Institute of Anatomy, Basic Medical College of Dali University, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
17
|
Luo B, Chen K, Feng Q, Xiao W, Ma D, Yang H, Zhang C. The interplay of BMP4 and IL‑7 regulates the apoptosis of intestinal intraepithelial lymphocytes under conditions of ischemia̸reperfusion. Int J Mol Med 2018; 41:2640-2650. [PMID: 29436597 PMCID: PMC5846653 DOI: 10.3892/ijmm.2018.3480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/11/2018] [Indexed: 12/22/2022] Open
Abstract
The number and function of intestinal intraepithelial lymphocytes (IELs) have been found to be significantly reduced following induction of acute intestinal mucosal damage via intestinal ischemia̸reperfusion (I̸R). However, the mechanism underlying this reduction remains unclear. Therefore, it was hypothesized that the interplay of bone morphogenetic protein (BMP)4 and interleukin (IL)‑7 regulates IEL function and number. Recent studies have demonstrated that the different components of the BMP2̸4 signaling pathway are expressed in intestinal epithelial cells (IECs) via the activation of nuclear factor (NF)‑κB. In the present study, reverse transcription‑polymerase chain reaction analysis and flow cytometry demonstrated that IELs express BMP receptors (BMPRIA, BMPRIB, ActRIA and BMPRII) and non‑canonical signal transduction molecules (NF‑κB). an in vivo mouse intestinal I̸R model was used, and I̸R was shown to increase the expression of BMP4 in IECs and upregulate the expression levels of BMPRIA, BMPRIB and phosphorylated NF‑κB in IELs. Following isolation and culture of IELs, it was observed that exogenous BMP4 also upregulated the expression of BMPRIA and BMPRIB and activated NF‑κB signaling in IELs, inducing IEL apoptosis. In addition, the rate of apoptosis of IELs decreased following treatment with the BMP‑specific antagonist Noggin or with the NF‑κB inhibitor pyrrolidine dithiocarbamate. Furthermore, it was observed that exogenous IL‑7 can decrease BMP4 protein expression in IECs and the expression of phosphorylated NF‑κB protein in IELs. The findings of the present study suggest that, under conditions of I̸R, IEC‑derived BMP4 activates NF‑κB signaling in IELs, inducing IEL apoptosis, further aggravating the dysfunction of the intestinal mucosal barrier. However, these effects may be alleviated by IL‑7 treatment. Therefore, BMP4 and IL‑7 appear to be involved in the interaction between IECs and IELs and in the mechanism underlying intestinal mucosal barrier dysfunction.
Collapse
Affiliation(s)
- Binyu Luo
- Department of General Surgery Two, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical University, Nanchong, Sichuan 637000, P.R. China
| | - Kang Chen
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Qi Feng
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Dan Ma
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Chaojun Zhang
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
18
|
Herrera B, Addante A, Sánchez A. BMP Signalling at the Crossroad of Liver Fibrosis and Regeneration. Int J Mol Sci 2017; 19:ijms19010039. [PMID: 29295498 PMCID: PMC5795989 DOI: 10.3390/ijms19010039] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Bone Morphogenetic Proteins (BMPs) belong to the Transforming Growth Factor-β (TGF-β) family. Initially identified due to their ability to induce bone formation, they are now known to have multiple functions in a variety of tissues, being critical not only during development for tissue morphogenesis and organogenesis but also during adult tissue homeostasis. This review focus on the liver as a target tissue for BMPs actions, devoting most efforts to summarize our knowledge on their recently recognized and/or emerging roles on regulation of the liver regenerative response to various insults, either acute or chronic and their effects on development and progression of liver fibrosis in different pathological conditions. In an attempt to provide the basis for guiding research efforts in this field both the more solid and more controversial areas of research were highlighted.
Collapse
Affiliation(s)
- Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
19
|
Calpe S, Correia ACP, Sancho-Serra MDC, Krishnadath KK. Comparison of newly developed anti-bone morphogenetic protein 4 llama-derived antibodies with commercially available BMP4 inhibitors. MAbs 2017; 8:678-88. [PMID: 26967714 PMCID: PMC4966848 DOI: 10.1080/19420862.2016.1158380] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Due to improved understanding of the role of bone morphogenetic protein 4 (BMP4) in an increasing number of diseases, the development of selective inhibitors of BMP4 is an attractive therapeutic option. The currently available BMP4 inhibitors are not suitable as therapeutics because of their low specificity and low effectiveness. Here, we compared newly generated anti-BMP4 llama-derived antibodies (VHHs) with 3 different types of commercially available BMP4 inhibitors, natural antagonists, small molecule BMPR inhibitors and conventional anti-BMP4 monoclonal antibodies. We found that the anti-BMP4 VHHs were as effective as the natural antagonist or small molecule inhibitors, but had higher specificity. We also showed that commercial anti-BMP4 antibodies were inferior in terms of both specificity and effectiveness. These findings might result from the fact that the VHHs C4C4 and C8C8 target a small region within the BMPR1 epitope of BMP4, whereas the commercial antibodies target other areas of the BMP4 molecule. Our results show that the newly developed anti-BMP4 VHHs are promising antibodies with better specificity and effectivity for inhibition of BMP4, making them an attractive tool for research and for therapeutic applications.
Collapse
Affiliation(s)
- Silvia Calpe
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands
| | - Ana C P Correia
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands
| | - Maria Del Carmen Sancho-Serra
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands
| | - Kausilia K Krishnadath
- a Center for Experimental & Molecular Medicine , Academic Medical Center , Meibergdreef , Amsterdam , The Netherlands.,b Department of Gastroenterology & Hepatology , Academic Medical Center , Meibergdreef, Amsterdam , The Netherlands
| |
Collapse
|
20
|
Bonnet N. Bone-Derived Factors: A New Gateway to Regulate Glycemia. Calcif Tissue Int 2017; 100:174-183. [PMID: 27832316 DOI: 10.1007/s00223-016-0210-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 11/02/2016] [Indexed: 01/26/2023]
Abstract
Type 2 diabetes mellitus (T2DM) and osteoporosis are two major disorders which prevalence increases with aging and is predicted to worsen in the coming years. Preclinical investigations suggest common mechanisms implicated in the pathogenesis of both disorders. Recent evidence has established that there is a clear link between glucose and bone metabolism. The emergence of bone as an endocrine regulator through FGF23 and osteocalcin has led to the re-evaluation of the role of bone cells and bone-derived factors in the development of metabolic diseases such as T2DM. The development of bone morphogenetic proteins, fibroblast growth factor 23, and osteoprotegerin-deficient mice has allowed to elucidate their role in bone homeostasis, as well as revealed their potential important function in glucose homeostasis. This review proposes emerging perspectives for several bone-derived factors that may regulate glycemia through the activation or inhibition of bone remodeling or directly by regulating function of key organs such as pancreatic beta cell proliferation, insulin expression and secretion, storage and release of glucose from the liver, skeletal muscle contraction, and browning of the adipose tissue. Connections between organs including bone-derived factors should further be explored to understand the pathophysiology of glucose metabolism and diabetes.
Collapse
Affiliation(s)
- Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospitals and Faculty of Medicine, 64 Av de la Roseraie, 1205, Geneva 14, Switzerland.
| |
Collapse
|
21
|
Schmidt SF, Madsen JGS, Frafjord KØ, Poulsen LLC, Salö S, Boergesen M, Loft A, Larsen BD, Madsen MS, Holst JJ, Maechler P, Dalgaard LT, Mandrup S. Integrative Genomics Outlines a Biphasic Glucose Response and a ChREBP-RORγ Axis Regulating Proliferation in β Cells. Cell Rep 2016; 16:2359-72. [PMID: 27545881 DOI: 10.1016/j.celrep.2016.07.063] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/26/2016] [Accepted: 07/25/2016] [Indexed: 12/27/2022] Open
Abstract
Glucose is an important inducer of insulin secretion, but it also stimulates long-term adaptive changes in gene expression that can either promote or antagonize the proliferative potential and function of β cells. Here, we have generated time-resolved profiles of enhancer and transcriptional activity in response to glucose in the INS-1E pancreatic β cell line. Our data outline a biphasic response with a first transcriptional wave during which metabolic genes are activated, and a second wave where cell-cycle genes are activated and β cell identity genes are repressed. The glucose-sensing transcription factor ChREBP directly activates first wave enhancers, whereas repression and activation of second wave enhancers are indirect. By integrating motif enrichment within late-regulated enhancers with expression profiles of the associated transcription factors, we have identified multiple putative regulators of the second wave. These include RORγ, the activity of which is important for glucose-induced proliferation of both INS-1E and primary rat β cells.
Collapse
Affiliation(s)
- Søren Fisker Schmidt
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jesper Grud Skat Madsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; NNF Center of Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Kari Østerli Frafjord
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Lars la Cour Poulsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Sofia Salö
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Michael Boergesen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Anne Loft
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Bjørk Ditlev Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Maria Stahl Madsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jens Juul Holst
- NNF Center of Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva, Switzerland
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark.
| |
Collapse
|
22
|
Ibarra Urizar A, Friberg J, Christensen DP, Lund Christensen G, Billestrup N. Inflammatory Cytokines Stimulate Bone Morphogenetic Protein-2 Expression and Release from Pancreatic Beta Cells. J Interferon Cytokine Res 2016; 36:20-9. [DOI: 10.1089/jir.2014.0199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Adriana Ibarra Urizar
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Josefine Friberg
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Dan Ploug Christensen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Gitte Lund Christensen
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Nils Billestrup
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
23
|
Grgurevic L, Christensen GL, Schulz TJ, Vukicevic S. Bone morphogenetic proteins in inflammation, glucose homeostasis and adipose tissue energy metabolism. Cytokine Growth Factor Rev 2015; 27:105-18. [PMID: 26762842 DOI: 10.1016/j.cytogfr.2015.12.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/10/2015] [Accepted: 12/23/2015] [Indexed: 12/13/2022]
Abstract
Bore morphogenetic proteins (BMPs) are members of the transforming growth factor (TGF)-β superfamily, a group of secreted proteins that regulate embryonic development. This review summarizes the effects of BMPs on physiological processes not exclusively linked to the musculoskeletal system. Specifically, we focus on the involvement of BMPs in inflammatory disorders, e.g. fibrosis, inflammatory bowel disease, anchylosing spondylitis, rheumatoid arthritis. Moreover, we discuss the role of BMPs in the context of vascular disorders, and explore the role of these signalling proteins in iron homeostasis (anaemia, hemochromatosis) and oxidative damage. The second and third parts of this review focus on BMPs in the development of metabolic pathologies such as type-2 diabetes mellitus and obesity. The pancreatic beta cells are the sole source of the hormone insulin and BMPs have recently been implicated in pancreas development as well as control of adult glucose homeostasis. Lastly, we review the recently recognized role of BMPs in brown adipose tissue formation and their consequences for energy expenditure and adiposity. In summary, BMPs play a pivotal role in metabolism beyond their role in skeletal homeostasis. However, increased understanding of these pleiotropic functions also highlights the necessity of tissue-specific strategies when harnessing BMP action as a therapeutic target.
Collapse
Affiliation(s)
- Lovorka Grgurevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Zagreb, Croatia
| | | | - Tim J Schulz
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| | - Slobodan Vukicevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Zagreb, Croatia.
| |
Collapse
|
24
|
Shuai B, Shen L, Zhu R, Zhou P. Effect of Qing'e formula on the in vitro differentiation of bone marrow-derived mesenchymal stem cells from proximal femurs of postmenopausal osteoporotic mice. Altern Ther Health Med 2015. [PMID: 26205885 PMCID: PMC4513391 DOI: 10.1186/s12906-015-0777-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Qing'e formula (QEF), prepared from an ancient Chinese recipe, was previously suggested to regulate bone metabolism and improve bone mineral density in patients with osteoporosis. To study the effects of medicated serum containing QEF on the in vitro differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) isolated from the proximal femurs of postmenopausal osteoporosis (PMOP) mice. METHODS Using an established mouse model of PMOP, mononuclear cells were isolated from the bone marrow present in the proximal femurs and cultured. PMOP mice were also randomly divided into four groups: the untreated group (Group A) and the groups treated with respectively low (Group B), medium (Group C), and high (Group D) concentrations of QEF. Serum was isolated from each and used to treat the cultured BMSCs in conjunction with recombinant human bone morphogenetic protein-2 (rhBMP-2). Cell morphology, proliferation rates, intracellular alkaline phosphatase (ALP) activity, and transforming growth factor-beta 1 (TGF-β1) mRNA expression were evaluated. RESULTS QEF-treated serum, particularly that containing moderate and high concentrations, appears to enhance the rhBMP-2-mediated changes in cell morphology, proliferation, and differentiation (determined via the expression of TGF-β1 mRNA and ALP activity) observed in the BMSCs isolated from PMOP mice. CONCLUSIONS QEF may play a role in the prevention and treatment of PMOP by enhancing the activity of rhBMP-2.
Collapse
|
25
|
Stewart AF, Hussain MA, García-Ocaña A, Vasavada RC, Bhushan A, Bernal-Mizrachi E, Kulkarni RN. Human β-cell proliferation and intracellular signaling: part 3. Diabetes 2015; 64:1872-85. [PMID: 25999530 PMCID: PMC4439562 DOI: 10.2337/db14-1843] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This is the third in a series of Perspectives on intracellular signaling pathways coupled to proliferation in pancreatic β-cells. We contrast the large knowledge base in rodent β-cells with the more limited human database. With the increasing incidence of type 1 diabetes and the recognition that type 2 diabetes is also due in part to a deficiency of functioning β-cells, there is great urgency to identify therapeutic approaches to expand human β-cell numbers. Therapeutic approaches might include stem cell differentiation, transdifferentiation, or expansion of cadaver islets or residual endogenous β-cells. In these Perspectives, we focus on β-cell proliferation. Past Perspectives reviewed fundamental cell cycle regulation and its upstream regulation by insulin/IGF signaling via phosphatidylinositol-3 kinase/mammalian target of rapamycin signaling, glucose, glycogen synthase kinase-3 and liver kinase B1, protein kinase Cζ, calcium-calcineurin-nuclear factor of activated T cells, epidermal growth factor/platelet-derived growth factor family members, Wnt/β-catenin, leptin, and estrogen and progesterone. Here, we emphasize Janus kinase/signal transducers and activators of transcription, Ras/Raf/extracellular signal-related kinase, cadherins and integrins, G-protein-coupled receptors, and transforming growth factor β signaling. We hope these three Perspectives will serve to introduce these pathways to new researchers and will encourage additional investigators to focus on understanding how to harness key intracellular signaling pathways for therapeutic human β-cell regeneration for diabetes.
Collapse
Affiliation(s)
- Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehboob A Hussain
- Departments of Medicine and Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Ernesto Bernal-Mizrachi
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, MI, and VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
Christensen GL, Jacobsen MLB, Wendt A, Mollet IG, Friberg J, Frederiksen KS, Meyer M, Bruun C, Eliasson L, Billestrup N. Bone morphogenetic protein 4 inhibits insulin secretion from rodent beta cells through regulation of calbindin1 expression and reduced voltage-dependent calcium currents. Diabetologia 2015; 58:1282-90. [PMID: 25828920 DOI: 10.1007/s00125-015-3568-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 03/04/2015] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is characterised by progressive loss of pancreatic beta cell mass and function. Therefore, it is of therapeutic interest to identify factors with the potential to improve beta cell proliferation and insulin secretion. Bone morphogenetic protein 4 (BMP4) expression is increased in diabetic animals and BMP4 reduces glucose-stimulated insulin secretion (GSIS). Here, we investigate the molecular mechanism behind this inhibition. METHODS BMP4-mediated inhibition of GSIS was investigated in detail using single cell electrophysiological measurements and live cell Ca(2+) imaging. BMP4-mediated gene expression changes were investigated by microarray profiling, quantitative PCR and western blotting. RESULTS Prolonged exposure to BMP4 reduced GSIS from rodent pancreatic islets. This inhibition was associated with decreased exocytosis due to a reduced Ca(2+) current through voltage-dependent Ca(2+) channels. To identify proteins involved in the inhibition of GSIS, we investigated global gene expression changes induced by BMP4 in neonatal rat pancreatic islets. Expression of the Ca(2+)-binding protein calbindin1 was significantly induced by BMP4. Overexpression of calbindin1 in primary islet cells reduced GSIS, and the effect of BMP4 on GSIS was lost in islets from calbindin1 (Calb1) knockout mice. CONCLUSIONS/INTERPRETATION We found BMP4 treatment to markedly inhibit GSIS from rodent pancreatic islets in a calbindin1-dependent manner. Calbindin1 is suggested to mediate the effect of BMP4 by buffering Ca(2+) and decreasing Ca(2+) channel activity, resulting in diminished insulin exocytosis. Both BMP4 and calbindin1 are potential pharmacological targets for the treatment of beta cell dysfunction.
Collapse
Affiliation(s)
- Gitte L Christensen
- Department of Biomedical Sciences, University of Copenhagen, Nørre Alle 20, 2100, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|