1
|
de Ramos FC, Barth R, Santos MR, Almeida MDS, Ferreira SM, Rafacho A, Boschero AC, dos Santos GJ. Hepatocyte nuclear factor 4-α is necessary for high fat diet-induced pancreatic β-cell mass expansion and metabolic compensations. Front Endocrinol (Lausanne) 2024; 15:1511813. [PMID: 39741884 PMCID: PMC11685084 DOI: 10.3389/fendo.2024.1511813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Aims This study investigates the role of Hepatocyte Nuclear Factor 4α (HNF4α) in the adaptation of pancreatic β-cells to an HFD-induced obesogenic environment, focusing on β cell mass expansion and metabolic adaptations. Main methods We utilized an HNF4α knockout (KO) mouse model, with CRE-recombinase enzyme activation confirmed through tamoxifen administration. KO and Control (CTL) mice were fed an HFD for 20 weeks. We monitored body weight, food intake, glucose tolerance, insulin sensitivity, and insulinemia. Also, to assess structural and metabolic changes, histological analyses of pancreatic islets and liver tissue were conducted. Key findings KO mice displayed lower fasting blood glucose levels compared to CTL mice after tamoxifen administration, indicating impaired glucose-regulated insulin secretion. HFD-fed KO mice consumed less food but exhibited greater weight gain and perigonadal fat accumulation, reflecting higher energy efficiency. Histological analysis revealed more pronounced liver steatosis and fibrosis in KO mice on HFD. Glucose intolerance and insulin resistance were exacerbated in KO mice, highlighting their inability to adapt to increased metabolic demand. Structural analysis showed that KO mice failed to exhibit HFD-induced β cell mass expansion, resulting in reduced islet diameter and number, confirming the critical role of HNF4α in β cell adaptation. Significance This study demonstrates that HNF4α is essential for the proper metabolic and structural adaptation of pancreatic β-cells in response to an obesogenic environment. The lack of HNF4α impairs β cell functionality, leading to increased susceptibility to glucose intolerance and insulin resistance. These findings underscore the importance of HNF4α in maintaining glucose homeostasis and highlight its potential as a therapeutic target for diabetes management in obesity.
Collapse
Affiliation(s)
- Francieli Caroline de Ramos
- Islet Biology and Metabolism Lab – IBM Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Robson Barth
- Islet Biology and Metabolism Lab – IBM Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Marcos Rizzon Santos
- Islet Biology and Metabolism Lab – IBM Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Milena dos Santos Almeida
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Sandra Mara Ferreira
- Laboratory of Endocrine Pancreas And Metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, Brazil
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Antônio Carlos Boschero
- Laboratory of Endocrine Pancreas And Metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, Brazil
| | - Gustavo Jorge dos Santos
- Islet Biology and Metabolism Lab – IBM Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
- Laboratory of Endocrine Pancreas And Metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, Brazil
| |
Collapse
|
2
|
Chu Y, Zhao L, Liu X, Chen H, Zhao C, Chen S, Xiang S, Lu J, Wang X, Wan Y, Dong D, Yao S, Li C, Yin R, Ren G, Yang X, Yu M. Lysine 117 Residue Is Essential for the Function of the Hepatocyte Nuclear Factor 1α. Diabetes 2023; 72:1502-1516. [PMID: 37440709 DOI: 10.2337/db22-0672] [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] [Received: 08/07/2022] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
Hepatocyte nuclear factor 1α (HNF1α) plays essential roles in controlling development and metabolism; its mutations are clearly linked to the occurrence of maturity-onset diabetes of the young (MODY3) in humans. Lysine 117 (K117) to glutamic acid (E117) mutation in the HNF1α gene has been clinically associated with MODY3, but no functional data on this variant are available. Here, we addressed the role of lysine 117 in HNF1α function using a knock-in animal model and site-directed mutagenesis. HNF1α K117E homozygous mice exhibited dwarfism, hepatic dysfunction, renal Fanconi syndrome, and progressive wasting syndrome. These phenotypes were very similar to those of mice with complete HNF1α deficiency, suggesting that K117 is critical to HNF1α functions. K117E homozygotes developed diabetes in the early postnatal period. The relative deficiency of serum insulin levels and the normal response to insulin treatment in homozygous mice were markedly similar to those in the MODY3 disorder in humans. Moreover, K117E heterozygous mutant causes age-dependent glucose intolerance, which is similar to the pathogenesis of MODY3 as well. K117 mutants significantly reduced the overall transactivation and DNA binding capacity of HNF1α by disrupting dimerization. Collectively, our findings reveal a previously unappreciated role of POU domain of HNF1α in homodimerization and provide important clues for identifying the molecular basis of HNF1α-related diseases such as MODY3. ARTICLE HIGHLIGHTS HNF1α K117E homozygous mice exhibited dwarfism, hepatic dysfunction, renal Fanconi syndrome, and progressive wasting syndrome. K117E homozygotes developed diabetes in the early postnatal period. K117E heterozygous mutant causes age-dependent glucose intolerance, which is similar to the pathogenesis of maturity-onset diabetes of the young. K117 mutants significantly reduced the overall transactivation and DNA binding capacity of HNF1α by disrupting dimerization.
Collapse
Affiliation(s)
- Yuan Chu
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Long Zhao
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xian Liu
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chen Zhao
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- Department of Hygienic Toxicology and Environmental Hygiene, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Sicong Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Shensi Xiang
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jun Lu
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Xiaofang Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Institute of Life Sciences, He Bei University, Baoding, China
| | - Yue Wan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- School of Basic Medical Sciences, An Hui Medical University, Hefei, China
| | - Diandian Dong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Institute of Life Sciences, He Bei University, Baoding, China
| | - Songhui Yao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Changyan Li
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- School of Basic Medical Sciences, An Hui Medical University, Hefei, China
| | - Ronghua Yin
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Guangming Ren
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xiaoming Yang
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Institute of Life Sciences, He Bei University, Baoding, China
- School of Basic Medical Sciences, An Hui Medical University, Hefei, China
| |
Collapse
|
3
|
Weng C, Gu A, Zhang S, Lu L, Ke L, Gao P, Liu X, Wang Y, Hu P, Plummer D, MacDonald E, Zhang S, Xi J, Lai S, Leskov K, Yuan K, Jin F, Li Y. Single cell multiomic analysis reveals diabetes-associated β-cell heterogeneity driven by HNF1A. Nat Commun 2023; 14:5400. [PMID: 37669939 PMCID: PMC10480445 DOI: 10.1038/s41467-023-41228-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
Broad heterogeneity in pancreatic β-cell function and morphology has been widely reported. However, determining which components of this cellular heterogeneity serve a diabetes-relevant function remains challenging. Here, we integrate single-cell transcriptome, single-nuclei chromatin accessibility, and cell-type specific 3D genome profiles from human islets and identify Type II Diabetes (T2D)-associated β-cell heterogeneity at both transcriptomic and epigenomic levels. We develop a computational method to explicitly dissect the intra-donor and inter-donor heterogeneity between single β-cells, which reflect distinct mechanisms of T2D pathogenesis. Integrative transcriptomic and epigenomic analysis identifies HNF1A as a principal driver of intra-donor heterogeneity between β-cells from the same donors; HNF1A expression is also reduced in β-cells from T2D donors. Interestingly, HNF1A activity in single β-cells is significantly associated with lower Na+ currents and we nominate a HNF1A target, FXYD2, as the primary mitigator. Our study demonstrates the value of investigating disease-associated single-cell heterogeneity and provides new insights into the pathogenesis of T2D.
Collapse
Affiliation(s)
- Chen Weng
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Biomedical Sciences Training Program (BSTP), School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Anniya Gu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Medical Scientist Training Program (MSTP), School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Shanshan Zhang
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Biomedical Sciences Training Program (BSTP), School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Leina Lu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Luxin Ke
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Biomedical Sciences Training Program (BSTP), School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peidong Gao
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Xiaoxiao Liu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yuntong Wang
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peinan Hu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Biomedical Sciences Training Program (BSTP), School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Dylan Plummer
- Department of Computer and Data Sciences, School of Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Elise MacDonald
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Saixian Zhang
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jiajia Xi
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sisi Lai
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- The Biomedical Sciences Training Program (BSTP), School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Konstantin Leskov
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Kyle Yuan
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Fulai Jin
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Computer and Data Sciences, School of Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Yan Li
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
4
|
Hermann FM, Kjærgaard MF, Tian C, Tiemann U, Jackson A, Olsen LR, Kraft M, Carlsson PO, Elfving IM, Kettunen JLT, Tuomi T, Novak I, Semb H. An insulin hypersecretion phenotype precedes pancreatic β cell failure in MODY3 patient-specific cells. Cell Stem Cell 2023; 30:38-51.e8. [PMID: 36563694 DOI: 10.1016/j.stem.2022.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/04/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022]
Abstract
MODY3 is a monogenic hereditary form of diabetes caused by mutations in the transcription factor HNF1A. The patients progressively develop hyperglycemia due to perturbed insulin secretion, but the pathogenesis is unknown. Using patient-specific hiPSCs, we recapitulate the insulin secretion sensitivity to the membrane depolarizing agent sulfonylurea commonly observed in MODY3 patients. Unexpectedly, MODY3 patient-specific HNF1A+/R272C β cells hypersecrete insulin both in vitro and in vivo after transplantation into mice. Consistently, we identified a trend of increased birth weight in human HNF1A mutation carriers compared with healthy siblings. Reduced expression of potassium channels, specifically the KATP channel, in MODY3 β cells, increased calcium signaling, and rescue of the insulin hypersecretion phenotype by pharmacological targeting ATP-sensitive potassium channels or low-voltage-activated calcium channels suggest that more efficient membrane depolarization underlies the hypersecretion of insulin in MODY3 β cells. Our findings identify a pathogenic mechanism leading to β cell failure in MODY3.
Collapse
Affiliation(s)
- Florian M Hermann
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Maya Friis Kjærgaard
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Chenglei Tian
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany
| | - Ulf Tiemann
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Abigail Jackson
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Maria Kraft
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Jarno L T Kettunen
- Folkhalsan Research Center, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Finland, Helsinki, Finland; Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Tiinamaija Tuomi
- Folkhalsan Research Center, Helsinki, Finland; Institute for Molecular Medicine Finland, University of Finland, Helsinki, Finland; Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Ivana Novak
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Semb
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany.
| |
Collapse
|
5
|
Ren XY, Xue MR, Yan ZL, Zhang SJ, Liu M, Li AZ. Clinical Characteristics and Gene Mutations of Two Families with MODY 3 in Inner Mongolia. Pharmgenomics Pers Med 2022; 15:1019-1027. [PMID: 36567880 PMCID: PMC9785186 DOI: 10.2147/pgpm.s371141] [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: 06/16/2022] [Accepted: 09/29/2022] [Indexed: 12/23/2022] Open
Abstract
Objective This study aimed to analyze the clinical characteristics and gene mutations of two families with maturity-onset diabetes of the young 3 (MODY 3) in Inner Mongolia. Methods Fifty-three patients in Inner Mongolia suspected of having MODY 3 were enrolled in this study according to clinical manifestations. Blood samples were collected, and all exons of the HNF1α gene were analyzed; the second-generation DNA of the splicing regions of the gene was determined by direct sequencing. Results In Family 1, the proband, mother, and uncle all carried the missense heterozygous mutation on exon 2 of the HNF1α gene (c.512G>A, p.Arg171Gln), and both the proband and uncle had MODY 3. In Family 2, the proband, grandfather, father, uncle I, and uncle II all carried a missense mutation on exon 2 (c.391C>t, p.Arg131Trp), and all had MODY 3. The blood glucose control in these patients was stable while they were being treated with oral sulfonylurea hypoglycemic drugs alone or with insulin. Uncle II had serious macrovascular and microvascular complications. Conclusion Maturity-onset diabetes of the young 3 gene mutations (c.512G>A, p.Arg171Gln) and (c.391C>T, p.Arg131Trp) may be the main pathogenic genes of the two families with MODY 3. The two gene mutations found in this study have not been reported previously in China.
Collapse
Affiliation(s)
- Xiao-Yan Ren
- Department of Endocrinology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, People’s Republic of China
| | - Meng-Ruo Xue
- Department of Interventional Radiology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, People’s Republic of China
| | - Zhao-Li Yan
- Department of Endocrinology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, People’s Republic of China,Correspondence: Zhao-Li Yan, Department of Endocrinology, Affiliated Hospital of Inner Mongolia Medical University, No. 1, North Tongdao Street, Huiming District, Hohhot, 010050, People’s Republic of China, Tel +86 13848177245, Email
| | - Shao-Jie Zhang
- Department of Anatomy, Inner Mongolia Medical University, Hohhot, 010050, People’s Republic of China
| | - Min Liu
- Department of Endocrinology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, People’s Republic of China
| | - Ai-Zhen Li
- Department of Endocrinology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, People’s Republic of China
| |
Collapse
|
6
|
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
|
7
|
A novel splice-affecting HNF1A variant with large population impact on diabetes in Greenland. THE LANCET REGIONAL HEALTH. EUROPE 2022; 24:100529. [PMID: 36649380 PMCID: PMC9832271 DOI: 10.1016/j.lanepe.2022.100529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/15/2022] [Accepted: 10/03/2022] [Indexed: 11/12/2022]
Abstract
Background The genetic disease architecture of Inuit includes a large number of common high-impact variants. Identification of such variants contributes to our understanding of the genetic aetiology of diseases and improves global equity in genomic personalised medicine. We aimed to identify and characterise novel variants in genes associated with Maturity Onset Diabetes of the Young (MODY) in the Greenlandic population. Methods Using combined data from Greenlandic population cohorts of 4497 individuals, including 448 whole genome sequenced individuals, we screened 14 known MODY genes for previously identified and novel variants. We functionally characterised an identified novel variant and assessed its association with diabetes prevalence and cardiometabolic traits and population impact. Findings We identified a novel variant in the known MODY gene HNF1A with an allele frequency of 1.9% in the Greenlandic Inuit and absent elsewhere. Functional assays indicate that it prevents normal splicing of the gene. The variant caused lower 30-min insulin (β = -232 pmol/L, βSD = -0.695, P = 4.43 × 10-4) and higher 30-min glucose (β = 1.20 mmol/L, βSD = 0.441, P = 0.0271) during an oral glucose tolerance test. Furthermore, the variant was associated with type 2 diabetes (OR 4.35, P = 7.24 × 10-6) and HbA1c (β = 0.113 HbA1c%, βSD = 0.205, P = 7.84 × 10-3). The variant explained 2.5% of diabetes variance in Greenland. Interpretation The reported variant has the largest population impact of any previously reported variant within a MODY gene. Together with the recessive TBC1D4 variant, we show that close to 1 in 5 cases of diabetes (18%) in Greenland are associated with high-impact genetic variants compared to 1-3% in large populations. Funding Novo Nordisk Foundation, Independent Research Fund Denmark, and Karen Elise Jensen's Foundation.
Collapse
|
8
|
Beucher A, Miguel-Escalada I, Balboa D, De Vas MG, Maestro MA, Garcia-Hurtado J, Bernal A, Gonzalez-Franco R, Vargiu P, Heyn H, Ravassard P, Ortega S, Ferrer J. The HASTER lncRNA promoter is a cis-acting transcriptional stabilizer of HNF1A. Nat Cell Biol 2022; 24:1528-1540. [PMID: 36202974 PMCID: PMC9586874 DOI: 10.1038/s41556-022-00996-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/16/2022] [Indexed: 11/08/2022]
Abstract
The biological purpose of long non-coding RNAs (lncRNAs) is poorly understood. Haploinsufficient mutations in HNF1A homeobox A (HNF1A), encoding a homeodomain transcription factor, cause diabetes mellitus. Here, we examine HASTER, the promoter of an lncRNA antisense to HNF1A. Using mouse and human models, we show that HASTER maintains cell-specific physiological HNF1A concentrations through positive and negative feedback loops. Pancreatic β cells from Haster mutant mice consequently showed variegated HNF1A silencing or overexpression, resulting in hyperglycaemia. HASTER-dependent negative feedback was essential to prevent HNF1A binding to inappropriate genomic regions. We demonstrate that the HASTER promoter DNA, rather than the lncRNA, modulates HNF1A promoter-enhancer interactions in cis and thereby regulates HNF1A transcription. Our studies expose a cis-regulatory element that is unlike classic enhancers or silencers, it stabilizes the transcription of its target gene and ensures the fidelity of a cell-specific transcription factor program. They also show that disruption of a mammalian lncRNA promoter can cause diabetes mellitus.
Collapse
Affiliation(s)
- Anthony Beucher
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Irene Miguel-Escalada
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Diego Balboa
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Matías G De Vas
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Miguel Angel Maestro
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Javier Garcia-Hurtado
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Aina Bernal
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Roser Gonzalez-Franco
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Holger Heyn
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Philippe Ravassard
- Biotechnology and Biotherapy Team, Institut du Cerveau et de la Moelle, CNRS UMR7225, INSERM U975, University Pierre et Marie Curie, Paris, France
| | - Sagrario Ortega
- Transgenics Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Jorge Ferrer
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain.
| |
Collapse
|
9
|
Miyachi Y, Miyazawa T, Ogawa Y. HNF1A Mutations and Beta Cell Dysfunction in Diabetes. Int J Mol Sci 2022; 23:ijms23063222. [PMID: 35328643 PMCID: PMC8948720 DOI: 10.3390/ijms23063222] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/26/2022] Open
Abstract
Understanding the genetic factors of diabetes is essential for addressing the global increase in type 2 diabetes. HNF1A mutations cause a monogenic form of diabetes called maturity-onset diabetes of the young (MODY), and HNF1A single-nucleotide polymorphisms are associated with the development of type 2 diabetes. Numerous studies have been conducted, mainly using genetically modified mice, to explore the molecular basis for the development of diabetes caused by HNF1A mutations, and to reveal the roles of HNF1A in multiple organs, including insulin secretion from pancreatic beta cells, lipid metabolism and protein synthesis in the liver, and urinary glucose reabsorption in the kidneys. Recent studies using human stem cells that mimic MODY have provided new insights into beta cell dysfunction. In this article, we discuss the involvement of HNF1A in beta cell dysfunction by reviewing previous studies using genetically modified mice and recent findings in human stem cell-derived beta cells.
Collapse
|
10
|
Barbetti F, Rapini N, Schiaffini R, Bizzarri C, Cianfarani S. The application of precision medicine in monogenic diabetes. Expert Rev Endocrinol Metab 2022; 17:111-129. [PMID: 35230204 DOI: 10.1080/17446651.2022.2035216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/25/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Monogenic diabetes, a form of diabetes mellitus, is caused by a mutation in a single gene and may account for 1-2% of all clinical forms of diabetes. To date, more than 40 loci have been associated with either isolated or syndromic monogenic diabetes. AREAS COVERED While the request of a genetic test is mandatory for cases with diabetes onset in the first 6 months of life, a decision may be difficult for childhood or adolescent diabetes. In an effort to assist the clinician in this task, we have grouped monogenic diabetes genes according to the age of onset (or incidental discovery) of hyperglycemia and described the additional clinical features found in syndromic diabetes. The therapeutic options available are reviewed. EXPERT OPINION Technical improvements in DNA sequencing allow for rapid, simultaneous analysis of all genes involved in monogenic diabetes, progressively shrinking the area of unsolved cases. However, the complexity of the analysis of genetic data requires close cooperation between the geneticist and the diabetologist, who should play a proactive role by providing a detailed clinical phenotype that might match a specific disease gene.
Collapse
Affiliation(s)
- Fabrizio Barbetti
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Diabetology and Growth Disorders Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Novella Rapini
- Diabetology and Growth Disorders Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Riccardo Schiaffini
- Diabetology and Growth Disorders Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Carla Bizzarri
- Diabetology and Growth Disorders Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stefano Cianfarani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Dipartimento Pediatrico Universitario Ospedaliero, IRCCS "Bambino Gesù" Children's Hospital, Rome, Italy
- Department of Women's and Children Health, Karolisnska Institute and University Hospital, Sweden
| |
Collapse
|
11
|
Lien YC, Pinney SE, Lu XM, Simmons RA. Identification of Novel Regulatory Regions Induced by Intrauterine Growth Restriction in Rat Islets. Endocrinology 2022; 163:6459683. [PMID: 34894232 PMCID: PMC8743043 DOI: 10.1210/endocr/bqab251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 01/05/2023]
Abstract
Intrauterine growth restriction (IUGR) leads to the development of type 2 diabetes in adulthood, and the permanent alterations in gene expression implicate an epigenetic mechanism. Using a rat model of IUGR, we performed TrueSeq-HELP Tagging to assess the association of DNA methylation changes and gene dysregulation in islets. We identified 511 differentially methylated regions (DMRs) and 4377 significantly altered single CpG sites. Integrating the methylome and our published transcriptome data sets resulted in the identification of pathways critical for islet function. The identified DMRs were enriched with transcription factor binding motifs, such as Elk1, Etv1, Foxa1, Foxa2, Pax7, Stat3, Hnf1, and AR. In silico analysis of 3-dimensional chromosomal interactions using human pancreas and islet Hi-C data sets identified interactions between 14 highly conserved DMRs and 35 genes with significant expression changes at an early age, many of which persisted in adult islets. In adult islets, there were far more interactions between DMRs and genes with significant expression changes identified with Hi-C, and most of them were critical to islet metabolism and insulin secretion. The methylome was integrated with our published genome-wide histone modification data sets from IUGR islets, resulting in further characterization of important regulatory regions of the genome altered by IUGR containing both significant changes in DNA methylation and specific histone marks. We identified novel regulatory regions in islets after exposure to IUGR, suggesting that epigenetic changes at key transcription factor binding motifs and other gene regulatory regions may contribute to gene dysregulation and an abnormal islet phenotype in IUGR rats.
Collapse
Affiliation(s)
- Yu-Chin Lien
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Sara E Pinney
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Xueqing Maggie Lu
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Correspondence: Rebecca A. Simmons, MD, Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, BRB II/III, 13th Fl, Rm 1308, 421 Curie Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Barth R, Ruoso C, Ferreira SM, de Ramos FC, Lima FB, Boschero AC, Santos GJD. Hepatocyte Nuclear Factor 4-α (HNF4α) controls the insulin resistance-induced pancreatic β-cell mass expansion. Life Sci 2022; 289:120213. [PMID: 34902439 DOI: 10.1016/j.lfs.2021.120213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Regardless of the etiology, any type of DM presents a reduction of insulin-secreting cell mass, so it is important to investigate pathways that induce the increase of this cell mass. AIM Based on the fact that (1) HNF4α is crucial for β-cell proliferation, (2) DEX-induced IR promotes β-cell mass expansion, and (3) the stimulation of β-cell mass expansion may be an important target for DM therapies, we aimed to investigate whether DEX-induced proliferation of β pancreatic cells is dependent on HNF4α. METHODS We used WildType (WT) and Knockout (KO) mice for HNF4-α, treated or not with 100 mg/Kg/day of DEX, for 5 consecutive days. One day after the last injection of DEX the IR was confirmed by ipITT and the mice were euthanized for pancreas removal. RESULTS In comparison to WT, KO mice presented increased glucose tolerance, lower fasting glucose and increased glucose-stimulates insulin secretion (GSIS). DEX induced IR in both KO and WT mice. In addition, DEX-induced β-cell mass expansion and an increase in the Ki67 immunostaining were observed only in WT mice, evidencing that IR-induced β-cell mass expansion is dependent on HNF4α. Also, we observed that DEX-treatment, in an HNF4α-dependent way, promoted an increase in PDX1, PAX4 and NGN3 gene expression. CONCLUSIONS Our results strongly suggest that DEX-induced IR promotes β-cell mass expansion through processes of proliferation and neogenesis that depend on the HNF4α activity, pointing to HNF4α as a possible therapeutic target in DM treatment.
Collapse
Affiliation(s)
- Robson Barth
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Carolina Ruoso
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Sandra Mara Ferreira
- Laboratory of endocrine pancreas and metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, 13083-862 Campinas, Brazil
| | - Francieli Caroline de Ramos
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Fernanda Barbosa Lima
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Antônio Carlos Boschero
- Laboratory of endocrine pancreas and metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, 13083-862 Campinas, Brazil
| | - Gustavo Jorge Dos Santos
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil.
| |
Collapse
|
13
|
Role of Actionable Genes in Pursuing a True Approach of Precision Medicine in Monogenic Diabetes. Genes (Basel) 2022; 13:genes13010117. [PMID: 35052457 PMCID: PMC8774614 DOI: 10.3390/genes13010117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/16/2022] Open
Abstract
Monogenic diabetes is a genetic disorder caused by one or more variations in a single gene. It encompasses a broad spectrum of heterogeneous conditions, including neonatal diabetes, maturity onset diabetes of the young (MODY) and syndromic diabetes, affecting 1-5% of patients with diabetes. Some of these variants are harbored by genes whose altered function can be tackled by specific actions ("actionable genes"). In suspected patients, molecular diagnosis allows the implementation of effective approaches of precision medicine so as to allow individual interventions aimed to prevent, mitigate or delay clinical outcomes. This review will almost exclusively concentrate on the clinical strategy that can be specifically pursued in carriers of mutations in "actionable genes", including ABCC8, KCNJ11, GCK, HNF1A, HNF4A, HNF1B, PPARG, GATA4 and GATA6. For each of them we will provide a short background on what is known about gene function and dysfunction. Then, we will discuss how the identification of their mutations in individuals with this form of diabetes, can be used in daily clinical practice to implement specific monitoring and treatments. We hope this article will help clinical diabetologists carefully consider who of their patients deserves timely genetic testing for monogenic diabetes.
Collapse
|
14
|
Zhao Q, Ding L, Yang Y, Sun J, Wang M, Li X, Liu M. Clinical Characteristics of Patients With HNF1-alpha MODY: A Literature Review and Retrospective Chart Review. Front Endocrinol (Lausanne) 2022; 13:900489. [PMID: 35795147 PMCID: PMC9252268 DOI: 10.3389/fendo.2022.900489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical manifestation of hepatocyte nuclear factor-1-alpha (HNF1-alpha) maturity-onset diabetes of the young (MODY) is highly variable. This study aims to investigate the clinical characteristics of patients with HNF1-alpha MODY in general, by geographical regions (Asian or non-Asian), HNF1-alpha mutations, and islet autoantibody status. A literature review and a chart review of patients with HNF1-alpha MODY were performed. The means and proportions from studies were pooled using the inverse variance method for pooling, and subgroup analyses were performed. A total of 109 studies involving 1,325 patients [41.5%, 95% confidence interval (CI): 35.2, 48.1; male] were identified. The mean age of diagnosis was 20.3 years (95% CI: 18.3-22.2), and the mean glycated hemoglobin was 7.3% (95% CI: 7.2-7.5). In comparison, Asian patients exhibited significantly higher HbA1c (p = 0.007) and 2-h post-load C-peptide (p = 0.012) levels and lower levels of triglyceride (TG) (p < 0.001), total cholesterol (TC) (p < 0.001), and high-density lipoprotein cholesterol (HDL-c) (p < 0.001) and less often had macrovascular complications (p = 0.014). The age of diagnosis was oldest in patients with mutations in the transactivation domain (p < 0.001). The levels of 2-h post-load C-peptide (p < 0.001), TG (p = 0.007), TC (p = 0.017), and HDL-c (p = 0.001) were highest and the prevalence of diabetic neuropathy was lowest (p = 0.024) in patients with DNA-binding domain mutations. The fasting (p = 0.004) and 2-h post-load glucose (p = 0.003) levels and the prevalence of diabetic neuropathy (p = 0.010) were higher among patients with positive islet autoantibodies. The study demonstrated that the clinical manifestations of HNF1-alpha MODY differed by geographical regions, HNF1-alpha mutations, and islet autoantibody status.
Collapse
Affiliation(s)
- Qinying Zhao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhong Sun
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Xin Li, ; Ming Liu,
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- National Health Commission (NHC) Key Laboratory of Hormones and Development, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Endocrinology, Tianjin, China
- *Correspondence: Xin Li, ; Ming Liu,
| |
Collapse
|
15
|
Ontawong A, Duangjai A, Srimaroeng C. Coffea arabica bean extract inhibits glucose transport and disaccharidase activity in Caco-2 cells. Biomed Rep 2021; 15:73. [PMID: 34405045 PMCID: PMC8329997 DOI: 10.3892/br.2021.1449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/14/2021] [Indexed: 11/06/2022] Open
Abstract
The major constituents of Coffea arabica (coffee), including caffeine, chlorogenic acid and caffeic acid, exhibit antihyperglycemic properties in in vitro and in vivo models. However, whether Coffea arabica bean extract (CBE) regulates glucose uptake activity and the underlying mechanisms involved remain unclear. The aim of the present study was to examine the effects of CBE on glucose absorption and identify the mechanisms involved using an in vitro model. The uptake of a fluorescent glucose analog into Caco-2 colorectal adenocarcinoma cells was determined. The expression levels of sodium glucose co-transporter 1 (SGLT1) and glucose transporter 2 (GLUT2) were evaluated. In addition, glycoside hydrolase enzyme activity was investigated. It was observed that CBE inhibited disaccharidase enzyme activity. Furthermore, CBE exerted an inhibitory effect on intestinal glucose absorption by downregulating SGLT1- and GLUT2-mediated 5' AMP-activated protein kinase phosphorylation and suppressing hepatocyte nuclear factor 1α expression. These data suggest that CBE may attenuate glucose absorption and may have potentially beneficial antihyperglycemic effects in the body; however, the mechanisms underlying the effects of CBE must be elucidated through further investigation.
Collapse
Affiliation(s)
- Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, Muang Phayao, Phayao 56000, Thailand
| | - Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Muang Phayao, Phayao 56000, Thailand
| | - Chutima Srimaroeng
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Nong Khai 52000, Thailand
| |
Collapse
|
16
|
Oropeza D, Cigliola V, Romero A, Chera S, Rodríguez-Seguí SA, Herrera PL. Stage-specific transcriptomic changes in pancreatic α-cells after massive β-cell loss. BMC Genomics 2021; 22:585. [PMID: 34340653 PMCID: PMC8330016 DOI: 10.1186/s12864-021-07812-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Background Loss of pancreatic insulin-secreting β-cells due to metabolic or autoimmune damage leads to the development of diabetes. The discovery that α-cells can be efficiently reprogrammed into insulin-secreting cells in mice and humans has opened promising avenues for innovative diabetes therapies. β-cell loss triggers spontaneous reprogramming of only 1–2% of α-cells, limiting the extent of regeneration. Most α-cells are refractory to conversion and their global transcriptomic response to severe β-cell loss as well as the mechanisms opposing their reprogramming into insulin producers are largely unknown. Here, we performed RNA-seq on FAC-sorted α-cells to characterize their global transcriptional responses at different time points after massive β-cell ablation. Results Our results show that α-cells undergo stage-specific transcriptional changes 5- and 15-days post-diphtheria toxin (DT)-mediated β-cell ablation. At 5 days, α-cells transiently upregulate various genes associated with interferon signaling and proliferation, including Interferon Induced Protein with Tetratricopeptide Repeats 3 (Ifit3). Subsequently, at 15 days post β-cell ablation, α-cells undergo a transient downregulation of genes from several pathways including Insulin receptor, mTOR and MET signaling. Conclusions The results presented here pinpoint novel markers discriminating α-cells at different stages after acute β-cell loss, and highlight additional signaling pathways that are modulated in α-cells in this context. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07812-x.
Collapse
Affiliation(s)
- Daniel Oropeza
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valentina Cigliola
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Present address: Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.,Regeneration Next, Duke University, Durham, North Carolina, 27710, USA
| | - Agustín Romero
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Simona Chera
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Clinical Science, Center for Diabetes Research, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Santiago A Rodríguez-Seguí
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina.
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
17
|
Decreased GLUT2 and glucose uptake contribute to insulin secretion defects in MODY3/HNF1A hiPSC-derived mutant β cells. Nat Commun 2021; 12:3133. [PMID: 34035238 PMCID: PMC8149827 DOI: 10.1038/s41467-021-22843-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/04/2021] [Indexed: 12/13/2022] Open
Abstract
Heterozygous HNF1A gene mutations can cause maturity onset diabetes of the young 3 (MODY3), characterized by insulin secretion defects. However, specific mechanisms of MODY3 in humans remain unclear due to lack of access to diseased human pancreatic cells. Here, we utilize MODY3 patient-derived human induced pluripotent stem cells (hiPSCs) to study the effect(s) of a causal HNF1A+/H126D mutation on pancreatic function. Molecular dynamics simulations predict that the H126D mutation could compromise DNA binding and gene target transcription. Genome-wide RNA-Seq and ChIP-Seq analyses on MODY3 hiPSC-derived endocrine progenitors reveal numerous HNF1A gene targets affected by the mutation. We find decreased glucose transporter GLUT2 expression, which is associated with reduced glucose uptake and ATP production in the MODY3 hiPSC-derived β-like cells. Overall, our findings reveal the importance of HNF1A in regulating GLUT2 and several genes involved in insulin secretion that can account for the insulin secretory defect clinically observed in MODY3 patients. Heterozygous HNF1A mutations can give rise to maturity onset diabetes of the young 3 (MODY3), characterized by insulin secretion defects. Here the authors show that MODY3-related HNF1A mutation in patient hiPSCderived pancreatic cells decreases glucose transporter GLUT2 expression due to compromised DNA binding.
Collapse
|
18
|
Zhang L, Sheng C, Zhou F, Zhu K, Wang S, Liu Q, Yuan M, Xu Z, Liu Y, Lu J, Liu J, Zhou L, Wang X. CBP/p300 HAT maintains the gene network critical for β cell identity and functional maturity. Cell Death Dis 2021; 12:476. [PMID: 33980820 PMCID: PMC8116341 DOI: 10.1038/s41419-021-03761-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022]
Abstract
Loss of β cell identity and functional immaturity are thought to be involved in β cell failure in type 2 diabetes. CREB-binding protein (CBP) and its paralogue p300 act as multifunctional transcriptional co-activators and histone acetyltransferases (HAT) with extensive biological functions. However, whether the regulatory role of CBP/p300 in islet β cell function depends on the HAT activity remains uncertain. In this current study, A-485, a selective inhibitor of CBP/p300 HAT activity, greatly impaired glucose-stimulated insulin secretion from rat islets in vitro and in vivo. RNA-sequencing analysis showed a comprehensive downregulation of β cell and α cell identity genes in A-485-treated islets, without upregulation of dedifferentiation markers and derepression of disallowed genes. A-485 treatment decreased the expressions of genes involved in glucose sensing, not in glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation. In the islets of prediabetic db/db mice, CBP/p300 displayed a significant decrease with key genes for β cell function. The deacetylation of histone H3K27 as well as the transcription factors Hnf1α and Foxo1 was involved in CBP/p300 HAT inactivation-repressed expressions of β cell identity and functional genes. These findings highlight the dominant role of CBP/p300 HAT in the maintenance of β cell identity by governing transcription network.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunxiang Sheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feiye Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kecheng Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shushu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianqian Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miaomiao Yuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoqian Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Sanchez Caballero L, Gorgogietas V, Arroyo MN, Igoillo-Esteve M. Molecular mechanisms of β-cell dysfunction and death in monogenic forms of diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:139-256. [PMID: 33832649 DOI: 10.1016/bs.ircmb.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monogenetic forms of diabetes represent 1%-5% of all diabetes cases and are caused by mutations in a single gene. These mutations, that affect genes involved in pancreatic β-cell development, function and survival, or insulin regulation, may be dominant or recessive, inherited or de novo. Most patients with monogenic diabetes are very commonly misdiagnosed as having type 1 or type 2 diabetes. The severity of their symptoms depends on the nature of the mutation, the function of the affected gene and, in some cases, the influence of additional genetic or environmental factors that modulate severity and penetrance. In some patients, diabetes is accompanied by other syndromic features such as deafness, blindness, microcephaly, liver and intestinal defects, among others. The age of diabetes onset may also vary from neonatal until early adulthood manifestations. Since the different mutations result in diverse clinical presentations, patients usually need different treatments that range from just diet and exercise, to the requirement of exogenous insulin or other hypoglycemic drugs, e.g., sulfonylureas or glucagon-like peptide 1 analogs to control their glycemia. As a consequence, awareness and correct diagnosis are crucial for the proper management and treatment of monogenic diabetes patients. In this chapter, we describe mutations causing different monogenic forms of diabetes associated with inadequate pancreas development or impaired β-cell function and survival, and discuss the molecular mechanisms involved in β-cell demise.
Collapse
Affiliation(s)
- Laura Sanchez Caballero
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Vyron Gorgogietas
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Maria Nicol Arroyo
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/.
| |
Collapse
|
20
|
Berger C, Zdzieblo D. Glucose transporters in pancreatic islets. Pflugers Arch 2020; 472:1249-1272. [PMID: 32394191 PMCID: PMC7462922 DOI: 10.1007/s00424-020-02383-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
The fine-tuning of glucose uptake mechanisms is rendered by various glucose transporters with distinct transport characteristics. In the pancreatic islet, facilitative diffusion glucose transporters (GLUTs), and sodium-glucose cotransporters (SGLTs) contribute to glucose uptake and represent important components in the glucose-stimulated hormone release from endocrine cells, therefore playing a crucial role in blood glucose homeostasis. This review summarizes the current knowledge about cell type-specific expression profiles as well as proven and putative functions of distinct GLUT and SGLT family members in the human and rodent pancreatic islet and further discusses their possible involvement in onset and progression of diabetes mellitus. In context of GLUTs, we focus on GLUT2, characterizing the main glucose transporter in insulin-secreting β-cells in rodents. In addition, we discuss recent data proposing that other GLUT family members, namely GLUT1 and GLUT3, render this task in humans. Finally, we summarize latest information about SGLT1 and SGLT2 as representatives of the SGLT family that have been reported to be expressed predominantly in the α-cell population with a suggested functional role in the regulation of glucagon release.
Collapse
Affiliation(s)
- Constantin Berger
- Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Daniela Zdzieblo
- Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies, Neunerplatz 2, 97082, Würzburg, Germany.
| |
Collapse
|
21
|
Defective functions of HNF1A variants on BCL2L1 transactivation and beta-cell growth. Biochem Biophys Res Commun 2020; 529:826-833. [PMID: 32684311 DOI: 10.1016/j.bbrc.2020.05.155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 10/23/2022]
Abstract
Maturity-onset diabetes of the young type 3 (MODY3) is caused by mutations in a gene encoding transcription factor hepatocyte nuclear factor 1-alpha (HNF1A). Although the roles of HNF1A in regulation of hepatic and pancreatic genes to maintain glucose homeostasis were investigated, the functions of HNF1A are not completely elucidated. To better understand the functions of HNF1A, we characterized mutations of HNF1A in Thai MODY3 patients and studied the functions of wild-type HNF1A and variant proteins. We demonstrate for the first time that HNF1A upregulates transactivation of an anti-apoptotic gene BCL2 Like 1 (BCL2L1) and that all the identified HNF1A variants including p.D80V, p.R203C, p.P475L, and p.G554fsX556, reduce this ability. The four HNF1A variants impair HNF1A function in promoting INS-1 cell transition from G1 to S phase of cell cycle, which thereby retard cell growth. This finding indicates the role of HNF1A in beta-cell viability by upregulation of anti-apoptotic gene expression and also reaffirms its role in beta-cell growth through cell cycle control.
Collapse
|
22
|
Abstract
MODY (Maturity Onset Diabetes of the Young) is a type of diabetes resulting from a pathogenic effect of gene mutations. Up to date, 13 MODY genes are known. Gene HNF1A is one of the most common causes of MODY diabetes (HNF1A-MODY; MODY3). This gene is polymorphic and more than 1200 pathogenic and non-pathogenic HNF1A variants were described in its UTRs, exons and introns. For HNF1A-MODY, not just gene but also phenotype heterogeneity is typical. Although there are some clinical instructions, HNF1A-MODY patients often do not meet every diagnostic criteria or they are still misdiagnosed as type 1 and type 2 diabetics. There is a constant effort to find suitable biomarkers to help with in distinguishing of MODY3 from Type 1 Diabetes (T1D) and Type 2 Diabetes (T2D). DNA sequencing is still necessary for unambiguous confirmation of clinical suspicion of MODY. NGS (Next Generation Sequencing) methods brought discoveries of multiple new gene variants and new instructions for their pathogenicity classification were required. The most actual problem is classification of variants with uncertain significance (VUS) which is a stumbling-block for clinical interpretation. Since MODY is a hereditary disease, DNA analysis of family members is helpful or even crucial. This review is updated summary about HNF1A-MODY genetics, pathophysiology, clinics functional studies and variant classification.
Collapse
|
23
|
Nocera D, Menniti M, Belviso S, Bond HM, Lanzillotta D, Spoleti CB, Guagliardi MR, Malatesta P, Trapasso F, Irace C, Perrotti N, Iuliano R. Functional characterization of p.Pro409His variant in HNF1A, a hypomorphic mutation involved in pancreatic β-cell dysfunction. Acta Diabetol 2019; 56:883-888. [PMID: 30963309 DOI: 10.1007/s00592-019-01298-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/05/2019] [Indexed: 12/31/2022]
Abstract
AIMS HNF1A is a gene coding for the transcription factor HNF1-α, mutated in some forms of MODY and type 2 diabetes mellitus characterized by a strong genetic component. The penetrance of HNF1A variants differs considerably; thus, to assess the genetic risk of diabetes in carrier subjects of a HNF1A mutant allele, a functional characterization of mutant forms is of paramount importance. METHODS The HNF1A gene was sequenced in two patients with partly discordant diabetic phenotype, carrying the p.Pro409His variant. To evaluate the pathogenicity of the variant, we measured the transactivation power of the corresponding P408H HNF1-α mutant mouse form on HNF1-α target promoters. RESULTS We found a lower but detectable activity of transactivation of the mutant form compared with the wild-type form and we excluded mechanisms of protein degradation or nuclear mislocalization. CONCLUSIONS The HNF1A mutation p.Pro409His can be considered a mild variant that confers a moderate risk of type 2 diabetes mellitus in heterozygous carriers.
Collapse
Affiliation(s)
- Donatella Nocera
- Unit of Medical Genetics, University Hospital Mater Domini, Viale T. Campanella, 115, Catanzaro, Italy
| | - Miranda Menniti
- Department of Human Health, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Stefania Belviso
- Department of Human Health, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Heather Mandy Bond
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Delia Lanzillotta
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Cristina Barbara Spoleti
- Department of Human Health, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Maria Rosaria Guagliardi
- Unit of Medical Genetics, University Hospital Mater Domini, Viale T. Campanella, 115, Catanzaro, Italy
| | - Paola Malatesta
- Unit of Medical Genetics, University Hospital Mater Domini, Viale T. Campanella, 115, Catanzaro, Italy
| | - Francesco Trapasso
- Unit of Medical Genetics, University Hospital Mater Domini, Viale T. Campanella, 115, Catanzaro, Italy
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Concetta Irace
- Department of Human Health, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy
| | - Nicola Perrotti
- Unit of Medical Genetics, University Hospital Mater Domini, Viale T. Campanella, 115, Catanzaro, Italy.
- Department of Human Health, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy.
| | - Rodolfo Iuliano
- Unit of Medical Genetics, University Hospital Mater Domini, Viale T. Campanella, 115, Catanzaro, Italy.
- Department of Human Health, University Magna Graecia of Catanzaro, Campus S. Venuta, Viale Europa, località Germaneto, 88100, Catanzaro, Italy.
| |
Collapse
|
24
|
Ning M, Duarte JD, Stevison F, Isoherranen N, Rubin LH, Jeong H. Determinants of Cytochrome P450 2D6 mRNA Levels in Healthy Human Liver Tissue. Clin Transl Sci 2019; 12:416-423. [PMID: 30821899 PMCID: PMC6618095 DOI: 10.1111/cts.12632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/07/2019] [Indexed: 12/28/2022] Open
Abstract
Cytochrome P450 2D6 (CYP2D6) is a major drug‐metabolizing enzyme that exhibits large interindividual variability. Recent studies suggest that differential transcriptional regulation of CYP2D6 in part may be responsible for the variability. In this study, we characterized potential determinants of CYP 2D6 transcript levels in healthy human liver tissue samples (n = 115), including genetic polymorphisms in CYP2D6 and the genes encoding transcription regulators for CYP2D6 expression; mRNA expression of the transcription factors and their known target genes; and hepatic levels of bile acids and retinoids, agents that modulate the expression/activity of the transcription factors. Their associations with CYP2D6 mRNA levels in the tissues were examined. Results from multivariable linear regression analysis revealed CYP8B1 mRNA level and rs3892097, the single‐ nucleotide polymorphism defining the nonfunctional CYP2D6*4 allele, as the two most significant predictors of CYP2D6 mRNA levels in the liver tissue samples, explaining 30% of the variability.
Collapse
Affiliation(s)
- Miaoran Ning
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Julio D Duarte
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Center for Pharmacogenomics, University of Florida, Gainesville, Florida, USA
| | - Faith Stevison
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Leah H Rubin
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Neurology, Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Hyunyoung Jeong
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
25
|
The E3 SUMO ligase PIASγ is a novel interaction partner regulating the activity of diabetes associated hepatocyte nuclear factor-1α. Sci Rep 2018; 8:12780. [PMID: 30143652 PMCID: PMC6109179 DOI: 10.1038/s41598-018-29448-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023] Open
Abstract
The transcription factor hepatocyte nuclear factor-1α (HNF-1A) is involved in normal pancreas development and function. Rare variants in the HNF1A gene can cause monogenic diabetes, while common variants confer type 2 diabetes risk. The precise mechanisms for regulation of HNF-1A, including the role and function of post-translational modifications, are still largely unknown. Here, we present the first evidence for HNF-1A being a substrate of SUMOylation in cellulo and identify two lysine (K) residues (K205 and K273) as SUMOylation sites. Overexpression of protein inhibitor of activated STAT (PIASγ) represses the transcriptional activity of HNF-1A and is dependent on simultaneous HNF-1A SUMOylation at K205 and K273. Moreover, PIASγ is a novel HNF-1A interaction partner whose expression leads to translocation of HNF-1A to the nuclear periphery. Thus, our findings support that the E3 SUMO ligase PIASγ regulates HNF-1A SUMOylation with functional implications, representing new targets for drug development and precision medicine in diabetes.
Collapse
|
26
|
Functional assessment of genetic variants located in the promoter of SHP1 (NR0B2). Pharmacogenet Genomics 2018; 27:410-415. [PMID: 28873070 DOI: 10.1097/fpc.0000000000000310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Small heterodimer partner 1 (SHP1, NR0B2) is a member of the superfamily of nuclear receptors (NRs). Even if this orphan receptor, unlike other NRs, lacks the DNA-binding domain, it is capable of regulating transcription by repressing the activity of other NRs by direct protein-protein interaction. Accordingly, SHP1 is part of negative feedback loops of the transcriptional regulation of genes involved in drug metabolism and various metabolic pathways including bile acid and glucose homeostasis. Although it is known that several interacting partners of SHP1 also modulate its expression, there is little information about genetic variability of this regulatory mechanism. Our study aimed to identify genetic variants in the NR0B2 promoter region and to determine their impact on NR0B2 transcription. For this, DNA samples originating from 119 participants of the population-based cohort Study of Health in Pomerania were analyzed by Sanger sequencing revealing four genetic variants: NR0B2:c.-594T>C (rs71636795), NR0B2:c.-414G>C (newly identified), NR0B2:c.-423C>T (rs78182695), and NR0B2:c.-224delCTGA (rs145613139) localized in the 5' untranslated region of NR0B2. The impact of these variants on transactivation of the NR0B2 promoter by NRs known to be regulators of SHP1 expression (hepatocyte nuclear factor 4α, liver receptor homolog-1, and farnesoid X receptor) was assessed in a cell-based reporter gene assay, showing that transactivation by hepatocyte nuclear factor 4α and liver receptor homolog-1 was significantly decreased in the presence of the genetic variant NR0B2:c.-594T>C, even though this effect was cell specific. However, SHP1 mRNA expression in a small collection of human kidney samples was not affected by these genetic variants.
Collapse
|
27
|
The molecular functions of hepatocyte nuclear factors - In and beyond the liver. J Hepatol 2018; 68:1033-1048. [PMID: 29175243 DOI: 10.1016/j.jhep.2017.11.026] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/27/2022]
Abstract
The hepatocyte nuclear factors (HNFs) namely HNF1α/β, FOXA1/2/3, HNF4α/γ and ONECUT1/2 are expressed in a variety of tissues and organs, including the liver, pancreas and kidney. The spatial and temporal manner of HNF expression regulates embryonic development and subsequently the development of multiple tissues during adulthood. Though the HNFs were initially identified individually based on their roles in the liver, numerous studies have now revealed that the HNFs cross-regulate one another and exhibit synergistic relationships in the regulation of tissue development and function. The complex HNF transcriptional regulatory networks have largely been elucidated in rodent models, but less so in human biological systems. Several heterozygous mutations in these HNFs were found to cause diseases in humans but not in rodents, suggesting clear species-specific differences in mutational mechanisms that remain to be uncovered. In this review, we compare and contrast the expression patterns of the HNFs, the HNF cross-regulatory networks and how these liver-enriched transcription factors serve multiple functions in the liver and beyond, extending our focus to the pancreas and kidney. We also summarise the insights gained from both human and rodent studies of mutations in several HNFs that are known to lead to different disease conditions.
Collapse
|
28
|
Dyer EM, Amin R. Fifteen-minute consultation: Monogenic forms of diabetes with onset after age 6 months. Arch Dis Child Educ Pract Ed 2018; 103:58-64. [PMID: 28754804 DOI: 10.1136/archdischild-2016-311470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 06/08/2017] [Accepted: 06/10/2017] [Indexed: 11/03/2022]
Abstract
Monogenic forms of diabetes (historically known as Maturity Onset Diabetes of the Young (MODY)) are caused by single gene mutations inherited in an autosomal dominant fashion that result in reduced pancreatic beta cell function. Children with these forms of diabetes may be misdiagnosed as having type 1 or 2 diabetes, which has important implications for treatment, genetic counselling, screening of family members and prognosis. Useful tools now exist to aid in their diagnosis and management. Here, we attempt to outline the clinical features that will help the physician make the differentiation from other diabetes subtypes.
Collapse
Affiliation(s)
- Emma M Dyer
- Department of Paediatrics, Lewisham University Hospital, London, UK
| | - Rakesh Amin
- Department of Endocrinology and Diabetes, Great Ormond Street Hospital London, London, UK
| |
Collapse
|
29
|
Naqvi AAT, Hasan GM, Hassan MI. Investigating the role of transcription factors of pancreas development in pancreatic cancer. Pancreatology 2018; 18:184-190. [PMID: 29289465 DOI: 10.1016/j.pan.2017.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 11/20/2017] [Accepted: 12/22/2017] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer (PC) is the seventh most common cause of cancer-related deaths worldwide that kills more than 300,000 people every year. Prognosis of PC is very poor with a five-year survival rate about 5%. The most common and highly observed type of PC is pancreatic ductal adenocarcinoma (PDAC). It is preceded by the progression of precursor lesions such as Pancreatic Intraepithelial Neoplasia (PanIN), Intraductal Papillary Neoplasm (IPMN) and Mucinous Cystic Neoplasm (MCN). PanIN is the most common among these premalignant lesions. Genes orchestrating the origin and differentiation of cells during organogenesis have the tendency to produce tumor cells in response to activating or inactivating mutations. Based on the following premise, we discuss the role of transcription factors (TFs) of pancreas development and cell fate differentiation in PC. Pancreas/duodenum homeobox protein 1 (PDX1), Pancreas transcription factor 1 subunit alpha (PTF1A), Nuclear receptor subfamily 5 group A member 2 (NR5A2), Hepatocyte nuclear factor 1-alpha (HNF1A) and Hepatocyte nuclear factor 1-beta (HNF1B) play vital role in the development and differentiation of pancreatic precursor cells. Mutated KRAS induces abnormalities in the regular function of these TFs which in turn cause abnormal cell growth and proliferation that leads to cancer. Thus, these TFs are highly susceptible for the origin of PC. Therefore, we propose that these TFs can be treated as therapeutic targets for the development of anticancer drugs.
Collapse
Affiliation(s)
- Ahmad Abu Turab Naqvi
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
30
|
Bensellam M, Jonas JC, Laybutt DR. Mechanisms of β-cell dedifferentiation in diabetes: recent findings and future research directions. J Endocrinol 2018; 236:R109-R143. [PMID: 29203573 DOI: 10.1530/joe-17-0516] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/04/2017] [Indexed: 12/13/2022]
Abstract
Like all the cells of an organism, pancreatic β-cells originate from embryonic stem cells through a complex cellular process termed differentiation. Differentiation involves the coordinated and tightly controlled activation/repression of specific effectors and gene clusters in a time-dependent fashion thereby giving rise to particular morphological and functional cellular features. Interestingly, cellular differentiation is not a unidirectional process. Indeed, growing evidence suggests that under certain conditions, mature β-cells can lose, to various degrees, their differentiated phenotype and cellular identity and regress to a less differentiated or a precursor-like state. This concept is termed dedifferentiation and has been proposed, besides cell death, as a contributing factor to the loss of functional β-cell mass in diabetes. β-cell dedifferentiation involves: (1) the downregulation of β-cell-enriched genes, including key transcription factors, insulin, glucose metabolism genes, protein processing and secretory pathway genes; (2) the concomitant upregulation of genes suppressed or expressed at very low levels in normal β-cells, the β-cell forbidden genes; and (3) the likely upregulation of progenitor cell genes. These alterations lead to phenotypic reconfiguration of β-cells and ultimately defective insulin secretion. While the major role of glucotoxicity in β-cell dedifferentiation is well established, the precise mechanisms involved are still under investigation. This review highlights the identified molecular mechanisms implicated in β-cell dedifferentiation including oxidative stress, endoplasmic reticulum (ER) stress, inflammation and hypoxia. It discusses the role of Foxo1, Myc and inhibitor of differentiation proteins and underscores the emerging role of non-coding RNAs. Finally, it proposes a novel hypothesis of β-cell dedifferentiation as a potential adaptive mechanism to escape cell death under stress conditions.
Collapse
Affiliation(s)
- Mohammed Bensellam
- Garvan Institute of Medical ResearchSydney, New South Wales, Australia
- Université Catholique de LouvainInstitut de Recherche Expérimentale et Clinique, Pôle d'Endocrinologie, Diabète et Nutrition, Brussels, Belgium
| | - Jean-Christophe Jonas
- Université Catholique de LouvainInstitut de Recherche Expérimentale et Clinique, Pôle d'Endocrinologie, Diabète et Nutrition, Brussels, Belgium
| | - D Ross Laybutt
- Garvan Institute of Medical ResearchSydney, New South Wales, Australia
- St Vincent's Clinical SchoolUNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
31
|
Xu J, Wang L, Tang J, Jia G, Liu G, Chen X, Cai J, Shang H, Zhao H. Pancreatic atrophy caused by dietary selenium deficiency induces hypoinsulinemic hyperglycemia via global down-regulation of selenoprotein encoding genes in broilers. PLoS One 2017; 12:e0182079. [PMID: 28763492 PMCID: PMC5538751 DOI: 10.1371/journal.pone.0182079] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
This study was envisaged to comprehensively profile genes in selected tissues along with a few biochemical indicators and integrate resulting information with dietary selenium (Se) deficiency symptoms in broilers. A total of 120 one-day-old Cobb male broilers were equally divided into two groups and fed a Se deficient corn-soybean-based basal diet supplemented with 0.3 mg/kg sodium selenite (Control, Se adequate) or without selenite (Se deficiency) for five weeks. Effects of Se deficiency on mRNA abundance of twenty-three selenoprotein encoding genes and seventeen insulin signaling related genes were studied at day 35 in pancreas, liver and muscle along with plasma biochemical constituents and enzyme activities. Compared to healthy birds in control diet, Se deficient diet induced deficiency symptoms in 90% birds and classic nutritional pancreatic atrophy, depressed growth performance of broilers, and decreased (P < 0.01 to P < 0.05) total antioxidant capacity and activities of superoxide dismutase and glutathione peroxidase in plasma and three other tissues. Se deficiency resulted in 58% higher mortality than control birds. Dietary Se deficiency down-regulated (P < 0.01-0.05) eighteen selenoprotein encoding genes in pancreas, fourteen genes in muscle and nine genes in liver, and up-regulated (P < 0.05) Txnrd1 and Selx in liver. Meanwhile, six, thirteen and five insulin signaling related genes were down-regulated (P < 0.01-0.05) in pancreas, muscle and liver, respectively, and three genes were up-regulated (P < 0.01) in liver. The decrease (P < 0.05) in levels of plasma insulin, total triglyceride and total cholesterol, and concurrent elevated (P < 0.05) levels of plasma glucose and inflammatory cytokines accompanied the global down-regulation of selenoprotein encoding- and insulin signaling related- genes in Se deficient birds. It was concluded that dietary Se deficiency induces nutritional pancreatic atrophy and metabolic disorder of glucose and lipid in broilers via down-regulation of selenoprotein encoding- and insulin signaling related- genes, indicating potential roles of these genes in metabolic regulation.
Collapse
Affiliation(s)
- Jingyang Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Longqiong Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jiayong Tang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Trace Element Research Center, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Jia
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Trace Element Research Center, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guangmang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoling Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jingyi Cai
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Haiying Shang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hua Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Trace Element Research Center, Sichuan Agricultural University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
32
|
Wang X, Li W, Ma L, Ping F, Liu J, Wu X, Mao J, Wang X, Nie M. Variants in MODY genes associated with maternal lipids profiles in second trimester of pregnancy. J Gene Med 2017; 19. [PMID: 28591938 DOI: 10.1002/jgm.2962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/15/2017] [Accepted: 06/03/2017] [Indexed: 12/18/2022] Open
Affiliation(s)
- Xiaojing Wang
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Wei Li
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Liangkun Ma
- Department of Obstetrics and Gynecology; Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Fan Ping
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Juntao Liu
- Department of Obstetrics and Gynecology; Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Xueyan Wu
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Jiangfeng Mao
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Xi Wang
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| | - Min Nie
- Key Laboratory of Endocrinology, Ministry of Health; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences; Beijing China
| |
Collapse
|
33
|
Abstract
Maturity Onset Diabetes of the Young type 3 (MODY3), linked to mutations in the transcription factor HNF1A, is the most prevalent form of monogenic diabetes mellitus. HNF1alpha-deficiency leads to defective insulin secretion via a molecular mechanism that is still not completely understood. Moreover, in MODY3 patients the severity of insulin secretion can be extremely variable even in the same kindred, indicating that modifier genes may control the onset of the disease. With the use of a mouse model for HNF1alpha-deficiency, we show here that specific genetic backgrounds (C3H and CBA) carry a powerful genetic suppressor of diabetes. A genome scan analysis led to the identification of a major suppressor locus on chromosome 3 (Moda1). Moda1 locus contains 11 genes with non-synonymous SNPs that significantly interacts with other loci on chromosomes 4, 11 and 18. Mechanistically, the absence of HNF1alpha in diabetic-prone (sensitive) strains leads to postnatal defective islets growth that is remarkably restored in resistant strains. Our findings are relevant to human genetics since Moda1 is syntenic with a human locus identified by genome wide association studies of fasting glycemia in patients. Most importantly, our results show that a single genetic locus can completely suppress diabetes in Hnf1a-deficiency.
Collapse
|
34
|
Singh V, Saluja N. Phylogenetic and promoter analysis of islet amyloid polypeptide gene causing type 2 diabetes in mammalian species. Int J Diabetes Dev Ctries 2016. [DOI: 10.1007/s13410-016-0508-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
35
|
Khelifa SB, Dendana A, Barboura I, Khochtali I, Chahed H, Ferchichi S, Miled A. Successful switch from insulin to oral sulfonylurea therapy in HNF1A-MODY Tunisian patient with the P291fsinsC mutation. Diabetes Res Clin Pract 2016; 115:133-6. [PMID: 26822262 DOI: 10.1016/j.diabres.2016.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/23/2015] [Accepted: 01/08/2016] [Indexed: 12/16/2022]
Abstract
The hot spot mutation P291fsinsC was identified for the first time in a 26 years old Tunisian woman. The low serum level of high C-reactive protein was helpful to target the HNF1A gene. Due to the molecular diagnosis, the change from insulin to sulfonylurea therapy was performed successfully.
Collapse
Affiliation(s)
| | - Azza Dendana
- Biochemistry Laboratory, CHU Farhat Hached, Sousse 4000, Tunisia.
| | - Ilhem Barboura
- Biochemistry Laboratory, CHU Farhat Hached, Sousse 4000, Tunisia.
| | - Ines Khochtali
- Endocrinology Unit, University Hospital of Monastir, Monastir 5000, Tunisia.
| | - Hinda Chahed
- Biochemistry Laboratory, CHU Farhat Hached, Sousse 4000, Tunisia.
| | - Selima Ferchichi
- Biochemistry Laboratory, CHU Farhat Hached, Sousse 4000, Tunisia.
| | - Abdelhedi Miled
- Biochemistry Laboratory, CHU Farhat Hached, Sousse 4000, Tunisia.
| |
Collapse
|
36
|
Balamurugan K, Bjørkhaug L, Mahajan S, Kanthimathi S, Njølstad PR, Srinivasan N, Mohan V, Radha V. Structure-function studies of HNF1A (MODY3) gene mutations in South Indian patients with monogenic diabetes. Clin Genet 2016; 90:486-495. [PMID: 26853433 DOI: 10.1111/cge.12757] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 12/30/2022]
Abstract
Maturity-onset diabetes of the young (MODY) is a genetically heterogeneous monogenic form of diabetes characterized by onset of diabetes below 25 years of age, autosomal dominant mode of inheritance and primary defect in insulin secretion. Mutations in the gene (HNF1A) encoding transcription factor hepatocyte nuclear factor 1A (HNF-1A) results in one of the most common forms of MODY (MODY3). HNF-1A is mainly enriched in pancreatic β-cells and hepatocytes and important for organ development and normal pancreatic function. We here report on the functional interrogation of eight missense HNF1A mutations associated with MODY3 in South Indian subjects, and the contributing effect of common variant (S487N) within HNF1A. Of the eight mutations, three mutations (p.R171G, p.G245R and p.R263H), in particular, affected HNF-1A function in transfected HeLa cells by reducing both transcriptional activity and nuclear localization, possibly due to disruption of the integrity of the three dimensional structure. The common variant p.S487N contributed further to the loss-of-function of p.R271Q (p.R271Q+p.S487N double mutant), in vitro, on both activity and localization. Our data on the first functional study of HNF1A mutations in South India subjects confers that the defect of the HNF-1A mutant proteins are responsible for MODY3 diabetes in these patients.
Collapse
Affiliation(s)
- K Balamurugan
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - L Bjørkhaug
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - S Mahajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - S Kanthimathi
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - P R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - N Srinivasan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - V Mohan
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| | - V Radha
- Department of Molecular genetics, Madras Diabetes Research Foundation, ICMR Advanced Centre for Genomics of Type 2 Diabetes and Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention & Control, IDF Centre of Education, Chennai, India
| |
Collapse
|
37
|
Yuk JM, Jin HS, Jo EK. Small Heterodimer Partner and Innate Immune Regulation. Endocrinol Metab (Seoul) 2016; 31:17-24. [PMID: 26754583 PMCID: PMC4803555 DOI: 10.3803/enm.2016.31.1.17] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 12/21/2015] [Accepted: 12/31/2015] [Indexed: 12/31/2022] Open
Abstract
The nuclear receptor superfamily consists of the steroid and non-steroid hormone receptors and the orphan nuclear receptors. Small heterodimer partner (SHP) is an orphan family nuclear receptor that plays an essential role in the regulation of glucose and cholesterol metabolism. Recent studies reported a previously unidentified role for SHP in the regulation of innate immunity and inflammation. The innate immune system has a critical function in the initial response against a variety of microbial and danger signals. Activation of the innate immune response results in the induction of inflammatory cytokines and chemokines to promote anti-microbial effects. An excessive or uncontrolled inflammatory response is potentially harmful to the host, and can cause tissue damage or pathological threat. Therefore, the innate immune response should be tightly regulated to enhance host defense while preventing unwanted immune pathologic responses. In this review, we discuss recent studies showing that SHP is involved in the negative regulation of toll-like receptor-induced and NLRP3 (NACHT, LRR and PYD domains-containing protein 3)-mediated inflammatory responses in innate immune cells. Understanding the function of SHP in innate immune cells will allow us to prevent or modulate acute and chronic inflammation processes in cases where dysregulated innate immune activation results in damage to normal tissues.
Collapse
Affiliation(s)
- Jae Min Yuk
- Department of Infection Biology, Chungnam National University School of Medicine, Daejeon, Korea
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyo Sun Jin
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eun Kyeong Jo
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea.
| |
Collapse
|
38
|
Huang X, Tang J, Xu J, Jia G, Liu G, Chen X, Cai J, Shang H, Zhao H. Supranutritional dietary selenium induced hyperinsulinemia and dyslipidemia via affected expression of selenoprotein genes and insulin signal-related genes in broiler. RSC Adv 2016. [DOI: 10.1039/c6ra14932d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The purpose of this study was to investigate the effects of supranutritional selenium (Se) on the mRNA expression of selenoprotein genes and insulin signal-related genes in the liver, muscle and pancreas of the broiler.
Collapse
Affiliation(s)
- Xiaofeng Huang
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Jiayong Tang
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Jingyang Xu
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Gang Jia
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Guangmang Liu
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Xiaoling Chen
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Jingyi Cai
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Haiying Shang
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| | - Hua Zhao
- Animal Nutrition Institute
- Sichuan Agricultural University
- Chengdu
- China
| |
Collapse
|
39
|
Ahn HY, Kim HH, Kim YA, Kim M, Ohn JH, Chung SS, Lee YK, Park DJ, Park KS, Moore DD, Park YJ. Thyroid Hormone Regulates the mRNA Expression of Small Heterodimer Partner through Liver Receptor Homolog-1. Endocrinol Metab (Seoul) 2015; 30:584-92. [PMID: 26485468 PMCID: PMC4722415 DOI: 10.3803/enm.2015.30.4.584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/24/2015] [Accepted: 09/24/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Expression of hepatic cholesterol 7α-hydroxylase (CYP7A1) is negatively regulated by orphan nuclear receptor small heterodimer partner (SHP). In this study, we aimed to find whether thyroid hormone regulates SHP expression by modulating the transcriptional activities of liver receptor homolog-1 (LRH-1). METHODS We injected thyroid hormone (triiodothyronine, T3) to C57BL/6J wild type. RNA was isolated from mouse liver and used for microarray analysis and quantitative real-time polymerase chain reaction (PCR). Human hepatoma cell and primary hepatocytes from mouse liver were used to confirm the effect of T3 in vitro. Promoter assay and electrophoretic mobility-shift assay (EMSA) were also performed using human hepatoma cell line. RESULTS Initial microarray results indicated that SHP expression is markedly decreased in livers of T3 treated mice. We confirmed that T3 repressed SHP expression in the liver of mice as well as in mouse primary hepatocytes and human hepatoma cells by real-time PCR analysis. LRH-1 increased the promoter activity of SHP; however, this increased activity was markedly decreased after thyroid hormone receptor β/retinoid X receptor α/T3 administration. EMSA revealed that T3 inhibits specific LRH-1 DNA binding. CONCLUSION We found that thyroid hormone regulates the expression of SHP mRNA through interference with the transcription factor, LRH-1.
Collapse
Affiliation(s)
- Hwa Young Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hwan Hee Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ye An Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Hun Ohn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Soo Chung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yoon Kwang Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Do Joon Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
40
|
Han EH, Gorman AA, Singh P, Chi YI. Repression of HNF1α-mediated transcription by amino-terminal enhancer of split (AES). Biochem Biophys Res Commun 2015; 468:14-20. [PMID: 26549228 DOI: 10.1016/j.bbrc.2015.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/02/2015] [Indexed: 12/23/2022]
Abstract
HNF1α (Hepatocyte Nuclear Factor 1α) is one of the master regulators in pancreatic beta-cell development and function, and the mutations in Hnf1α are the most common monogenic causes of diabetes mellitus. As a member of the POU transcription factor family, HNF1α exerts its gene regulatory function through various molecular interactions; however, there is a paucity of knowledge in their functional complex formation. In this study, we identified the Groucho protein AES (Amino-terminal Enhancer of Split) as a HNF1α-specific physical binding partner and functional repressor of HNF1α-mediated transcription, which has a direct link to glucose-stimulated insulin secretion in beta-cells that is impaired in the HNF1α mutation-driven diabetes.
Collapse
Affiliation(s)
- Eun Hee Han
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Amanda A Gorman
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Puja Singh
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Young-In Chi
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN 55912, USA.
| |
Collapse
|
41
|
Fang C, Huang J, Huang Y, Chen L, Chen X, Hu J. A novel nonsense mutation of the HNF1α in maturity-onset diabetes of the young type 3 in Asian population. Diabetes Res Clin Pract 2015; 109:e5-7. [PMID: 26050565 DOI: 10.1016/j.diabres.2015.05.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 02/13/2015] [Accepted: 05/02/2015] [Indexed: 11/18/2022]
Abstract
We reported a novel nonsense mutation, R54X of the hepatic nuclear factor 1α (HNF1α) gene in a Chinese family. The mutation was identified in a 47 years old woman and her 19 years old daughter within a five-family members tested. Both the two patients were sensitive to insulin and glibenclamide treatments.
Collapse
Affiliation(s)
- Chen Fang
- Department of Endocrinology, 2nd Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Jian Huang
- School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou 215123, China
| | - Yun Huang
- Department of Endocrinology, 2nd Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Linqi Chen
- Department of Endocrinology, Children's Hospital of Soochow University, Suzhou 215003, China
| | - Xiuli Chen
- Department of Endocrinology, Children's Hospital of Soochow University, Suzhou 215003, China
| | - Ji Hu
- Department of Endocrinology, 2nd Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
42
|
Jonasson ME, Wicklow BA, Sellers EAC, Dolinsky VW, Doucette CA. Exploring the role of the HNF-1αG319S polymorphism in β cell failure and youth-onset type 2 diabetes: Lessons from MODY and Hnf-1α-deficient animal models. Biochem Cell Biol 2015; 93:487-94. [PMID: 26176428 DOI: 10.1139/bcb-2015-0021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The prevalence of youth-onset type 2 diabetes (T2D) is rapidly increasing worldwide, disproportionately affecting Indigenous youth with Oji-Cree heritage from central Canada. Candidate gene screening has uncovered a novel and private polymorphism in the Oji-Cree population in the hepatocyte nuclear factor-1 alpha (HNF-1α) gene, where a highly conserved glycine residue at position 319 is changed to a serine (termed HNF-1αG319S or simply G319S). Oji-Cree youth who carry one or two copies of the "S-allele" present at diagnosis with less obesity, reduced indicators of insulin resistance, and lower plasma insulin levels at diagnosis, suggestive of a primary defect in the insulin-secreting β cells. Few studies on the impact of the HNF-1αG319S variant on β cell function have been performed to date; however, much can be learned from other clinical phenotypes of HNF-1α-deficiency, including HNF-1α mutations that cause maturity-onset diabetes of the young 3 (MODY3). In addition, evaluation of Hnf-1α-deficient murine models reveals that HNF-1α plays a central role in the regulation of insulin secretion by regulating the expression of key genes involved in β cell glucose-sensing, mitochondrial function, and the maintenance of the β cell phenotype in differentiated β cells. The overall goal of this minireview is to explore the impact of HNF-1α-deficiency on the β cell to better inform future research into the mechanisms of β cell dysfunction in Oji-Cree youth with T2D.
Collapse
Affiliation(s)
- Michael E Jonasson
- d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Brandy A Wicklow
- b Department of Pediatrics and Child Health, University of Manitoba, CE-208 Childrens Hospital, 840 Sherbrook Street, Health Sciences Centre, Winnipeg, MB R3A 1S1, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Elizabeth A C Sellers
- b Department of Pediatrics and Child Health, University of Manitoba, CE-208 Childrens Hospital, 840 Sherbrook Street, Health Sciences Centre, Winnipeg, MB R3A 1S1, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Vernon W Dolinsky
- c Department of Pharmacology and Therapeutics, A203 Chown Bldg., 753 McDermot Avenue, University of Manitoba, Winnipeg, MB R3E 0T6, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Christine A Doucette
- a College of Medicine, Faculty of Health Sciences, Department of Physiology & Pathophysiology, 432 Basic Medical Sciences Building, 745 Bannatyne Avenue, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| |
Collapse
|
43
|
Benet M, Guzmán C, Pisonero-Vaquero S, García-Mediavilla MV, Sánchez-Campos S, Martínez-Chantar ML, Donato MT, Castell JV, Jover R. Repression of the nuclear receptor small heterodimer partner by steatotic drugs and in advanced nonalcoholic fatty liver disease. Mol Pharmacol 2015; 87:582-94. [PMID: 25576488 DOI: 10.1124/mol.114.096313] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The small heterodimer partner (SHP) (NR0B2) is an atypical nuclear receptor that lacks a DNA-binding domain. It interacts with and inhibits many transcription factors, affecting key metabolic processes, including bile acid, cholesterol, fatty acid, and drug metabolism. Our aim was to determine the influence of steatotic drugs and nonalcoholic fatty liver disease (NAFLD) on SHP expression and investigate the potential mechanisms. SHP was found to be repressed by steatotic drugs (valproate, doxycycline, tetracycline, and cyclosporin A) in cultured hepatic cells and the livers of different animal models of NAFLD: iatrogenic (tetracycline-treated rats), genetic (glycine N-methyltransferase-deficient mice), and nutritional (mice fed a methionine- and choline-deficient diet). Among the different transcription factors investigated, CCAAT-enhancer-binding protein α (C/EBPα) showed the strongest dominant-repressive effect on SHP expression in HepG2 and human hepatocytes. Reporter assays revealed that the inhibitory effect of C/EBPα and steatotic drugs colocalize between -340 and -509 base pair of the SHP promoter, and mutation of a predicted C/EBPα response element at -473 base pair abolished SHP repression by both C/EBPα and drugs. Moreover, inhibition of major stress signaling pathways demonstrated that the mitogen-activated protein kinase kinase 1/2 pathway activates, while the phosphatidylinositol 3 kinase pathway represses SHP in a C/EBP-dependent manner. We conclude that SHP is downregulated by several steatotic drugs and in advanced NAFLD. These conditions can activate signals that target C/EBPα and consequently repress SHP, thus favoring the progression and severity of NAFLD.
Collapse
Affiliation(s)
- Marta Benet
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Carla Guzmán
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Sandra Pisonero-Vaquero
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Victoria García-Mediavilla
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Sonia Sánchez-Campos
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Luz Martínez-Chantar
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Teresa Donato
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - José Vicente Castell
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Ramiro Jover
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| |
Collapse
|
44
|
Ahluwalia TS, Allin KH, Sandholt CH, Sparsø TH, Jørgensen ME, Rowe M, Christensen C, Brandslund I, Lauritzen T, Linneberg A, Husemoen LL, Jørgensen T, Hansen T, Grarup N, Pedersen O. Discovery of coding genetic variants influencing diabetes-related serum biomarkers and their impact on risk of type 2 diabetes. J Clin Endocrinol Metab 2015; 100:E664-71. [PMID: 25599387 DOI: 10.1210/jc.2014-3677] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Type 2 diabetes (T2D) prevalence is spiraling globally, and knowledge of its pathophysiological signatures is crucial for a better understanding and treatment of the disease. OBJECTIVE We aimed to discover underlying coding genetic variants influencing fasting serum levels of nine biomarkers associated with T2D: adiponectin, C-reactive protein, ferritin, heat shock 70-kDa protein 1B, IGF binding protein 1 and IGF binding protein 2, IL-18, IL-2 receptor-α, and leptin. DESIGN AND PARTICIPANTS A population-based sample of 6215 adult Danes was genotyped for 16 340 coding single-nucleotide polymorphisms and were tested for association with each biomarker. Identified loci were tested for association with T2D through a large-scale meta-analysis involving up to 17 024 T2D cases and up to 64 186 controls. RESULTS We discovered 11 associations between single-nucleotide polymorphisms and five distinct biomarkers at a study-wide P < 3.4 × 10(-7). Nine associations were novel: IL18: BIRC6, RAD17, MARVELD2; ferritin: F5; IGF binding protein 1: SERPING1, KLKB, GCKR, CELSR2, and heat shock 70-kDa protein 1B: CFH. Three of the identified loci (CELSR2, HNF1A, and GCKR) were significantly associated with T2D, of which the association with the CELSR2 locus has not been shown previously. CONCLUSION The identified loci influence processes related to insulin signaling, cell communication, immune function, apoptosis, DNA repair, and oxidative stress, all of which could provide a rationale for novel diabetes therapeutic strategies.
Collapse
Affiliation(s)
- Tarunveer Singh Ahluwalia
- Novo Nordisk Foundation Center for Basic Metabolic Research (T.S.A., K.H.A., C.H.S., T.H.A., T.H., N.G., O.P.), Section of Metabolic Genetics, Faculty of Health and Medical Sciences, Department of Clinical Medicine (A.L.), Faculty of Health and Medical Sciences, and Institute of Public Health (T.J.), University of Copenhagen, 2100 Copenhagen, Denmark; Copenhagen Prospective Studies on Asthma in Childhood (T.S.A.), Faculty of Health and Medical Sciences, University of Copenhagen, 2820 Copenhagen, Denmark; The Danish Paediatric Asthma Center (T.S.A.), Gentofte Hospital, The Capital Region, 2820 Copenhagen, Denmark; Steno Diabetes Center (E.J.), 2820 Gentofte, Denmark; Tethys Bioscience Inc (M.R.), Emeryville, California 94608; Departments of Internal Medicine and Endocrinology (C.C.) and Clinical Biochemistry (I.B.), Vejle Hospital, 7100 Vejle, Denmark; Institute of Regional Health Research (I.B.), and Faculty of Health Sciences (T.H.), University of Southern Denmark, 5230 Odense, Denmark; Department of General Practice (T.L.), Aarhus University, 8000 C Aarhus, Denmark; Research Centre for Prevention and Health (A.L., L.L.H., T.J.), The Capital Region of Denmark, Copenhagen, Denmark; Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, 2600 Glostrup, Denmark; and Faculty of Medicine (T.J.), University of Aalborg, 9220 Aalborg, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Shende VR, Wu M, Singh AB, Dong B, Kan CFK, Liu J. Reduction of circulating PCSK9 and LDL-C levels by liver-specific knockdown of HNF1α in normolipidemic mice. J Lipid Res 2015; 56:801-9. [PMID: 25652089 DOI: 10.1194/jlr.m052969] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The transcription factors hepatic nuclear factor (HNF)1α and HNF1β can bind to the HNF1 site on the proprotein convertase subtilisin/kexin type 9 (PCSK9) promoter to activate transcription in HepG2 cells. However, it is unknown whether one or both HNF1 factors are obligatory for transactivating hepatic PCSK9 gene expression in vivo. We developed shRNA adenoviral constructs (Ad-shHNF1α and Ad-shHNF1β) to examine the effects of knockdown of HNF1α or HNF1β on PCSK9 expression and its consequent impact on LDL receptor (LDLR) protein levels in cultured hepatic cells and liver tissue. We demonstrated that infection with Ad-shHNF1α, but not Ad-shHNF1β, markedly reduced PCSK9 mRNA expression in HepG2 cells with a concomitant increase in LDLR protein abundance. Injecting Ad-shHNF1α in mice fed a normal diet significantly (∼ 50%) reduced liver mRNA expression and serum concentration of PCSK9 with a concomitant increase (∼ 1.9-fold) in hepatic LDLR protein abundance. Furthermore, we observed a modest but significant reduction in circulating LDL cholesterol after knockdown of HNF1α in these normolipidemic mice. Consistent with the observation that knockdown of HNF1β did not affect PCSK9 mRNA or protein expression in cultured hepatic cells, Ad-shHNF1β infection in mice resulted in no change in the hepatic mRNA expression or serum content of PCSK9. Altogether, our study demonstrates that HNF1α, but not HNF1β, is the primary positive regulator of PCSK9 transcription in mouse liver.
Collapse
Affiliation(s)
- Vikram Ravindra Shende
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 Department of Medicine, Stanford University, Stanford, CA 94305
| | - Minhao Wu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | - Amar Bahadur Singh
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 Department of Medicine, Stanford University, Stanford, CA 94305
| | - Bin Dong
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | | | - Jingwen Liu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| |
Collapse
|
46
|
Yamagata K. Roles of HNF1α and HNF4α in pancreatic β-cells: lessons from a monogenic form of diabetes (MODY). VITAMINS AND HORMONES 2015; 95:407-23. [PMID: 24559927 DOI: 10.1016/b978-0-12-800174-5.00016-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the genes encoding hepatocyte nuclear factor (HNF)1α and HNF4α cause a monogenic form of diabetes mellitus known as maturity-onset diabetes of the young (MODY). The primary cause of MODY is an impairment of glucose-stimulated insulin secretion by pancreatic β-cells, indicating the important roles of HNF1α and HNF4α in β-cells. Large-scale genetic studies have clarified that the common variants of HNF1α and HNF4α genes are also associated with type 2 diabetes, suggesting that they are involved in the pathogenesis of both diseases. Recent experimental studies revealed that HNF1α controls both β-cell function and growth by regulating target genes such as glucose transporter 2, pyruvate kinase, collectrin, hepatocyte growth factor activator, and HNF4α. In contrast, HNF4α mainly regulates the function of β-cells. Although direct target genes of HNF4α in β-cells are largely unknown, we recently identified Anks4b as a novel target of HNF4α that regulates β-cell susceptibility to endoplasmic reticulum stress. Studies of MODY have led to a better understanding of the molecular mechanism of glucose-stimulated insulin secretion by pancreatic β-cells.
Collapse
Affiliation(s)
- Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
47
|
Dong B, Li H, Singh AB, Cao A, Liu J. Inhibition of PCSK9 transcription by berberine involves down-regulation of hepatic HNF1α protein expression through the ubiquitin-proteasome degradation pathway. J Biol Chem 2014; 290:4047-58. [PMID: 25540198 DOI: 10.1074/jbc.m114.597229] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our previous in vitro studies have identified hepatocyte nuclear factor 1α (HNF1α) as an obligated trans-activator for PCSK9 gene expression and demonstrated its functional involvement in the suppression of PCSK9 expression by berberine (BBR), a natural cholesterol-lowering compound. In this study, we investigated the mechanism underlying the inhibitory effect of BBR on HNF1α-mediated PCSK9 transcription. Administration of BBR to hyperlipidemic mice and hamsters lowered circulating PCSK9 concentrations and hepatic PCSK9 mRNA levels without affecting the gene expression of HNF1α. However, hepatic HNF1α protein levels were markedly reduced in BBR-treated animals as compared with the control. Using HepG2 cells as a model system, we obtained evidence that BBR treatment let to accelerated degradation of HNF1α protein. By applying inhibitors to selectively block the ubiquitin proteasome system (UPS) and autophagy-lysosomal pathway, we show that HNF1α protein content in HepG2 cells was not affected by bafilomycin A1 treatment, but it was dose-dependently increased by UPS inhibitors bortezomib and MG132. Bortezomib treatment elevated HNF1α and PCSK9 cellular levels with concomitant reductions of LDL receptor protein. Moreover, HNF1α protein displayed a multiubiquitination ladder pattern in cells treated with BBR or overexpressing ubiquitin. By expressing GFP-HNF1α fusion protein in cells, we observed that blocking UPS resulted in accumulation of GFP-HNF1α in cytoplasm. Importantly, we show that the BBR reducing effects on HNF1α protein and PCSK9 gene transcription can be eradicated by proteasome inhibitors. Altogether, our studies using BBR as a probe uncovered a new aspect of PCSK9 regulation by ubiquitin-induced proteasomal degradation of HNF1α.
Collapse
Affiliation(s)
- Bin Dong
- From the Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - Hai Li
- From the Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - Amar Bahadur Singh
- From the Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - Aiqin Cao
- From the Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - Jingwen Liu
- From the Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
48
|
Vassar R, Kuhn PH, Haass C, Kennedy ME, Rajendran L, Wong PC, Lichtenthaler SF. Function, therapeutic potential and cell biology of BACE proteases: current status and future prospects. J Neurochem 2014; 130:4-28. [PMID: 24646365 PMCID: PMC4086641 DOI: 10.1111/jnc.12715] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/12/2014] [Accepted: 03/14/2014] [Indexed: 01/18/2023]
Abstract
The β-site APP cleaving enzymes 1 and 2 (BACE1 and BACE2) were initially identified as transmembrane aspartyl proteases cleaving the amyloid precursor protein (APP). BACE1 is a major drug target for Alzheimer's disease because BACE1-mediated cleavage of APP is the first step in the generation of the pathogenic amyloid-β peptides. BACE1, which is highly expressed in the nervous system, is also required for myelination by cleaving neuregulin 1. Several recent proteomic and in vivo studies using BACE1- and BACE2-deficient mice demonstrate a much wider range of physiological substrates and functions for both proteases within and outside of the nervous system. For BACE1 this includes axon guidance, neurogenesis, muscle spindle formation, and neuronal network functions, whereas BACE2 was shown to be involved in pigmentation and pancreatic β-cell function. This review highlights the recent progress in understanding cell biology, substrates, and functions of BACE proteases and discusses the therapeutic options and potential mechanism-based liabilities, in particular for BACE inhibitors in Alzheimer's disease. The protease BACE1 is a major drug target in Alzheimer disease. Together with its homolog BACE2, both proteases have an increasing number of functions within and outside of the nervous system. This review highlights recent progress in understanding cell biology, substrates, and functions of BACE proteases and discusses the therapeutic options and potential mechanism-based liabilities, in particular for BACE inhibitors in Alzheimer disease.
Collapse
Affiliation(s)
- Robert Vassar
- Department of Cell and Molecular Biology, Feinberg University School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peer-Hendrik Kuhn
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute for Advanced Study, Technische Universität München, Garching, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Adolf-Butenandt Institute, Biochemistry, Ludwig-Maximilians University Munich, Munich, Germany
| | - Matthew E. Kennedy
- Neurosciences, Merck Research Labs, Boston, Massachusetts, USA
- Division of Psychiatry Research, University of Zurich, Zurich, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, Division of Psychiatry Research, University of Zurich, Zurich, Switzerland
- Graduate programs of the Zurich Center for Integrative Human Physiology and Zurich Neuroscience Center, University of Zurich, Zurich, Switzerland
| | - Philip C. Wong
- Departments of Pathology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute for Advanced Study, Technische Universität München, Garching, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
49
|
A new susceptibility locus for myocardial infarction, hypertension, type 2 diabetes mellitus, and dyslipidemia on chromosome 12q24. DISEASE MARKERS 2014; 2014:291419. [PMID: 25057215 PMCID: PMC4098619 DOI: 10.1155/2014/291419] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/15/2014] [Accepted: 05/28/2014] [Indexed: 02/03/2023]
Abstract
We examined the role of hepatic nuclear factor-1 alpha (HNF1a) gene polymorphism on coronary artery disease (CAD) traits in 4631 Saudi angiographed individuals (2419 CAD versus 2212 controls) using TaqMan assay on ABI Prism 7900HT sequence detection system. Following adjustment for confounders, the rs2259820_CC (1.19 (1.01–1.42); P = 0.041), rs2464196_TT (1.19 (1.00–1.40); P = 0.045), and rs2259816_T (1.13 (1.01–1.26); P = 0.031) were associated with MI. The rs2259820_T (1.14 (1.03–1.26); P = 0.011) and rs2464196_C (1.12 (1.02–1.24); P = 0.024) were associated with type 2 diabetes mellitus (T2DM), while the rs2393791_T (1.14 (1.01–1.28); P = 0.032), rs7310409_G (1.16 (1.03–1.30); P = 0.013), and rs2464196_AG+GG (1.25 (1.05–1.49); P = 0.012) were implicated in hypertension. Hypertriglyceridemia was linked to the rs2393791_T (1.14 (1.02–1.27); P = 0.018), rs7310409_G (1.12 (1.01–1.25); P = 0.031), rs1169310_G (1.15 (1.04–1.28); P = 0.010), and rs1169313_CT+TT (1.24 (1.06–1.45); P = 0.008) and high low density lipoprotein-cholesterol levels were associated with rs2259820_T (1.23 (1.07–1.41); P = 0.004), rs2464196_T (1.22 (1.06–1.39); P = 0.004), and rs2259816_T (1.18 (1.02–1.36); P = 0.023). A 7-mer haplotype CATATAC (χ2 = 7.50; P = 0.0062), constructed from the studied SNPs, was associated with MI, and CATATA implicated in T2DM (χ2 = 3.94; P = 0.047). Hypertriglyceridemia was linked to TGCGGG (χ2 = 4.26; P = 0.039), and obesity to ACGGGT (χ2 = 5.04; P = 0.025). Our results suggest that the HNF1a is a common susceptibility gene for MI, T2DM, hypertension, and dyslipidemia.
Collapse
|
50
|
Zhao L, Chen H, Zhan YQ, Li CY, Ge CH, Zhang JH, Wang XH, Yu M, Yang XM. Serine 249 phosphorylation by ATM protein kinase regulates hepatocyte nuclear factor-1α transactivation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:604-20. [PMID: 24821553 DOI: 10.1016/j.bbagrm.2014.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 05/02/2014] [Accepted: 05/05/2014] [Indexed: 12/15/2022]
Abstract
Hepatocyte nuclear factor-1 alpha (HNF1α) exerts important effects on gene expression in multiple tissues. Several studies have directly or indirectly supported the role of phosphorylation processes in the activity of HNF1α. However, the molecular mechanism of this phosphorylation remains largely unknown. Using microcapillary liquid chromatography MS/MS and biochemical assays, we identified a novel phosphorylation site in HNF1α at Ser249. We also found that the ATM protein kinase phosphorylated HNF1α at Ser249 in vitro in an ATM-dependent manner and that ATM inhibitor KU55933 treatment inhibited phosphorylation of HNF1α at Ser249 in vivo. Coimmunoprecipitation assays confirmed the association between HNF1α and ATM. Moreover, ATM enhanced HNF1α transcriptional activity in a dose-dependent manner, whereas the ATM kinase-inactive mutant did not. The use of KU55933 confirmed our observation. Compared with wild-type HNF1α, a mutation in Ser249 resulted in a pronounced decrease in HNF1α transactivation, whereas no dominant-negative effect was observed. The HNF1αSer249 mutant also exhibited normal nuclear localization but decreased DNA-binding activity. Accordingly, the functional studies of HNF1αSer249 mutant revealed a defect in glucose metabolism. Our results suggested that ATM regulates the activity of HNF1α by phosphorylation of serine 249, particularly in glucose metabolism, which provides valuable insights into the undiscovered mechanisms of ATM in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Long Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Yi-Qun Zhan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Chang-Yan Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Chang-Hui Ge
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Jian-Hong Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiao-Hui Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Miao Yu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China.
| | - Xiao-Ming Yang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 102206, China.
| |
Collapse
|