1
|
Lou QM, Lai FF, Li JW, Mao KJ, Wan HT, He Y. Mechanisms of cuproptosis and its relevance to distinct diseases. Apoptosis 2024; 29:981-1006. [PMID: 38824478 DOI: 10.1007/s10495-024-01983-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
Copper is a trace element required by the organism, but once the level of copper exceeds the threshold, it becomes toxic and even causes death. The underlying mechanisms of copper-induced death are inconclusive, with different studies showing different opinions on the mechanism of copper-induced death. Multiple investigations have shown that copper induces oxidative stress, endoplasmic reticulum stress, nucleolar stress, and proteasome inhibition, all of which can result in cell death. The latest research elucidates a copper-dependent death and denominates it as cuproptosis. Cuproptosis takes place through the combination of copper and lipoylated proteins of the tricarboxylic acid cycle, triggering agglomeration of lipoylated proteins and loss of iron-sulfur cluster proteins, leading to proteotoxic stress and ultimately death. Given the toxicity and necessity of copper, abnormal levels of copper lead to diseases such as neurological diseases and cancer. The development of cancer has a high demand for copper, neurological diseases involve the change of copper contents and the binding of copper to proteins. There is a close relationship between these two kinds of diseases and copper. Here, we summarize the mechanisms of copper-related death, and the association between copper and diseases, to better figure out the influence of copper in cell death and diseases, thus advancing the clinical remedy of these diseases.
Collapse
Affiliation(s)
- Qiao-Mei Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Fei-Fan Lai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jing-Wei Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Kun-Jun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hai-Tong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Bogdanski AM, Onnekink AM, Inderson A, Boekestijn B, Bonsing BA, Vasen HFA, van Hooft JE, Boonstra JJ, Mieog JSD, Wasser MNJM, Feshtali S, Potjer TP, Klatte DCF, van Leerdam ME. The Added Value of Blood Glucose Monitoring in High-Risk Individuals Undergoing Pancreatic Cancer Surveillance. Pancreas 2024; 53:e566-e572. [PMID: 38598368 DOI: 10.1097/mpa.0000000000002335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
OBJECTIVES The study aimed to investigate the added value of blood glucose monitoring in high-risk individuals (HRIs) participating in pancreatic cancer surveillance. MATERIALS AND METHODS High-risk individuals with a CDKN2A/p16 germline pathogenic variant participating in pancreatic cancer surveillance were included in this study. Multivariable logistic regression was performed to assess the relationship between new-onset diabetes (NOD) and pancreatic ductal adenocarcinoma (PDAC). To quantify the diagnostic performance of NOD as a marker for PDAC, receiver operating characteristic curve with area under the curve was computed. RESULTS In total, 220 HRIs were included between 2000 and 2019. Median age was 61 (interquartile range. 53-71) years and 62.7% of participants were female. During the study period, 26 (11.8%) HRIs developed NOD, of whom 5 (19.2%) later developed PDAC. The other 23 (82.1%) PDAC cases remained NOD-free. Multivariable analysis showed no statistically significant relationship between NOD and PDAC (odds ratio, 1.21; 95% confidence interval, 0.39-3.78) and 4 of 5 PDAC cases seemed to have NOD within 3 months before diagnosis. Furthermore, NOD did not differentiate between HRIs with and without PDAC (area under the curve, 0.54; 95% confidence interval, 0.46-0.61). CONCLUSIONS In this study, we found no added value for longitudinal glucose monitoring in CDKN2A pathogenic variant carriers participating in an imaging-based pancreatic cancer surveillance program.
Collapse
Affiliation(s)
| | | | - Akin Inderson
- From the Departments of Gastroenterology and Hepatology
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Wang Y, Qiu X, Liu J, Liu X, Pan J, Cai J, Liu X, Qu S. Cuproptosis-Related Biomarkers and Characterization of Immune Infiltration in Sepsis. J Inflamm Res 2024; 17:2459-2478. [PMID: 38681070 PMCID: PMC11048236 DOI: 10.2147/jir.s452980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction Sepsis is a worldwide epidemic, with high morbidity and mortality. Cuproptosis is a form of cell death that is associated with a wide range of diseases. This study aimed to explore genes associated with cuproptosis in sepsis, construct predictive models and screen for potential targets. Methods The LASSO algorithm and SVM-RFE model has been analysed the expression of cuproptosis-related genes in sepsis and immune infiltration characteristics and identified the marker genes under a diagnostic model. Gene-drug networks, mRNA-miRNA networks and PPI networks were constructed to screen for potential biological targets. The expression of marker genes was validated based on the GSE57065 dataset. Consensus clustering method was used to classify sepsis samples. Results We found 381 genes associated with the development of sepsis and discovered significantly differentially expressed cuproptosis-related genes of 16 cell types in sepsis and immune infiltration with CD8/CD4 T cells being lower. NFE2L2, NLRP3, SLC31A1, DLD, DLAT, PDHB, MTF1, CDKN2A and DLST were identified as marker genes by the LASSO algorithm and the SVM-RFE model. AUC > 0.9 was constructed for PDHB and MTF1 alone respectively. The validation group data for PDHB (P=0.00099) and MTF1 (P=7.2e-14) were statistically significant. Consistent clustering analysis confirmed two subtypes. The C1 subtype may be more relevant to cellular metabolism and the C2 subtype has some relevance to immune molecules.The results of animal experiments showed that the gene expression was consistent with the bioinformatics analysis. Discussion Our study systematically explored the relationship between sepsis and cuproptosis and constructed a diagnostic model. And, several cuproptosis-related genes may interfere with the progression of sepsis through immune cell infiltration.
Collapse
Affiliation(s)
- Yuanfeng Wang
- College of Public Health and Management, Zhejiang Provincial Key Laboratory of Watershed Science and Health, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Xu Qiu
- College of Public Health and Management, Zhejiang Provincial Key Laboratory of Watershed Science and Health, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jiao Liu
- College of Public Health and Management, Zhejiang Provincial Key Laboratory of Watershed Science and Health, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Xuanyi Liu
- College of Public Health and Management, Zhejiang Provincial Key Laboratory of Watershed Science and Health, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jialu Pan
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jiayi Cai
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Xiaodong Liu
- College of Public Health and Management, Zhejiang Provincial Key Laboratory of Watershed Science and Health, Wenzhou Medical University, Wenzhou, People’s Republic of China
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, People’s Republic of China
| | - Shugen Qu
- College of Public Health and Management, Zhejiang Provincial Key Laboratory of Watershed Science and Health, Wenzhou Medical University, Wenzhou, People’s Republic of China
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, People’s Republic of China
| |
Collapse
|
4
|
Privalova V, Sobczyk Ł, Szlachcic E, Labecka AM, Czarnoleski M. Heat tolerance in Drosophila melanogaster is influenced by oxygen conditions and mutations in cell size control pathways. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220490. [PMID: 38186282 PMCID: PMC10772611 DOI: 10.1098/rstb.2022.0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 01/09/2024] Open
Abstract
Understanding metabolic performance limitations is key to explaining the past, present and future of life. We investigated whether heat tolerance in actively flying Drosophila melanogaster is modified by individual differences in cell size and the amount of oxygen in the environment. We used two mutants with loss-of-function mutations in cell size control associated with the target of rapamycin (TOR)/insulin pathways, showing reduced (mutant rictorΔ2) or increased (mutant Mnt1) cell size in different body tissues compared to controls. Flies were exposed to a steady increase in temperature under normoxia and hypoxia until they collapsed. The upper critical temperature decreased in response to each mutation type as well as under hypoxia. Females, which have larger cells than males, had lower heat tolerance than males. Altogether, mutations in cell cycle control pathways, differences in cell size and differences in oxygen availability affected heat tolerance, but existing theories on the roles of cell size and tissue oxygenation in metabolic performance can only partially explain our results. A better understanding of how the cellular composition of the body affects metabolism may depend on the development of research models that help separate various interfering physiological parameters from the exclusive influence of cell size. This article is part of the theme issue 'The evolutionary significance of variation in metabolic rates'.
Collapse
Affiliation(s)
- Valeriya Privalova
- Life History Evolution Group, Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Łukasz Sobczyk
- Life History Evolution Group, Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Ewa Szlachcic
- Life History Evolution Group, Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Anna Maria Labecka
- Life History Evolution Group, Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Marcin Czarnoleski
- Life History Evolution Group, Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
5
|
Cha J, Aguayo-Mazzucato C, Thompson PJ. Pancreatic β-cell senescence in diabetes: mechanisms, markers and therapies. Front Endocrinol (Lausanne) 2023; 14:1212716. [PMID: 37720527 PMCID: PMC10501801 DOI: 10.3389/fendo.2023.1212716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Cellular senescence is a response to a wide variety of stressors, including DNA damage, oncogene activation and physiologic aging, and pathologically accelerated senescence contributes to human disease, including diabetes mellitus. Indeed, recent work in this field has demonstrated a role for pancreatic β-cell senescence in the pathogenesis of Type 1 Diabetes, Type 2 Diabetes and monogenic diabetes. Small molecule or genetic targeting of senescent β-cells has shown promise as a novel therapeutic approach for preventing and treating diabetes. Despite these advances, major questions remain around the molecular mechanisms driving senescence in the β-cell, identification of molecular markers that distinguish senescent from non-senescent β-cell subpopulations, and translation of proof-of-concept therapies into novel treatments for diabetes in humans. Here, we summarize the current state of the field of β-cell senescence, highlighting insights from mouse models as well as studies on human islets and β-cells. We identify markers that have been used to detect β-cell senescence to unify future research efforts in this field. We discuss emerging concepts of the natural history of senescence in β-cells, heterogeneity of senescent β-cells subpopulations, role of sex differences in senescent responses, and the consequences of senescence on integrated islet function and microenvironment. As a young and developing field, there remain many open research questions which need to be addressed to move senescence-targeted approaches towards clinical investigation.
Collapse
Affiliation(s)
- Jeeyeon Cha
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Peter J. Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
6
|
Oger F, Bourouh C, Friano ME, Courty E, Rolland L, Gromada X, Moreno M, Carney C, Rabhi N, Durand E, Amanzougarene S, Berberian L, Derhourhi M, Blanc E, Hannou SA, Denechaud PD, Benfodda Z, Meffre P, Fajas L, Kerr-Conte J, Pattou F, Froguel P, Pourcet B, Bonnefond A, Collombat P, Annicotte JS. β-Cell-Specific E2f1 Deficiency Impairs Glucose Homeostasis, β-Cell Identity, and Insulin Secretion. Diabetes 2023; 72:1112-1126. [PMID: 37216637 DOI: 10.2337/db22-0604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 05/01/2023] [Indexed: 05/24/2023]
Abstract
The loss of pancreatic β-cell identity has emerged as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cell-autonomous role of the cell-cycle regulator and transcription factor E2F1 in the maintenance of β-cell identity, insulin secretion, and glucose homeostasis. We show that the β-cell-specific loss of E2f1 function in mice triggers glucose intolerance associated with defective insulin secretion, altered endocrine cell mass, downregulation of many β-cell genes, and concomitant increase of non-β-cell markers. Mechanistically, epigenomic profiling of the promoters of these non-β-cell upregulated genes identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, promoters of downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that specific E2f1 transcriptional, cistromic, and epigenomic signatures are associated with these β-cell dysfunctions, with E2F1 directly regulating several β-cell genes at the chromatin level. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of β-cell identity genes. Our data suggest that E2F1 is critical for maintaining β-cell identity and function through sustained control of β-cell and non-β-cell transcriptional programs. ARTICLE HIGHLIGHTS β-Cell-specific E2f1 deficiency in mice impairs glucose tolerance. Loss of E2f1 function alters the ratio of α- to β-cells but does not trigger β-cell conversion into α-cells. Pharmacological inhibition of E2F activity inhibits glucose-stimulated insulin secretion and alters β- and α-cell gene expression in human islets. E2F1 maintains β-cell function and identity through control of transcriptomic and epigenetic programs.
Collapse
Affiliation(s)
- Frédérik Oger
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Cyril Bourouh
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Marika Elsa Friano
- INSERM, CNRS, Institut de Biologie Valrose, Université Côte d'Azur, Nice, France
| | - Emilie Courty
- INSERM, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Laure Rolland
- INSERM, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Xavier Gromada
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Maeva Moreno
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Charlène Carney
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Nabil Rabhi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Emmanuelle Durand
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Souhila Amanzougarene
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Lionel Berberian
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Mehdi Derhourhi
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Etienne Blanc
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Sarah Anissa Hannou
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | | | | | | | - Lluis Fajas
- Center for Integrative Genomics, Université de Lausanne, Lausanne, Switzerland
| | - Julie Kerr-Conte
- INSERM, U1190 - EGID, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - François Pattou
- INSERM, U1190 - EGID, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Philippe Froguel
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
- Department of Metabolism, Hammersmith Hospital, Imperial College London, London, U.K
| | - Benoit Pourcet
- INSERM, U1011 - EGID, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| | - Amélie Bonnefond
- INSERM, U1283 - UMR8199 - European Genomic Institute for Diabetes (EGID), CNRS, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
- Department of Metabolism, Hammersmith Hospital, Imperial College London, London, U.K
| | - Patrick Collombat
- INSERM, CNRS, Institut de Biologie Valrose, Université Côte d'Azur, Nice, France
| | - Jean-Sébastien Annicotte
- INSERM, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, CHU Lille, Université de Lille, Lille, France
| |
Collapse
|
7
|
Beck JS, Madaj Z, Cheema CT, Kara B, Bennett DA, Schneider JA, Gordon MN, Ginsberg SD, Mufson EJ, Counts SE. Co-expression network analysis of frontal cortex during the progression of Alzheimer's disease. Cereb Cortex 2022; 32:5108-5120. [PMID: 35076713 PMCID: PMC9667180 DOI: 10.1093/cercor/bhac001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/29/2023] Open
Abstract
Mechanisms of Alzheimer's disease (AD) and its putative prodromal stage, amnestic mild cognitive impairment (aMCI), involve the dysregulation of multiple candidate molecular pathways that drive selective cellular vulnerability in cognitive brain regions. However, the spatiotemporal overlap of markers for pathway dysregulation in different brain regions and cell types presents a challenge for pinpointing causal versus epiphenomenal changes characterizing disease progression. To approach this problem, we performed Weighted Gene Co-expression Network Analysis and STRING interactome analysis of gene expression patterns quantified in frontal cortex samples (Brodmann area 10) from subjects who died with a clinical diagnosis of no cognitive impairment, aMCI, or mild/moderate AD. Frontal cortex was chosen due to the relatively protracted involvement of this region in AD, which might reveal pathways associated with disease onset. A co-expressed network correlating with clinical diagnosis was functionally associated with insulin signaling, with insulin (INS) being the most highly connected gene within the network. Co-expressed networks correlating with neuropathological diagnostic criteria (e.g., NIA-Reagan Likelihood of AD) were associated with platelet-endothelium-leucocyte cell adhesion pathways and hypoxia-oxidative stress. Dysregulation of these functional pathways may represent incipient alterations impacting disease progression and the clinical presentation of aMCI and AD.
Collapse
Affiliation(s)
- John S Beck
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI 49503, USA
| | - Zachary Madaj
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Calvin T Cheema
- Department of Mathematics and Computer Science, Kalamazoo College, Kalamazoo, MI 49006, USA
| | - Betul Kara
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI 49503, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Rush Alzheimer’s Disease Research Center, Chicago, IL 60612, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Rush Alzheimer’s Disease Research Center, Chicago, IL 60612, USA
| | - Marcia N Gordon
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI 49503, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Scott E Counts
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI 49503, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Hauenstein Neurosciences Center, Mercy Health Saint Mary’s Hospital, Grand Rapids, MI 49503, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Safwan-Zaiter H, Wagner N, Wagner KD. P16INK4A-More Than a Senescence Marker. Life (Basel) 2022; 12:1332. [PMID: 36143369 PMCID: PMC9501954 DOI: 10.3390/life12091332] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is a biological feature that is characterized by gradual degeneration of function in cells, tissues, organs, or an intact organism due to the accumulation of environmental factors and stresses with time. Several factors have been attributed to aging such as oxidative stress and augmented production or exposure to reactive oxygen species, inflammatory cytokines production, telomere shortening, DNA damage, and, importantly, the deposit of senescent cells. These are irreversibly mitotically inactive, yet metabolically active cells. The reason underlying their senescence lies within the extrinsic and the intrinsic arms. The extrinsic arm is mainly characterized by the expression and the secretory profile known as the senescence-associated secretory phenotype (SASP). The intrinsic arm results from the impact of several genes meant to regulate the cell cycle, such as tumor suppressor genes. P16INK4A is a tumor suppressor and cell cycle regulator that has been linked to aging and senescence. Extensive research has revealed that p16 expression is significantly increased in senescent cells, as well as during natural aging or age-related pathologies. Based on this fact, p16 is considered as a specific biomarker for detecting senescent cells and aging. Other studies have found that p16 is not only a senescence marker, but also a protein with many functions outside of senescence and aging. In this paper, we discuss and shed light on several studies that show the different functions of p16 and provide insights in its role in several biological processes besides senescence and aging.
Collapse
Affiliation(s)
| | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France
| | | |
Collapse
|
9
|
Wagner KD, Wagner N. The Senescence Markers p16INK4A, p14ARF/p19ARF, and p21 in Organ Development and Homeostasis. Cells 2022; 11:cells11121966. [PMID: 35741095 PMCID: PMC9221567 DOI: 10.3390/cells11121966] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
It is widely accepted that senescent cells accumulate with aging. They are characterized by replicative arrest and the release of a myriad of factors commonly called the senescence-associated secretory phenotype. Despite the replicative cell cycle arrest, these cells are metabolically active and functional. The release of SASP factors is mostly thought to cause tissue dysfunction and to induce senescence in surrounding cells. As major markers for aging and senescence, p16INK4, p14ARF/p19ARF, and p21 are established. Importantly, senescence is also implicated in development, cancer, and tissue homeostasis. While many markers of senescence have been identified, none are able to unambiguously identify all senescent cells. However, increased levels of the cyclin-dependent kinase inhibitors p16INK4A and p21 are often used to identify cells with senescence-associated phenotypes. We review here the knowledge of senescence, p16INK4A, p14ARF/p19ARF, and p21 in embryonic and postnatal development and potential functions in pathophysiology and homeostasis. The establishment of senolytic therapies with the ultimate goal to improve healthy aging requires care and detailed knowledge about the involvement of senescence and senescence-associated proteins in developmental processes and homeostatic mechanism. The review contributes to these topics, summarizes open questions, and provides some directions for future research.
Collapse
|
10
|
Bin-Jumah MN, Nadeem MS, Gilani SJ, Al-Abbasi FA, Ullah I, Alzarea SI, Ghoneim MM, Alshehri S, Uddin A, Murtaza BN, Kazmi I. Genes and Longevity of Lifespan. Int J Mol Sci 2022; 23:1499. [PMID: 35163422 PMCID: PMC8836117 DOI: 10.3390/ijms23031499] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/04/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex process indicated by low energy levels, declined physiological activity, stress induced loss of homeostasis leading to the risk of diseases and mortality. Recent developments in medical sciences and an increased availability of nutritional requirements has significantly increased the average human lifespan worldwide. Several environmental and physiological factors contribute to the aging process. However, about 40% human life expectancy is inherited among generations, many lifespan associated genes, genetic mechanisms and pathways have been demonstrated during last decades. In the present review, we have evaluated many human genes and their non-human orthologs established for their role in the regulation of lifespan. The study has included more than fifty genes reported in the literature for their contributions to the longevity of life. Intact genomic DNA is essential for the life activities at the level of cell, tissue, and organ. Nucleic acids are vulnerable to oxidative stress, chemotherapies, and exposure to radiations. Efficient DNA repair mechanisms are essential for the maintenance of genomic integrity, damaged DNA is not replicated and transferred to next generations rather the presence of deleterious DNA initiates signaling cascades leading to the cell cycle arrest or apoptosis. DNA modifications, DNA methylation, histone methylation, histone acetylation and DNA damage can eventually lead towards apoptosis. The importance of calorie restriction therapy in the extension of lifespan has also been discussed. The role of pathways involved in the regulation of lifespan such as DAF-16/FOXO (forkhead box protein O1), TOR and JNK pathways has also been particularized. The study provides an updated account of genetic factors associated with the extended lifespan and their interactive contributory role with cellular pathways.
Collapse
Affiliation(s)
- May Nasser Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
- Environment and Biomaterial Unit, Health Sciences Research Center, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Sadaf Jamal Gilani
- Department of Basic Health Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Inam Ullah
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan;
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Aziz Uddin
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra 21300, Pakistan;
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology (AUST), Abbottabad 22310, Pakistan;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
11
|
Montano E, Pollice A, Lucci V, Falco G, Affinito O, La Mantia G, Vivo M, Angrisano T. Pancreatic Progenitor Commitment Is Marked by an Increase in Ink4a/Arf Expression. Biomolecules 2021; 11:biom11081124. [PMID: 34439790 PMCID: PMC8392192 DOI: 10.3390/biom11081124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 01/06/2023] Open
Abstract
The identification of the molecular mechanisms controlling early cell fate decisions in mammals is of paramount importance as the ability to determine specific lineage differentiation represents a significant opportunity for new therapies. Pancreatic Progenitor Cells (PPCs) constitute a regenerative reserve essential for the maintenance and regeneration of the pancreas. Besides, PPCs represent an excellent model for understanding pathological pancreatic cellular remodeling. Given the lack of valid markers of early endoderm, the identification of new ones is of fundamental importance. Both products of the Ink4a/Arf locus, in addition to being critical cell-cycle regulators, appear to be involved in several disease pathologies. Moreover, the locus' expression is epigenetically regulated in ES reprogramming processes, thus constituting the ideal candidates to modulate PPCs homeostasis. In this study, starting from mouse embryonic stem cells (mESCs), we analyzed the early stages of pancreatic commitment. By inducing mESCs commitment to the pancreatic lineage, we observed that both products of the Cdkn2a locus, Ink4a and Arf, mark a naïve pancreatic cellular state that resembled PPC-like specification. Treatment with epi-drugs suggests a role for chromatin remodeling in the CDKN2a (Cycline Dependent Kinase Inhibitor 2A) locus regulation in line with previous observations in other cellular systems. Our data considerably improve the comprehension of pancreatic cellular ontogeny, which could be critical for implementing pluripotent stem cells programming and reprogramming toward pancreatic lineage commitment.
Collapse
Affiliation(s)
- Elena Montano
- Department of Biology, University of Naples “Federico II”, 80147 Naples, Italy; (E.M.); (A.P.); (V.L.); (G.F.); (G.L.M.)
| | - Alessandra Pollice
- Department of Biology, University of Naples “Federico II”, 80147 Naples, Italy; (E.M.); (A.P.); (V.L.); (G.F.); (G.L.M.)
| | - Valeria Lucci
- Department of Biology, University of Naples “Federico II”, 80147 Naples, Italy; (E.M.); (A.P.); (V.L.); (G.F.); (G.L.M.)
- Department of Nuclear Medicine, IRCCS—Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy
| | - Geppino Falco
- Department of Biology, University of Naples “Federico II”, 80147 Naples, Italy; (E.M.); (A.P.); (V.L.); (G.F.); (G.L.M.)
- Department of Nuclear Medicine, IRCCS—Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy
- Biogem Scarl, Istituto di Ricerche Genetiche “Gaetano Salvatore”, 83031 Ariano Irpino, Italy
| | | | - Girolama La Mantia
- Department of Biology, University of Naples “Federico II”, 80147 Naples, Italy; (E.M.); (A.P.); (V.L.); (G.F.); (G.L.M.)
| | - Maria Vivo
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano, Italy
- Correspondence: (M.V.); (T.A.); Tel.: +39-081-679721 (T.A.)
| | - Tiziana Angrisano
- Department of Biology, University of Naples “Federico II”, 80147 Naples, Italy; (E.M.); (A.P.); (V.L.); (G.F.); (G.L.M.)
- Correspondence: (M.V.); (T.A.); Tel.: +39-081-679721 (T.A.)
| |
Collapse
|
12
|
Zhang Z, Wen H, Peng B, Weng J, Zeng F. CDKN2A deregulation in fatty liver disease and its accelerative role in the process of lipogenesis. FASEB J 2021; 35:e21230. [PMID: 33769609 DOI: 10.1096/fj.202000683r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 01/07/2023]
Abstract
Previous literature has indicated that cyclin-dependent kinase inhibitor 2 A (CDKN2A) is upregulated, while the Protein Inhibitor of Activated STAT1 (PIAS1) is downregulated in the liver tissues of obese mice. The current study aimed to investigate the relationship between CDKN2A and PIAS1 in the lipogenesis of fatty liver disease. In the C57BL/6J db/db mouse model and hepatocyte model of fatty liver, the expression pattern of CDKN2A, PIAS1, Protein arginine methyltransferase 1 (PRMT1) and CASP8 and FADD-like apoptosis regulator (CFLAR) was characterized by RNA quantitative and Western blot analysis. The lipogenesis-related genes (Srebp1c and Fas) in the liver tissues and cells were employed in the assessment of lipogenesis in response to gain- or loss-of-function of CDKN2A, PIAS1, PRMT1, and CFLAR, while triglyceride and fat content were evaluated in relation to fat accumulation. Western blot analysis was conducted to determine c-Jun amino-terminal kinase (JNK) phosphorylation, while the ubiquitination of CFLAR and SUMOylation of PIAS1 was examined by immunoprecipitation. PIAS1 and CFLAR were downregulated, while CDKN2A, PRMT1, and phosphorylation of JNK was elevated in the tissues and cells of the fatty liver models. Our results suggested that CDKN2A enhanced the SUMOylation of PIAS1 to reduce the expression of PIAS1. PRMT1 downregulated CFLAR by triggering its ubiquitination, while CFLAR repressed phosphorylation of JNK. The in vitro and in vivo results indicated that CDKN2A silencing prevented lipogenesis and fat accumulation by impairing the PRMT1-dependent ubiquitination of CFLAR and blocking the phosphorylation of JNK. Taken together, the central observations of our study demonstrate that targeting CDKN2A contributes to the suppression of lipogenesis and fat accumulation in fatty liver disease. The findings of our study highlight the potential of CDKN2A as a promising target against fatty liver.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Hepatobiliary Surgery, the Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Huiqing Wen
- Department of Hepatobiliary Surgery, the Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Bangjian Peng
- Department of Hepatobiliary Surgery, the Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Fanhong Zeng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
13
|
Huang L, Yu X, Jiang Z, Zeng P. Novel Autophagy-Related Gene Signature Investigation for Patients With Oral Squamous Cell Carcinoma. Front Genet 2021; 12:673319. [PMID: 34220946 PMCID: PMC8248343 DOI: 10.3389/fgene.2021.673319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022] Open
Abstract
The correlation between autophagy defects and oral squamous cell carcinoma (OSCC) has been previously studied, but only based on a limited number of autophagy-related genes in cell lines or animal models. The aim of the present study was to analyze differentially expressed autophagy-related genes through The Cancer Genome Atlas (TCGA) database to explore enriched pathways and potential biological function. Based on TCGA database, a signature composed of four autophagy-related genes (CDKN2A, NKX2-3, NRG3, and FADD) was established by using multivariate Cox regression models and two Gene Expression Omnibus datasets were applied for external validation. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to study the function of autophagy-related genes and their pathways. The most significant GO and KEGG pathways were enriched in several key pathways that were related to the progression of autophagy and OSCC. Furthermore, a prognostic risk score was constructed based on the four genes; patients were then divided into two groups (i.e., high risk and low risk) in terms of the median of risk score. Prognosis of the two groups and results showed that patients at the low-risk group had a much better prognosis than those at the high-risk group, regardless of whether they were in the training datasets or validation datasets. Multivariate Cox regression results indicated that the risk score of the autophagy-related gene signatures could greatly predict the prognosis of patients after controlling for several clinical covariates. The findings of the present study revealed that autophagy-related gene signatures play an important role in OSCC and are potential prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Lihong Huang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinghao Yu
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Zhou Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Ping Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China.,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Park J, Shin S, Liu L, Jahan I, Ong SG, Xu P, Berry DC, Jiang Y. Progenitor-like characteristics in a subgroup of UCP1+ cells within white adipose tissue. Dev Cell 2021; 56:985-999.e4. [PMID: 33711247 DOI: 10.1016/j.devcel.2021.02.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/17/2020] [Accepted: 02/16/2021] [Indexed: 12/19/2022]
Abstract
Thermogenic beige fat found in white adipose tissue is a potential therapeutic target to curb the global obesity and diabetes epidemic. However, these inducible thermogenic beige adipocytes have been thought to be short-lived and to rapidly convert to "white-like" adipocytes after discontinuing stimuli. In this study, using effective labeling techniques and genetic mouse tools, we demonstrate that a subset of UCP1+ cells that exist within white adipose tissue are able to self-divide and contribute to new beige adipocyte recruitment in response to β3 stimuli. When these cells are depleted or their adipogenic capability is blocked, β3-induced beige adipocyte formation is impaired. We also identify a cell-cycle machinery of p21 and CDKN2A as a molecular basis of beige adipocyte regulation. Collectively, our findings provide new insights into the cellular and molecular mechanisms of beige adipocyte regulation and potential therapeutic opportunities to induce the beige phenotype and treat metabolic disease.
Collapse
Affiliation(s)
- Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunhye Shin
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lifeng Liu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Iffat Jahan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Daniel C Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
15
|
Kahoul Y, Oger F, Montaigne J, Froguel P, Breton C, Annicotte JS. Emerging Roles for the INK4a/ARF ( CDKN2A) Locus in Adipose Tissue: Implications for Obesity and Type 2 Diabetes. Biomolecules 2020; 10:biom10091350. [PMID: 32971832 PMCID: PMC7563355 DOI: 10.3390/biom10091350] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022] Open
Abstract
Besides its role as a cell cycle and proliferation regulator, the INK4a/ARF (CDKN2A) locus and its associated pathways are thought to play additional functions in the control of energy homeostasis. Genome-wide association studies in humans and rodents have revealed that single nucleotide polymorphisms in this locus are risk factors for obesity and related metabolic diseases including cardiovascular complications and type-2 diabetes (T2D). Recent studies showed that both p16INK4a-CDK4-E2F1/pRB and p19ARF-P53 (p14ARF in humans) related pathways regulate adipose tissue (AT) physiology and adipocyte functions such as lipid storage, inflammation, oxidative activity, and cellular plasticity (browning). Targeting these metabolic pathways in AT emerged as a new putative therapy to alleviate the effects of obesity and prevent T2D. This review aims to provide an overview of the literature linking the INK4a/ARF locus with AT functions, focusing on its mechanisms of action in the regulation of energy homeostasis.
Collapse
|
16
|
Vincent EE, Yaghootkar H. Using genetics to decipher the link between type 2 diabetes and cancer: shared aetiology or downstream consequence? Diabetologia 2020; 63:1706-1717. [PMID: 32705315 PMCID: PMC7406536 DOI: 10.1007/s00125-020-05228-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Recent developments in the field of genetics have accelerated our understanding of the aetiology of complex diseases. Type 2 diabetes mellitus and cancer are no exception, with large-scale genome-wide association studies (GWAS) facilitating exploration of the underlying pathology. Here, we discuss how genetics studies can be used to investigate the relationship between these complex diseases. Observational epidemiological studies consistently report that people with type 2 diabetes have a higher risk of several types of cancer. Indeed, type 2 diabetes and cancer share many common risk factors, such as obesity, ageing, poor diet and low levels of physical activity. However, questions remain regarding the biological mechanisms that link these two diseases. Large-scale GWAS of type 2 diabetes and cancer allow us to consider the evidence for shared genetic architecture. Several shared susceptibility genes have been identified, yet tissue specificity and direction of effect must be taken into account when considering common genetic aetiology. We also consider how GWAS, and associated techniques such as Mendelian randomisation, allow us to dissect the link between the two diseases and address questions such as 'Does type 2 diabetes cause cancer or is the increased risk observed driven by higher adiposity or another associated metabolic feature?' Graphical abstract.
Collapse
Affiliation(s)
- Emma E Vincent
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
- School of Cellular and Molecular Medicine, Biomedical Science Building, University of Bristol, Bristol, BS8 1TW, UK.
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, UK
- School of Life Sciences, College of Liberal Arts and Science, University of Westminster, London, UK
- Division of Medical Sciences, Department of Health Sciences, Luleå University of Technology, Luleå, Sweden
| |
Collapse
|
17
|
Luan Y, Zhang W, Xie J, Mao J. CDKN2A inhibits cell proliferation and invasion in cervical cancer through LDHA-mediated AKT/mTOR pathway. Clin Transl Oncol 2020; 23:222-228. [DOI: 10.1007/s12094-020-02409-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022]
|
18
|
Abstract
Life expectancy has increased substantially over the last few decades, leading to a worldwide increase in the prevalence and burden of aging-associated diseases. Recent evidence has proven that cellular senescence contributes substantially to the development of these disorders. Cellular senescence is a state of cell cycle arrest with suppressed apoptosis and concomitant secretion of multiple bioactive factors (the senescence-associated secretory phenotype-SASP) that plays a physiological role in embryonic development and healing processes. However, DNA damage and oxidative stress that occur during aging cause the accumulation of senescent cells, which through their SASP bring about deleterious effects on multiple organ and systemic functions. Ablation of senescent cells through genetic or pharmacological means leads to improved life span and health span in animal models, and preliminary evidence suggests it may also have a positive impact on human health. Thus, strategies to reduce or eliminate the burden of senescent cells or their products have the potential to impact multiple clinical outcomes with a single intervention. In this review, we touch upon the basics of cell senescence and summarize the current state of development of therapies against cell senescence for human use.
Collapse
|
19
|
Wu C, Yang P, Liu B, Tang Y. Is there a CDKN2A-centric network in pancreatic ductal adenocarcinoma? Onco Targets Ther 2020; 13:2551-2562. [PMID: 32273725 PMCID: PMC7108878 DOI: 10.2147/ott.s232464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has a high mortality rate and its incidence has risen rapidly in recent years. Meanwhile, the diagnosis and treatment of this cancer remain challenging. Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, but, currently, no sufficiently effective modalities for its treatment exist. The early diagnosis rate of pancreatic cancer is low and most patients have reached an advanced stage at the time of diagnosis. PDAC evolves from precancerous lesions and is highly aggressive and metastatic. It is essential to understand how the disease progresses and metastasizes. CDKN2A mutations are very common in PDAC. Therefore, here, we have performed a literature review and discuss the role of CDKN2A and some related genes in the development of PDAC, as well as the basis of gene targeting with a correlation coefficient of CDKN2A above 0.9 on the STRING website. It is noteworthy that the interaction of CDKN2A with each gene has been reported in the literature. The role of these genes and CDKN2A in PDAC may provide new directions that will advance the current knowledge base and treatment options since cancer progression is realized through interactions among cells. Our findings provide new insights into the treatment of PADC that can, to some extent, improve the diagnosis rate and quality of life of patients.
Collapse
Affiliation(s)
- Chu Wu
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Ping Yang
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Bingxue Liu
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yunlian Tang
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
20
|
Ingelsson E, McCarthy MI. Human Genetics of Obesity and Type 2 Diabetes Mellitus: Past, Present, and Future. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002090. [PMID: 29899044 DOI: 10.1161/circgen.118.002090] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Type 2 diabetes mellitus (T2D) and obesity already represent 2 of the most prominent risk factors for cardiovascular disease, and are destined to increase in importance given the global changes in lifestyle. Ten years have passed since the first round of genome-wide association studies for T2D and obesity. During this decade, we have witnessed remarkable developments in human genetics. We have graduated from the despair of candidate gene-based studies that generated few consistently replicated genotype-phenotype associations, to the excitement of an exponential harvest of loci robustly associated with medical outcomes through ever larger genome-wide association study meta-analyses. As well as discovering hundreds of loci, genome-wide association studies have provided transformative insights into the genetic architecture of T2D and other complex traits, highlighting the extent of polygenicity and the tiny effect sizes of many common risk alleles. Genome-wide association studies have also provided a critical starting point for discovering new biology relevant to these traits. Expectations are high that these discoveries will foster development of more effective strategies for intervention, through optimization of precision medicine approaches. In this article, we review current knowledge and provide suggestions for the next steps in genetic research for T2D and obesity. We focus on four areas relevant to precision medicine: genetic architecture, pharmacogenetics and other gene-environment interactions, mechanistic inference, and drug development. As we describe, the genetic architecture of complex traits has major implications for the prospects of precision medicine, rendering some anticipated approaches decidedly unrealistic. We highlight obstacles to the translation of human genetic findings into mechanism inference but are optimistic that, as these are overcome, there is untapped potential for novel drugs and more effective strategies for treating and preventing T2D and obesity.
Collapse
Affiliation(s)
- Erik Ingelsson
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA (E.I.) .,Stanford Cardiovascular Institute, Stanford University, CA (E.I.)
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics (M.I.M.).,Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, United Kingdom (M.I.M.).,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, United Kingdom (M.I.M.)
| |
Collapse
|
21
|
Guo T, Li B, Gu C, Chen X, Han M, Liu X, Xu C. GCN-5/PGC-1α signaling is activated and associated with metabolism in cyclin E1-driven ovarian cancer. Aging (Albany NY) 2019; 11:4890-4899. [PMID: 31313989 PMCID: PMC6682509 DOI: 10.18632/aging.102082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
Abstract
AIM We previously found Cyclin E1-driven high grade serous ovarian cancer (HGSOC) showed metabolic shift. In this study, we aimed to elucidate signaling pathway therein. METHODS In silico reproduction of TCGA ovarian cancer dataset and pathway analysis were performed. Candidate metabolic pathway was validated using in vitro and in vivo assays. RESULTS We found CCNE1-amplified HGSOC showed significant metabolic alteration besides canonical cell cycle control. Using CCNE1-amplified OVCAR-3 and A2780 OvCa cells, we found that knockdown of CDK2 and GCN5 resulted in decreased G6PC and increased PGC-1α level, and that both genetic and pharmaceutical (MB-3) inhibition of GCN5 resulted in significant decrease in acetylation of PGC-1α. Silencing of CDK2 also resulted in significant decrease in acetylation of both PGC-1α and Rb. GCN5-KD significantly decreased glucose uptake, increased lactate production and decreased SDH activity. Western blots showed hierarchy of the elements indicating Cyclin E1-CDK2/GCN5/PGC-1α regulatory axis from up- to down- stream. Inhibitory effect of Dinaciclib was similar to that of GCN5 silencing, whereas combination therapy further inhibited cell proliferation significantly. Similar findings were noted also in cell cycle arrest, apoptosis, invasion, migration and colony formation assays. Xenograft experiments showed that GCN5-KD alone did not alter tumor growth yet combination therapy of Dinaciclib and GCN5-KD conferred significant inhibition of tumor growth compared with either therapy alone with no toxicity observed. CONCLUSION GCN-5/PGC-1α signaling is activated and associated with metabolism in Cyclin E1-driven HGSOC. Targeting GCN5 hold promise to augment current CDK2-targeting strategy and further studies are warranted for clinical translation.
Collapse
Affiliation(s)
- Ting Guo
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Bin Li
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Chao Gu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Xiuying Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Mengxin Han
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Xiaocheng Liu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Congjian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| |
Collapse
|
22
|
Zhao Y, Yang J, Liu Y, Fan J, Yang H. HSV-2-encoded miRNA-H4 Regulates Cell Cycle Progression and Act-D-induced Apoptosis in HeLa Cells by Targeting CDKL2 and CDKN2A. Virol Sin 2019; 34:278-286. [PMID: 30953292 DOI: 10.1007/s12250-019-00101-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/25/2019] [Indexed: 11/30/2022] Open
Abstract
MicroRNAs (miRNAs) encoded by latency-associated transcript are associated with both latent and acute stages of herpes simplex virus 2 (HSV-2) infection. In this study, miRNA-H4-5p and miRNA-H4-3p were ectopically expressed in HeLa cells to explore potential cellular targets of viral miRNAs and demonstrate their potential biological functions. The results showed that miRNA-H4-5p could reverse apoptosis induced by actinomycin D (Act-D) and promote cell cycle progression, but miRNA-H4-3p had no such obvious functions. Bioinformatics analysis, luciferase report assay, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and Western blotting demonstrated that miRNA-H4-5p could bind to the 3'-untranslated region (UTR) of cyclin-dependent kinase inhibitor 2A (CDKN2A) and cyclin-dependent kinase-like 2 (CDKL2) to negatively regulate their expression. We verified that these two targeted genes were associated with cell apoptosis and cell cycle. Furthermore, in HeLa cells infected with HSV-2, we detected significantly reduced expression of CDKN2A and CDKL2 and demonstrated the negative regulation effect of miRNA-H4-5p on these two target genes. Our findings show that viral miRNAs play a vital role in regulating the expression of the host's cellular genes that participate in cell apoptosis and progression to reshape the cellular environment in response to HSV-2 infection, providing further information on the roles of encoded herpesvirus miRNAs in pathogen-host interaction.
Collapse
Affiliation(s)
- Yang Zhao
- Guangzhou School of Clinical Medicine, Southern Medical University, Guangzhou, 510010, China
| | - Jingjing Yang
- Guangzhou School of Clinical Medicine, Southern Medical University, Guangzhou, 510010, China
| | - Yan Liu
- Department of Dermatology, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
| | - Jianyong Fan
- Department of Dermatology, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China
| | - Huilan Yang
- Guangzhou School of Clinical Medicine, Southern Medical University, Guangzhou, 510010, China. .,Department of Dermatology, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, China.
| |
Collapse
|
23
|
Benzel J, Fendrich V. Familial Pancreatic Cancer. Oncol Res Treat 2018; 41:611-618. [PMID: 30269130 DOI: 10.1159/000493473] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Abstract
Familial pancreatic cancer accounts for 10% of all patients with pancreatic cancer. Because the 5-year survival rate of pancreatic cancer is only 7%, screening programs for high-risk individuals are essential and might be advantageous. Pancreatic ductal adenocarcinoma mostly shows symptoms at an advanced state and treatment is not efficient enough to cure most patients. People with hereditary tumor syndromes or their affected relatives can also be included in such screening programs. Besides the collection of data to investigate the background of the disease, these screening programs aim to diagnose and treat precursor lesions so that more dangerous, invasive lesions are prevented. These precursor lesions can be pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm, and mucinous cystic neoplasm. This review summarizes the latest knowledge of pancreatic screening programs, shows the procedure of pancreatic cancer screening, and gives an overview of current guidelines.
Collapse
|
24
|
Rabhi N, Hannou SA, Gromada X, Salas E, Yao X, Oger F, Carney C, Lopez-Mejia IC, Durand E, Rabearivelo I, Bonnefond A, Caron E, Fajas L, Dani C, Froguel P, Annicotte JS. Cdkn2a deficiency promotes adipose tissue browning. Mol Metab 2017; 8:65-76. [PMID: 29237539 PMCID: PMC5985036 DOI: 10.1016/j.molmet.2017.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/23/2017] [Indexed: 01/01/2023] Open
Abstract
Objectives Genome-wide association studies have reported that DNA polymorphisms at the CDKN2A locus modulate fasting glucose in human and contribute to type 2 diabetes (T2D) risk. Yet the causal relationship between this gene and defective energy homeostasis remains elusive. Here we sought to understand the contribution of Cdkn2a to metabolic homeostasis. Methods We first analyzed glucose and energy homeostasis from Cdkn2a-deficient mice subjected to normal or high fat diets. Subsequently Cdkn2a-deficient primary adipose cells and human-induced pluripotent stem differentiated into adipocytes were further characterized for their capacity to promote browning of adipose tissue. Finally CDKN2A levels were studied in adipocytes from lean and obese patients. Results We report that Cdkn2a deficiency protects mice against high fat diet-induced obesity, increases energy expenditure and modulates adaptive thermogenesis, in addition to improving insulin sensitivity. Disruption of Cdkn2a associates with increased expression of brown-like/beige fat markers in inguinal adipose tissue and enhances respiration in primary adipose cells. Kinase activity profiling and RNA-sequencing analysis of primary adipose cells further demonstrate that Cdkn2a modulates gene networks involved in energy production and lipid metabolism, through the activation of the Protein Kinase A (PKA), PKG, PPARGC1A and PRDM16 signaling pathways, key regulators of adipocyte beiging. Importantly, CDKN2A expression is increased in adipocytes from obese compared to lean subjects. Moreover silencing CDKN2A expression during human-induced pluripotent stem cells adipogenic differentiation promoted UCP1 expression. Conclusion Our results offer novel insight into brown/beige adipocyte functions, which has recently emerged as an attractive therapeutic strategy for obesity and T2D. Modulating Cdkn2a-regulated signaling cascades may be of interest for the treatment of metabolic disorders. Cdkn2a deficiency protects mice against high fat diet-induced obesity. Cdkn2a modulates brown-like/beige fat gene networks involved in energy production and lipid metabolism. Increased CDKN2A expression in human obese adipocytes. Increased UCP1 levels in adipocytes differentiated from CDKN2A-silenced hiPS cells.
Collapse
Affiliation(s)
- Nabil Rabhi
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Sarah Anissa Hannou
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Xavier Gromada
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Elisabet Salas
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Xi Yao
- Université Côte d'Azur, CNRS, INSERM, iBV, Faculté de Médecine, F-06107 Nice Cedex 2, France
| | - Frédérik Oger
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Charlène Carney
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Isabel C Lopez-Mejia
- Center for Integrative Genomics, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Emmanuelle Durand
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Iandry Rabearivelo
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Amélie Bonnefond
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France
| | - Emilie Caron
- INSERM, UMR S-1172, Development and Plasticity of Postnatal Brain, F-59000 Lille, France
| | - Lluis Fajas
- Center for Integrative Genomics, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Christian Dani
- Université Côte d'Azur, CNRS, INSERM, iBV, Faculté de Médecine, F-06107 Nice Cedex 2, France
| | - Philippe Froguel
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France; Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London W12 0NN, UK.
| | - Jean-Sébastien Annicotte
- Lille University, UMR 8199 - EGID, F-59000 Lille, France; CNRS, UMR 8199, F-59000 Lille, France; Institut Pasteur de Lille, F-59000 Lille, France.
| |
Collapse
|
25
|
Sharma A, Vella A. Obstacles to Translating Genotype-Phenotype Correlates in Metabolic Disease. Physiology (Bethesda) 2017; 32:42-50. [PMID: 27927804 DOI: 10.1152/physiol.00009.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Type 2 diabetes mellitus is a polygenic disease with a variable phenotype. Many genetic associations have been described; however, understanding their underlying pathophysiological role in Type 2 diabetes mellitus is important for development of future therapeutic targets. Here, we review the physiological mechanisms of diabetes-associated variants that affect glycemia.
Collapse
Affiliation(s)
- Anu Sharma
- Department of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, Minnesota
| | - Adrian Vella
- Department of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Deciphering the mechanisms of type 2 diabetes (T2DM) risk loci can greatly inform on disease pathology. This review discusses current knowledge of mechanisms through which genetic variants influence T2DM risk and considerations for future studies. RECENT FINDINGS Over 100 T2DM risk loci to date have been identified. Candidate causal variants at risk loci map predominantly to non-coding sequence. Physiological, epigenomic and gene expression data suggest that variants at many known T2DM risk loci affect pancreatic islet regulation, although variants at other loci also affect protein function and regulatory processes in adipose, pre-adipose, liver, skeletal muscle and brain. The effects of T2DM variants on regulatory activity in these tissues appear largely, but not exclusively, due to altered transcription factor binding. Putative target genes of T2DM variants have been defined at an increasing number of loci and some, such as FTO, may entail several genes and multiple tissues. Gene networks in islets and adipocytes have been implicated in T2DM risk, although the molecular pathways of risk genes remain largely undefined. Efforts to fully define the mechanisms of T2DM risk loci are just beginning. Continued identification of risk mechanisms will benefit from combining genetic fine-mapping with detailed phenotypic association data, high-throughput epigenomics data from diabetes-relevant tissue, functional screening of candidate genes and genome editing of cellular and animal models.
Collapse
Affiliation(s)
- Kyle J Gaulton
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA.
| |
Collapse
|
27
|
Padhi SS, Roy S, Kar M, Saha A, Roy S, Adhya A, Baisakh M, Banerjee B. Role of CDKN2A/p16 expression in the prognostication of oral squamous cell carcinoma. Oral Oncol 2017; 73:27-35. [PMID: 28939073 DOI: 10.1016/j.oraloncology.2017.07.030] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/11/2017] [Accepted: 07/28/2017] [Indexed: 01/11/2023]
Abstract
OBJECTIVE CDKN2A/p16 is a known tumor suppressor gene with a homologous deletion in Oral Squamous cell carcinoma. CDKN2A/p16 is found to be inactivated in a broad spectrum of solid tumors and in more than 80% of OSCC. Molecular alteration of CDKN2A/p16 in progression of OSCC can pose an important tool for the prognosis of squamous cell carcinoma. MATERIAL AND METHOD Systematic network analysis was carried out to obtain involvement of CDKN2A/p16 in oral cancer by polysearch and FunDO. In the present study we have screened 104 OSCC patients from eastern region of India for CDKN2A/p16 expression in recurrent and non-recurrent OSCC. The observation was validated by Comparative Genomic Hybridisation and Next generation sequencing in recurrent cases. RESULT Systematic analysis revealed direct involvement of CDKN2A/p16 in oral cancer. There was a consistent downregulated expression of CDKN2A/p16 in the recurrent cases. The gene expression study confirmed a >5-fold downregulation of CDKN2A/p16 in recurrent tumors as compared to non-recurrent ones. Array CGH analysis revealed a copy number deletion in the recurrent case. Furthermore, next generation sequencing validated deletion of CDKN2A/p16 and reported it asa common variant with a nonsense mutation having stop /loss of function of the gene in recurrent cases. Recurrent cases with deleted CDKN2A/p16 expression had poor prognosis and low survival rate. CONCLUSION CDKN2A/p16 frequently alters in oral cancer progression with a deletion/loss of function in the recurrent cases displaying its role in aiding several molecular events for the malignant transformations occurring throughout disease progression.
Collapse
Affiliation(s)
- Swati Shree Padhi
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha 751024, India
| | - Souvick Roy
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha 751024, India
| | - Madhabananda Kar
- Department of Surgical Oncology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, Odisha 751016, India
| | - Arka Saha
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha 751024, India
| | - Shomereeta Roy
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha 751024, India
| | - Amit Adhya
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha 751024, India
| | - Manas Baisakh
- Department of Pathology, Apollo Hospitals, Bhubaneswar, Odisha 751004, India
| | - Birendranath Banerjee
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
28
|
He S, Sharpless NE. Senescence in Health and Disease. Cell 2017; 169:1000-1011. [PMID: 28575665 DOI: 10.1016/j.cell.2017.05.015] [Citation(s) in RCA: 1086] [Impact Index Per Article: 155.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 02/07/2023]
Abstract
Many cellular stresses activate senescence, a persistent hyporeplicative state characterized in part by expression of the p16INK4a cell-cycle inhibitor. Senescent cell production occurs throughout life and plays beneficial roles in a variety of physiological and pathological processes including embryogenesis, wound healing, host immunity, and tumor suppression. Meanwhile, the steady accumulation of senescent cells with age also has adverse consequences. These non-proliferating cells occupy key cellular niches and elaborate pro-inflammatory cytokines, contributing to aging-related diseases and morbidity. This model suggests that the abundance of senescent cells in vivo predicts "molecular," as opposed to chronologic, age and that senescent cell clearance may mitigate aging-associated pathology.
Collapse
Affiliation(s)
- Shenghui He
- Departments of Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA; The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA
| | - Norman E Sharpless
- Departments of Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA; The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7295, USA.
| |
Collapse
|
29
|
Ndiaye FK, Ortalli A, Canouil M, Huyvaert M, Salazar-Cardozo C, Lecoeur C, Verbanck M, Pawlowski V, Boutry R, Durand E, Rabearivelo I, Sand O, Marselli L, Kerr-Conte J, Chandra V, Scharfmann R, Poulain-Godefroy O, Marchetti P, Pattou F, Abderrahmani A, Froguel P, Bonnefond A. Expression and functional assessment of candidate type 2 diabetes susceptibility genes identify four new genes contributing to human insulin secretion. Mol Metab 2017; 6:459-470. [PMID: 28580277 PMCID: PMC5444093 DOI: 10.1016/j.molmet.2017.03.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 03/17/2017] [Accepted: 03/24/2017] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVES Genome-wide association studies (GWAS) have identified >100 loci independently contributing to type 2 diabetes (T2D) risk. However, translational implications for precision medicine and for the development of novel treatments have been disappointing, due to poor knowledge of how these loci impact T2D pathophysiology. Here, we aimed to measure the expression of genes located nearby T2D associated signals and to assess their effect on insulin secretion from pancreatic beta cells. METHODS The expression of 104 candidate T2D susceptibility genes was measured in a human multi-tissue panel, through PCR-free expression assay. The effects of the knockdown of beta-cell enriched genes were next investigated on insulin secretion from the human EndoC-βH1 beta-cell line. Finally, we performed RNA-sequencing (RNA-seq) so as to assess the pathways affected by the knockdown of the new genes impacting insulin secretion from EndoC-βH1, and we analyzed the expression of the new genes in mouse models with altered pancreatic beta-cell function. RESULTS We found that the candidate T2D susceptibility genes' expression is significantly enriched in pancreatic beta cells obtained by laser capture microdissection or sorted by flow cytometry and in EndoC-βH1 cells, but not in insulin sensitive tissues. Furthermore, the knockdown of seven T2D-susceptibility genes (CDKN2A, GCK, HNF4A, KCNK16, SLC30A8, TBC1D4, and TCF19) with already known expression and/or function in beta cells changed insulin secretion, supporting our functional approach. We showed first evidence for a role in insulin secretion of four candidate T2D-susceptibility genes (PRC1, SRR, ZFAND3, and ZFAND6) with no previous knowledge of presence and function in beta cells. RNA-seq in EndoC-βH1 cells with decreased expression of PRC1, SRR, ZFAND6, or ZFAND3 identified specific gene networks related to T2D pathophysiology. Finally, a positive correlation between the expression of Ins2 and the expression of Prc1, Srr, Zfand6, and Zfand3 was found in mouse pancreatic islets with altered beta-cell function. CONCLUSIONS This study showed the ability of post-GWAS functional studies to identify new genes and pathways involved in human pancreatic beta-cell function and in T2D pathophysiology.
Collapse
Affiliation(s)
- Fatou K. Ndiaye
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Ana Ortalli
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Mickaël Canouil
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Marlène Huyvaert
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Clara Salazar-Cardozo
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Cécile Lecoeur
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Marie Verbanck
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Valérie Pawlowski
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Raphaël Boutry
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Emmanuelle Durand
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Iandry Rabearivelo
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Olivier Sand
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56100 Pisa, Italy
| | - Julie Kerr-Conte
- Inserm U1190, EGID, CHU Lille, University of Lille, 59000 Lille, France
| | - Vikash Chandra
- Inserm U1016, Institut Cochin, Faculté de Médecine, Paris Descartes University, Sorbonne Paris Cité, 75014 Paris, France
| | - Raphaël Scharfmann
- Inserm U1016, Institut Cochin, Faculté de Médecine, Paris Descartes University, Sorbonne Paris Cité, 75014 Paris, France
| | - Odile Poulain-Godefroy
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56100 Pisa, Italy
| | - François Pattou
- Inserm U1190, EGID, CHU Lille, University of Lille, 59000 Lille, France
| | - Amar Abderrahmani
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
- Department of Genomics of Common Disease, Imperial College London, W12 0NN London, United Kingdom
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
- Department of Genomics of Common Disease, Imperial College London, W12 0NN London, United Kingdom
| | - Amélie Bonnefond
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000 Lille, France
- Department of Genomics of Common Disease, Imperial College London, W12 0NN London, United Kingdom
| |
Collapse
|
30
|
Thomsen SK, Ceroni A, van de Bunt M, Burrows C, Barrett A, Scharfmann R, Ebner D, McCarthy MI, Gloyn AL. Systematic Functional Characterization of Candidate Causal Genes for Type 2 Diabetes Risk Variants. Diabetes 2016; 65:3805-3811. [PMID: 27554474 PMCID: PMC5402869 DOI: 10.2337/db16-0361] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/18/2016] [Indexed: 12/30/2022]
Abstract
Most genetic association signals for type 2 diabetes risk are located in noncoding regions of the genome, hindering translation into molecular mechanisms. Physiological studies have shown a majority of disease-associated variants to exert their effects through pancreatic islet dysfunction. Systematically characterizing the role of regional transcripts in β-cell function could identify the underlying disease-causing genes, but large-scale studies in human cellular models have previously been impractical. We developed a robust and scalable strategy based on arrayed gene silencing in the human β-cell line EndoC-βH1. In a screen of 300 positional candidates selected from 75 type 2 diabetes regions, each gene was assayed for effects on multiple disease-relevant phenotypes, including insulin secretion and cellular proliferation. We identified a total of 45 genes involved in β-cell function, pointing to possible causal mechanisms at 37 disease-associated loci. The results showed a strong enrichment for genes implicated in monogenic diabetes. Selected effects were validated in a follow-up study, including several genes (ARL15, ZMIZ1, and THADA) with previously unknown or poorly described roles in β-cell biology. We have demonstrated the feasibility of systematic functional screening in a human β-cell model and successfully prioritized plausible disease-causing genes at more than half of the regions investigated.
Collapse
Affiliation(s)
- Soren K Thomsen
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Alessandro Ceroni
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Martijn van de Bunt
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, U.K
| | - Carla Burrows
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Amy Barrett
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Daniel Ebner
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, U.K
- National Institute for Health Research Oxford Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K.
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, U.K
- National Institute for Health Research Oxford Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| |
Collapse
|
31
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
32
|
Kong Y, Sharma RB, Nwosu BU, Alonso LC. Islet biology, the CDKN2A/B locus and type 2 diabetes risk. Diabetologia 2016; 59:1579-93. [PMID: 27155872 PMCID: PMC4930689 DOI: 10.1007/s00125-016-3967-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes, fuelled by the obesity epidemic, is an escalating worldwide cause of personal hardship and public cost. Diabetes incidence increases with age, and many studies link the classic senescence and ageing protein p16(INK4A) to diabetes pathophysiology via pancreatic islet biology. Genome-wide association studies (GWASs) have unequivocally linked the CDKN2A/B locus, which encodes p16 inhibitor of cyclin-dependent kinase (p16(INK4A)) and three other gene products, p14 alternate reading frame (p14(ARF)), p15(INK4B) and antisense non-coding RNA in the INK4 locus (ANRIL), with human diabetes risk. However, the mechanism by which the CDKN2A/B locus influences diabetes risk remains uncertain. Here, we weigh the evidence that CDKN2A/B polymorphisms impact metabolic health via islet biology vs effects in other tissues. Structured in a bedside-to-bench-to-bedside approach, we begin with a summary of the evidence that the CDKN2A/B locus impacts diabetes risk and a brief review of the basic biology of CDKN2A/B gene products. The main emphasis of this work is an in-depth look at the nuanced roles that CDKN2A/B gene products and related proteins play in the regulation of beta cell mass, proliferation and insulin secretory function, as well as roles in other metabolic tissues. We finish with a synthesis of basic biology and clinical observations, incorporating human physiology data. We conclude that it is likely that the CDKN2A/B locus influences diabetes risk through both islet and non-islet mechanisms.
Collapse
Affiliation(s)
- Yahui Kong
- AS7-2047, Division of Diabetes, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Rohit B Sharma
- AS7-2047, Division of Diabetes, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Benjamin U Nwosu
- Division of Endocrinology, Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Laura C Alonso
- AS7-2047, Division of Diabetes, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
33
|
Abstract
As with other complex diseases, unbiased association studies followed by physiological and experimental characterization have for years formed a paradigm for identifying genes or processes of relevance to type 2 diabetes mellitus (T2D). Recent large-scale common and rare variant genome-wide association studies (GWAS) suggest that substantially larger association studies are needed to identify most T2D loci in the population. To hasten clinical translation of genetic discoveries, new paradigms are also required to aid specialized investigation of nascent hypotheses. We argue for an integrated T2D knowledgebase, designed for a worldwide community to access aggregated large-scale genetic data sets, as one paradigm to catalyse convergence of these efforts.
Collapse
|
34
|
Saunders D, Powers AC. Replicative capacity of β-cells and type 1 diabetes. J Autoimmun 2016; 71:59-68. [PMID: 27133598 DOI: 10.1016/j.jaut.2016.03.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 03/28/2016] [Indexed: 12/16/2022]
Abstract
Efforts to restore β-cell number or mass in type 1 diabetes (T1D) must combine an intervention to stimulate proliferation of remaining β-cells and an intervention to mitigate or control the β-cell-directed autoimmunity. This review highlights features of the β-cell, including it being part of a pancreatic islet, a mini-organ that is highly vascularized and highly innervated, and efforts to promote β-cell proliferation. In addition, the β-cell in T1D exists in a microenvironment with interactions and input from other islet cell types, extracellular matrix, vascular endothelial cells, neuronal projections, and immune cells, all of which likely influence the β-cell's capacity for replication. Physiologic β-cell proliferation occurs in human and rodents in the neonatal period and early in life, after which there is an age-dependent decline in β-cell proliferation, and also as part of the β-cell's compensatory response to the metabolic challenges of pregnancy and insulin resistance. This review reviews the molecular pathways involved in this β-cell proliferation and highlights recent work in two areas: 1) Investigators, using high-throughput screening to discover small molecules that promote human β-cell proliferation, are now focusing on the dual-specificity tyrosine-regulated kinase-1a and cell cycle-dependent kinase inhibitors CDKN2C/p18 or CDKN1A/p21as targets of compounds to stimulate adult human β-cell proliferation. 2) Local inflammation, macrophages, and the local β-cell microenvironment promote β-cell proliferation. Future efforts to harness the responsible mechanisms may lead to new approaches to promote β-cell proliferation in T1D.
Collapse
Affiliation(s)
- Diane Saunders
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, United States; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; VA Tennessee Valley Healthcare System, Nashville, TN, United States.
| |
Collapse
|
35
|
Thomsen SK, McCarthy MI, Gloyn AL. The Importance of Context: Uncovering Species- and Tissue-Specific Effects of Genetic Risk Variants for Type 2 Diabetes. Front Endocrinol (Lausanne) 2016; 7:112. [PMID: 27630614 PMCID: PMC5005446 DOI: 10.3389/fendo.2016.00112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 08/04/2016] [Indexed: 12/30/2022] Open
Affiliation(s)
- Soren K. Thomsen
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Anna L. Gloyn
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
- *Correspondence: Anna L. Gloyn,
| |
Collapse
|