1
|
Giroud J, Combémorel E, Pourtier A, Abbadie C, Pluquet O. Unraveling the functional and molecular interplay between cellular senescence and the unfolded protein response. Am J Physiol Cell Physiol 2025; 328:C1764-C1782. [PMID: 40257464 DOI: 10.1152/ajpcell.00091.2025] [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: 01/28/2025] [Revised: 02/12/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Senescence is a complex cellular state that can be considered as a stress response phenotype. A decade ago, we suggested the intricate connections between unfolded protein response (UPR) signaling and the development of the senescent phenotype. Over the past ten years, significant advances have been made in understanding the multifaceted role of the UPR in regulating cellular senescence, highlighting its contribution to biological processes such as oxidative stress and autophagy. In this updated review, we expand these interconnections with the benefit of new insights, and we suggest that targeting specific components of the UPR could provide novel therapeutic strategies to mitigate the deleterious effects of senescence, with significant implications for age-related pathologies and geroscience.
Collapse
Affiliation(s)
- Joëlle Giroud
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University of Lille, Lille, France
| | - Emilie Combémorel
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University of Lille, Lille, France
| | - Albin Pourtier
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University of Lille, Lille, France
| | - Corinne Abbadie
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University of Lille, Lille, France
| | - Olivier Pluquet
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University of Lille, Lille, France
| |
Collapse
|
2
|
MacDonald TL, Ryback B, Aparecida da Silva Pereira J, Wei S, Mendez B, Cai EP, Ishikawa Y, Arbeau M, Weir G, Bonner-Weir S, Kissler S, Yi P. Renalase inhibition defends against acute and chronic β cell stress by regulating cell metabolism. Mol Metab 2025; 95:102115. [PMID: 39988068 PMCID: PMC11981795 DOI: 10.1016/j.molmet.2025.102115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVE Renalase (Rnls) is annotated as an oxidase enzyme. It has been implicated in Type 1 diabetes (T1D) risk via genome-wide association studies (GWAS). We previously discovered through CRISPR screening and validation experiments that Rnls inhibition prevents or delays T1D in multiple mouse models of diabetes in vivo, and protects pancreatic β cells against autoimmune killing, ER and oxidative stress in vitro. The molecular biochemistry and functions of Rnls are largely uncharted. Here we studied the mechanisms of Rnls inhibition that underlie β cell protection during diabetogenic stress. METHODS Akita mice were treated with oral Pargyline (PG) in vivo to bind and inhibit Rnls, and pancreas or islets were harvested for β cell mass and β cell function analyses. Genetic and pharmacological tools were used to inhibit Rnls in β cell lines. RNA sequencing, metabolomics and metabolic function experiments were conducted in vitro in NIT-1 mouse β cell lines and human stem cell-derived β cells. RESULTS In vivo, PG improved glycemia and mildly preserved β cell mass and function in females. Genetic strategies to mutate (Rnlsmut) or knockout (Rnls KO) Rnls induced a robust metabolic shift towards glycolysis in both mouse and human β cell lines, in vitro. Stress protection was abolished when glycolysis was blocked with 2-deoxyglucose (2-DG). Pharmacological Rnls inhibition with PG did not strongly mimic these newly identified metabolic mechanisms. CONCLUSIONS Our work illustrates a role for Rnls in regulating cell metabolism. We show that inhibiting Rnls protects against chronic stress in vivo, and shields against acute stress in β cell lines in vitro by rewiring cell metabolism towards glycolysis.
Collapse
Affiliation(s)
- Tara L MacDonald
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Section for Immunobiology, Joslin Diabetes Center, Boston, USA; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Birgitta Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA
| | - Jéssica Aparecida da Silva Pereira
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Siying Wei
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bryhan Mendez
- Section for Immunobiology, Joslin Diabetes Center, Boston, USA
| | - Erica P Cai
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yuki Ishikawa
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Meagan Arbeau
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gordon Weir
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Susan Bonner-Weir
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
3
|
Yagan M, Najam S, Hu R, Wang Y, Dickerson MT, Dadi PK, Xu Y, Simmons AJ, Stein R, Adams CM, Jacobson DA, Lau KS, Liu Q, Gu G. Atf4 Protects Islet β-Cell Identity and Function Under Acute Glucose-Induced Stress but Promotes β-Cell Failure in the Presence of Free Fatty Acid. Diabetes 2025; 74:838-849. [PMID: 39899446 PMCID: PMC12015139 DOI: 10.2337/db24-0360] [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: 04/27/2024] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Glucolipotoxicity, caused by combined hyperglycemia and hyperlipidemia, results in β-cell failure and type 2 diabetes via cellular stress-related mechanisms. Activating transcription factor 4 (Atf4) is an essential effector of stress response. We show here that Atf4 expression in β-cells is minimally required for glucose homeostasis in juvenile and adolescent mice but it is needed for β-cell function during aging and under obesity-related metabolic stress. Henceforth, Atf4-deficient β-cells older than 2 months after birth display compromised secretory function under acute hyperglycemia. In contrast, they are resistant to acute free fatty acid-induced dysfunction and reduced production of several factors essential for β-cell identity. Atf4-deficient β-cells downregulate genes involved in protein translation. They also upregulate several lipid metabolism or signaling genes, likely contributing to their resistance to free fatty acid-induced dysfunction. These results suggest that Atf4 activation is required for β-cell identity and function under high glucose. But Atf4 activation paradoxically induces β-cell failure in high levels of free fatty acids. Different transcriptional targets of Atf4 could be manipulated to protect β-cells from metabolic stress-induced failure. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mahircan Yagan
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Sadia Najam
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Ruiying Hu
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Yu Wang
- Department of Biostatistics and Center for Quantitative Sciences, Vanderbilt Medical Center, Nashville, TN
| | - Mathew T. Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Prasanna K. Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Yanwen Xu
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Computational Systems Biology and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Alan J. Simmons
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Computational Systems Biology and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Christopher M. Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Ken S. Lau
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
- Center for Computational Systems Biology and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Qi Liu
- Department of Biostatistics and Center for Quantitative Sciences, Vanderbilt Medical Center, Nashville, TN
| | - Guoqiang Gu
- Department of Cell and Developmental Biology and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
4
|
Wu S, Zhao S, Hai L, Yang Z, Wang S, Cui D, Xie J. Macrophage polarization regulates the pathogenesis and progression of autoimmune diseases. Autoimmun Rev 2025; 24:103820. [PMID: 40268127 DOI: 10.1016/j.autrev.2025.103820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/28/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Macrophages are integral components of the innate immune system, present in nearly all tissues and organs throughout the body. They exhibit a high degree of plasticity and heterogeneity, participating in immune responses to maintain immune homeostasis. When the immune system loses tolerance, macrophages rapidly proliferate and polarize in response to various signaling pathways within a disrupted microenvironment. The direction of macrophage polarization can be regulated by a variety of factors, including transcription factors, non-coding RNAs, and metabolic reprogramming. Autoimmune diseases arise from the immune system's activation against host cells, with macrophage polarization playing a critical role in the pathogenesis of numerous chronic inflammatory and autoimmune conditions, such as rheumatoid arthritis, systemic lupus erythematosus, immune thrombocytopenic purpura, and type 1 diabetes. Consequently, elucidating the molecular mechanisms underlying macrophage development and function presents opportunities for the development of novel therapeutic targets. This review outlines the functions of macrophage polarization in prevalent autoimmune diseases and the underlying mechanisms involved. Furthermore, we discuss the immunotherapeutic potential of targeting macrophage polarization and highlight the characteristics and recent advancements of promising therapeutic targets. Our aim is to inspire further strategies to restore macrophage balance in preventing and treating autoimmune diseases.
Collapse
Affiliation(s)
- Siwen Wu
- Department of Blood Transfusion, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shubi Zhao
- Department of Critical Medicine, School of Medicine, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, China
| | - Lei Hai
- Department of Blood Transfusion, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyin Yang
- Department of Blood Transfusion, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shifen Wang
- Department of Blood Transfusion, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dawei Cui
- Department of Blood Transfusion, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jue Xie
- Department of Blood Transfusion, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Mitra J, Kodavati M, Dharmalingam P, Guerrero EN, Rao KS, Garruto RM, Hegde ML. Endogenous TDP-43 mislocalization in a novel knock-in mouse model reveals DNA repair impairment, inflammation, and neuronal senescence. Acta Neuropathol Commun 2025; 13:54. [PMID: 40057796 PMCID: PMC11889789 DOI: 10.1186/s40478-025-01962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 03/14/2025] Open
Abstract
TDP-43 mislocalization and aggregation are key pathological features of amyotrophic lateral sclerosis (ALS)- and frontotemporal dementia (FTD). However, existing transgenic hTDP-43 WT or ∆NLS-overexpression animal models primarily focus on late-stage TDP-43 proteinopathy. To complement these models and to study the early-stage motor neuron-specific pathology during pre-symptomatic phases of disease progression, we generated a new endogenous knock-in (KI) mouse model using a combination of CRISPR/Cas9 and FLEX Cre-switch strategy for the conditional expression of a mislocalized Tdp-43∆NLS variant of mouse Tdp-43. This variant is expressed either in the whole body (WB) or specifically in the motor neurons (MNs) in two distinct models. These mice exhibit loss of nuclear Tdp-43, with concomitant cytosolic accumulation and aggregation in targeted cells, leading to increased DNA double-strand breaks (DSBs), signs of inflammation, and associated cellular senescence. Notably, unlike WT Tdp-43, which functionally interacts with Xrcc4 and DNA Ligase 4, the key DSB repair proteins in the non-homologous end-joining (NHEJ) pathway, the Tdp-43∆NLS mutant sequesters them into cytosolic aggregates, exacerbating neuronal damage in mouse brain. The mutant mice also exhibit myogenic degeneration in hindlimb soleus muscles and distinct motor deficits, consistent with the characteristics of motor neuron disease (MND). Our findings reveal progressive degenerative mechanisms in motor neurons expressing endogenous Tdp-43∆NLS mutant, independent of Tdp-43 overexpression or other confounding factors. Thus, this unique Tdp-43 KI mouse model, which displays key molecular and phenotypic features of Tdp-43 proteinopathy, offers a significant opportunity to characterize the early-stage progression of MND further and also opens avenues for developing DNA repair-targeted approaches for treating TDP-43 pathology-linked neurodegenerative diseases.
Collapse
Affiliation(s)
- Joy Mitra
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA.
| | - Manohar Kodavati
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Prakash Dharmalingam
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Erika N Guerrero
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Gorgas Memorial Institute for Health Studies, Avenida Justo Arosemena y Calle 35, Panama City, Republic of Panama
- Sistema Nacional de Investigación, SENACYT, Panama City, Republic of Panama
| | - K S Rao
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation Deemed to Be University, Green Fields, Vaddeswaram, Andhra Pradesh, 522502, India
| | - Ralph M Garruto
- Department of Anthropology, Binghamton University, State University of New York, Binghamton, NY, 13902, USA
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY, 13902, USA
| | - Muralidhar L Hegde
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Neuroscience, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
6
|
Oguh AU, Haemmerle MW, Sen S, Rozo AV, Shrestha S, Cartailler JP, Fazelinia H, Ding H, Preza S, Yang J, Yang X, Sussel L, Alvarez-Dominguez JR, Doliba N, Spruce LA, Arrojo E Drigo R, Stoffers DA. E3 ligase substrate adaptor SPOP fine-tunes the UPR of pancreatic β cells. Genes Dev 2025; 39:261-279. [PMID: 39797759 PMCID: PMC11789638 DOI: 10.1101/gad.352010.124] [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: 06/10/2024] [Accepted: 11/07/2024] [Indexed: 01/13/2025]
Abstract
The Cullin-3 E3 ligase adaptor protein SPOP targets proteins for ubiquitination and proteasomal degradation. We previously established the β-cell transcription factor (TF) and human diabetes gene PDX1 as an SPOP substrate, suggesting a functional role for SPOP in the β cell. Here, we generated a β-cell-specific Spop deletion mouse strain (Spop βKO) and found that Spop is necessary to prevent aberrant basal insulin secretion and for maintaining glucose-stimulated insulin secretion through impacts on glycolysis and glucose-stimulated calcium flux. Integration of proteomic, TF-regulatory gene network, and biochemical analyses identified XBP1 as a functionally important SPOP substrate in pancreatic β cells. Furthermore, loss of SPOP strengthened the IRE1α-XBP1 axis of unfolded protein response (UPR) signaling. ER stress promoted proteasomal degradation of SPOP, supporting a model whereby SPOP fine-tunes XBP1 activation during the UPR. These results position SPOP as a regulator of β-cell function and proper UPR activation.
Collapse
Affiliation(s)
- Alexis U Oguh
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Matthew W Haemmerle
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Sabyasachi Sen
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Andrea V Rozo
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Shristi Shrestha
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Jean-Philippe Cartailler
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Hossein Fazelinia
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Hua Ding
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Sam Preza
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Juxiang Yang
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Xiaodun Yang
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Lori Sussel
- Department of Pediatrics and Cell and Developmental Biology, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Juan R Alvarez-Dominguez
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Nicolai Doliba
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Lynn A Spruce
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Rafael Arrojo E Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Doris A Stoffers
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA;
| |
Collapse
|
7
|
Bandesh K, Motakis E, Nargund S, Kursawe R, Selvam V, Bhuiyan RM, Eryilmaz GN, Krishnan SN, Spracklen CN, Ucar D, Stitzel ML. Single-cell decoding of human islet cell type-specific alterations in type 2 diabetes reveals converging genetic- and state-driven β -cell gene expression defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633590. [PMID: 39896672 PMCID: PMC11785113 DOI: 10.1101/2025.01.17.633590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Pancreatic islets maintain glucose homeostasis through coordinated action of their constituent endocrine and affiliate cell types and are central to type 2 diabetes (T2D) genetics and pathophysiology. Our understanding of robust human islet cell type-specific alterations in T2D remains limited. Here, we report comprehensive single cell transcriptome profiling of 245,878 human islet cells from a 48-donor cohort spanning non-diabetic (ND), pre-diabetic (PD), and T2D states, identifying 14 distinct cell types detected in every donor from each glycemic state. Cohort analysis reveals ~25-30% loss of functional beta cell mass in T2D vs. ND or PD donors resulting from (1) reduced total beta cell numbers/proportions and (2) reciprocal loss of 'high function' and gain of senescent β -cell subpopulations. We identify in T2D β -cells 511 differentially expressed genes (DEGs), including new (66.5%) and validated genes (e.g., FXYD2, SLC2A2, SYT1), and significant neuronal transmission and vitamin A metabolism pathway alterations. Importantly, we demonstrate newly identified DEG roles in human β -cell viability and/or insulin secretion and link 47 DEGs to diabetes-relevant phenotypes in knockout mice, implicating them as potential causal islet dysfunction genes. Additionally, we nominate as candidate T2D causal genes and therapeutic targets 27 DEGs for which T2D genetic risk variants (GWAS SNPs) and pathophysiology (T2D vs. ND) exert concordant expression effects. We provide this freely accessible atlas for data exploration, analysis, and hypothesis testing. Together, this study provides new genomic resources for and insights into T2D pathophysiology and human islet dysfunction.
Collapse
Affiliation(s)
- Khushdeep Bandesh
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Efthymios Motakis
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Siddhi Nargund
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
| | - Giray Naim Eryilmaz
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Sai Nivedita Krishnan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
| | - Cassandra N. Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
- Institute for Systems Genomics, UConn, Farmington, CT 06032 USA
| | - Michael L. Stitzel
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
- Institute for Systems Genomics, UConn, Farmington, CT 06032 USA
| |
Collapse
|
8
|
Kaniuka O, Deregowska A, Bandura Y, Sabadashka M, Chala D, Kulachkovskyi O, Kubis H, Adamczyk-Grochala J, Sybirna N. Upregulation of GRP78 is accompanied by decreased antioxidant response and mitophagy promotion in streptozotocin-induced type 1 diabetes in rats. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167531. [PMID: 39353543 DOI: 10.1016/j.bbadis.2024.167531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Endoplasmic reticulum stress, oxidative stress, and mitochondrial dysfunction are interconnected processes involved in the pathogenesis of diabetes mellitus (DM). In the present study, we demonstrate a distinct unfolded protein response (UPR) signaling pathways in two mammalian models of DM: β-TC-6 cell line and streptozotocin-induced type 1 diabetes model in rats. However, a feature common to both systems was the upregulation of the GRP78 protein. Moreover, in vivo studies showed the disruption of the antioxidant system and an escalation of mitophagy against the background of a depletion of the level of ATP in pancreatic cells. In conclusion, we suggest that glucotoxic conditions induced GRP78 upregulation, and next cause depletion of the antioxidant pool and disruption of the functioning of antioxidant defense enzymes and in consequence promote mitophagy in pancreatic cells. Therefore, GRP78 may be considered as a potential therapeutic factor in patients with diabetes.
Collapse
Affiliation(s)
- O Kaniuka
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - A Deregowska
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - Yu Bandura
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - M Sabadashka
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - D Chala
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - O Kulachkovskyi
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - H Kubis
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - J Adamczyk-Grochala
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - N Sybirna
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| |
Collapse
|
9
|
Zhao H, Liu Z, Chen H, Han M, Zhang M, Liu K, Jin H, Liu X, Shi M, Pu W, Werner M, Meister M, Kauschke SG, Sun R, Wang J, Shen R, Wang QD, Ma X, Tchorz JS, Zhou B. Identifying specific functional roles for senescence across cell types. Cell 2024; 187:7314-7334.e21. [PMID: 39368477 DOI: 10.1016/j.cell.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/16/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Cellular senescence plays critical roles in aging, regeneration, and disease; yet, the ability to discern its contributions across various cell types to these biological processes remains limited. In this study, we generated an in vivo genetic toolbox consisting of three p16Ink4a-related intersectional genetic systems, enabling pulse-chase tracing (Sn-pTracer), Cre-based tracing and ablation (Sn-cTracer), and gene manipulation combined with tracing (Sn-gTracer) of defined p16Ink4a+ cell types. Using liver injury and repair as an example, we found that macrophages and endothelial cells (ECs) represent distinct senescent cell populations with different fates and functions during liver fibrosis and repair. Notably, clearance of p16Ink4a+ macrophages significantly mitigates hepatocellular damage, whereas eliminating p16Ink4a+ ECs aggravates liver injury. Additionally, targeted reprogramming of p16Ink4a+ ECs through Kdr overexpression markedly reduces liver fibrosis. This study illuminates the functional diversity of p16Ink4a+ cells and offers insights for developing cell-type-specific senolytic therapies in the future.
Collapse
Affiliation(s)
- Huan Zhao
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zixin Liu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hui Chen
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Maoying Han
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Mingjun Zhang
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kuo Liu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Hengwei Jin
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuxiu Liu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Mengyang Shi
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenjuan Pu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Markus Werner
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Michael Meister
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Stefan G Kauschke
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ruilin Sun
- Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Jinjin Wang
- Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xin Ma
- Department of Pharmacology, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jan S Tchorz
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Bin Zhou
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
10
|
Liu Y, Yang X, Zhou J, Yang H, Yang R, Zhu P, Zhou R, Wu T, Gao Y, Ye Z, Li X, Liu R, Zhang W, Zhou H, Li Q. OSGEP regulates islet β-cell function by modulating proinsulin translation and maintaining ER stress homeostasis in mice. Nat Commun 2024; 15:10479. [PMID: 39622811 PMCID: PMC11612026 DOI: 10.1038/s41467-024-54905-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Proinsulin translation and folding is crucial for glucose homeostasis. However, islet β-cell control of Proinsulin translation remains incompletely understood. Here, we identify OSGEP, an enzyme responsible for t6A37 modification of tRNANNU that tunes glucose metabolism in β-cells. Global Osgep deletion causes glucose intolerance, while β-cell-specific deletion induces hyperglycemia and glucose intolerance due to impaired insulin activity. Transcriptomics and proteomics reveal activation of the unfolded protein response (UPR) and apoptosis signaling pathways in Osgep-deficient islets, linked to an increase in misfolded Proinsulin from reduced t6A37 modification. Osgep overexpression in pancreas rescues insulin secretion and mitigates diabetes in high-fat diet mice. Osgep enhances translational fidelity and alleviates UPR signaling, highlighting its potential as a therapeutic target for diabetes. Individuals carrying the C allele at rs74512655, which promotes OSGEP transcription, may show reduced susceptibility to T2DM. These findings show OSGEP is essential for islet β-cells and a potential diabetes therapy target.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
- Department of Pharmacy, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xuechun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Jian Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Ruimeng Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Tianyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Rong Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China.
| |
Collapse
|
11
|
Hetz C, Dillin A. Central role of the ER proteostasis network in healthy aging. Trends Cell Biol 2024:S0962-8924(24)00209-5. [PMID: 39547881 DOI: 10.1016/j.tcb.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Aging trajectories vary among individuals, characterized by progressive functional decline, often leading to disease states. One of the central hallmarks of aging is the deterioration of proteostasis, where the function of the endoplasmic reticulum (ER) is dramatically affected. ER stress is monitored and adjusted by the unfolded protein response (UPR); a signaling pathway that mediates adaptive processes to restore proteostasis. Studies in multiple model organisms (yeast, worms, flies, and mice) in addition to human tissue indicates that adaptive UPR signaling contributes to healthy aging. Strategies to improve ER proteostasis using small molecules and gene therapy reduce the decline of organ function during normal aging in mammals. This article reviews recent advances in understanding the significance of the ER proteostasis network to normal aging and its relationship with other hallmarks of aging such as senescence.
Collapse
Affiliation(s)
- Claudio Hetz
- The Buck Institute for Research in Aging, Novato, CA 94945, USA; Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile.
| | - Andrew Dillin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
12
|
Yi X, Eizirik DL. β-Cell gene expression stress signatures in types 1 and 2 diabetes. J Diabetes 2024; 16:e70026. [PMID: 39505716 PMCID: PMC11540585 DOI: 10.1111/1753-0407.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024] Open
Affiliation(s)
- Xiaoyan Yi
- ULB Center for Diabetes Research, Medical FacultyUniversité Libre de BruxellesBrusselsBelgium
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical FacultyUniversité Libre de BruxellesBrusselsBelgium
| |
Collapse
|
13
|
Yang Y, Wang B, Dong H, Lin H, Yuen-man Ho M, Hu K, Zhang N, Ma J, Xie R, Cheng KKY, Li X. The mitochondrial enzyme pyruvate carboxylase restricts pancreatic β-cell senescence by blocking p53 activation. Proc Natl Acad Sci U S A 2024; 121:e2401218121. [PMID: 39436667 PMCID: PMC11536080 DOI: 10.1073/pnas.2401218121] [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: 02/03/2024] [Accepted: 09/07/2024] [Indexed: 10/23/2024] Open
Abstract
Defective glucose-stimulated insulin secretion (GSIS) and β-cell senescence are hallmarks in diabetes. The mitochondrial enzyme pyruvate carboxylase (PC) has been shown to promote GSIS and β-cell proliferation in the clonal β-cell lines, yet its physiological relevance remains unknown. Here, we provide animal and human data showing a role of PC in protecting β-cells against senescence and maintaining GSIS under different physiological and pathological conditions. β-cell-specific deletion of PC impaired GSIS and induced β-cell senescence in the mouse models under either a standard chow diet or prolonged high-fat diet feeding. Transcriptomic analysis indicated that p53-related senescence and cell cycle arrest are activated in PC-deficient islets. Overexpression of PC inhibited hyperglycemia- and aging-induced p53-related senescence in human and mouse islets as well as INS-1E β-cells, whereas knockdown of PC provoked senescence. Mechanistically, PC interacted with MDM2 to prevent its degradation via the MDM2 binding motif, which in turn restricts the p53-dependent senescent program in β-cells. On the contrary, the regulatory effects of PC on GSIS and the tricarboxylic acid (TCA) anaplerotic flux are p53-independent. We illuminate a function of PC in controlling β-cell senescence through the MDM2-p53 axis.
Collapse
Affiliation(s)
- Yumei Yang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200030, China
| | - Baomin Wang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200030, China
| | - Haoru Dong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200030, China
| | - Huige Lin
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen518000, China
| | - Melody Yuen-man Ho
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong999077, China
| | - Ke Hu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200030, China
| | - Na Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200030, China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200030, China
| | - Rong Xie
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200030, China
| | - Kenneth King-yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen518000, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200030, China
| |
Collapse
|
14
|
Rampazzo Morelli N, Préfontaine C, Pipella J, Thompson PJ. Secreted GDF15 maintains transcriptional responses during DNA damage-mediated senescence in human beta cells. Am J Physiol Endocrinol Metab 2024; 327:E552-E562. [PMID: 39196800 PMCID: PMC11482276 DOI: 10.1152/ajpendo.00257.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 08/30/2024]
Abstract
Type 1 diabetes (T1D) is a chronic metabolic disease resulting from an autoimmune destruction of pancreatic beta cells. Beta cells activate various stress responses during the development of T1D, including senescence, which involves cell cycle arrest, prosurvival signaling, and a proinflammatory secretome termed the senescence-associated secretory phenotype (SASP). We previously identified growth and differentiation factor 15 (GDF15) as a major SASP factor in human islets and human EndoC-βH5 beta cells in a model of DNA damage-mediated senescence that recapitulates features of senescent beta cells in T1D. Soluble GDF15 has been shown to exert protective effects on human and mouse beta cells during various forms of stress relevant to T1D; therefore, we hypothesized that secreted GDF15 may play a prosurvival role during DNA damage-mediated senescence in human beta cells. We found that elevated GDF15 secretion was associated with endogenous senescent beta cells in an islet preparation from a T1D donor, supporting the validity of our DNA damage model. Using antibody-based neutralization, we found that secreted endogenous GDF15 was not required for senescent human islet or EndoC cell viability. Rather, neutralization of GDF15 led to reduced expression of specific senescence-associated genes, including GDF15 itself and the prosurvival gene BCL2-like protein 1 (BCL2L1). Taken together, these data suggest that SASP factor GDF15 is not required to sustain senescent human islet viability, but it is required to maintain senescence-associated transcriptional responses.NEW & NOTEWORTHY Beta cell senescence is an emerging contributor to the pathogenesis of type 1 diabetes, but candidate therapeutic targets have not been identified in human beta cells. In this study, we examined the role of a secreted factor, GDF15, and found that although it is not required to maintain viability during senescence, it is required to fine-tune gene expression programs involved in the senescence response during DNA damage in human beta cells.
Collapse
Affiliation(s)
- Nayara Rampazzo Morelli
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Camille Préfontaine
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jasmine Pipella
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter J Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
15
|
Rodvold JJ, Grimmer M, Ruiz K, Marsters SA, Oikonomidi I, Tan-Aristy E, Pham VC, Sarkar T, Harnoss JM, Shatz-Binder W, Modrusan ZD, Wu TD, Lill JR, Villemure E, Rudolph J, de Sousa e Melo F, Ashkenazi A. ATF6 Promotes Colorectal Cancer Growth and Stemness by Regulating the Wnt Pathway. CANCER RESEARCH COMMUNICATIONS 2024; 4:2734-2755. [PMID: 39324706 PMCID: PMC11492184 DOI: 10.1158/2767-9764.crc-24-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/07/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
SIGNIFICANCE ATF6 intervention reduces colorectal cancer cell and organoid viability by interrupting dysregulated Wnt signaling, identifying a novel facilitator and potential therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Jeffrey J. Rodvold
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Matthew Grimmer
- Department of Computational Science, Genentech, Inc., South San Francisco, California
| | - Karen Ruiz
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California
| | - Scot A. Marsters
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Ioanna Oikonomidi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Eileen Tan-Aristy
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Victoria C. Pham
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California
| | - Tamal Sarkar
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University Hospital Giessen, Giessen, Germany
| | - Jonathan M. Harnoss
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University Hospital Giessen, Giessen, Germany
| | - Whitney Shatz-Binder
- Department of Pharmaceutical Development, Genentech, Inc., South San Francisco, California
| | - Zora D. Modrusan
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California
| | - Thomas D. Wu
- Department of Computational Science, Genentech, Inc., South San Francisco, California
| | - Jennie R. Lill
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California
| | - Elisia Villemure
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California
| | - Joachim Rudolph
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California
| | | | - Avi Ashkenazi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| |
Collapse
|
16
|
Cheng H, Zhao Z, Liu D, Wang Y, Zhang M. Early senescence of pancreatic β cells induced by unfolded protein response deficiency prevents type 1 diabetes. J Zhejiang Univ Sci B 2024; 25:796-799. [PMID: 39308069 PMCID: PMC11422799 DOI: 10.1631/jzus.b2400013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Indexed: 09/27/2024]
Abstract
Type 1 diabetes (T1D) is a T lymphocyte-mediated autoimmune disease caused by pancreatic β-cell destruction, which eventually leads to reduced insulin level and increased blood glucose level (Syed, 2022). As a multifactorial disease, T1D is characterized by a genetic predisposition associated with various environmental and cellular elements (Syed, 2022). Pancreatic β cells have long been considered the "innocent victims" in T1D pathogenesis since the pancreas is attacked by the immune cells, resulting in a process known as insulitis, in which the immune cells infiltrate pancreatic islets and secrete pro-inflammatory cytokines. However, growing evidence suggests that various β-cell stresses, dysfunction, and death contribute to T1D pathogenesis, as it has been observed that β-cell dysfunction in autoantibody-positive (Aab+) individuals exists long before T1D diagnosis (Evans-Molina et al., 2018).
Collapse
Affiliation(s)
- Haipeng Cheng
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zhenwang Zhao
- School of Basic Medicine, Health Science Center, Hubei University of Arts and Science, Xiangyang 441053, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541199, China
| | - Dan Liu
- Medical Education Department, Guangdong Provincial People's Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai 519041, China
| | - Yufei Wang
- School of Basic Medicine, Health Science Center, Hubei University of Arts and Science, Xiangyang 441053, China. ,
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541199, China. ,
| | - Min Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Department of Bioinformatics and Medical Big Data, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
17
|
Wang X, Zhang C, Su J, Ren S, Wang X, Zhang Y, Yuan Z, He X, Wu X, Li M, Du F, Chen Y, Deng S, Zhao Y, Wang X, Sun Y, Shen J, Ji H, Hou Y, Xiao Z. Rejuvenation Strategy for Inducing and Enhancing Autoimmune Response to Eliminate Senescent Cells. Aging Dis 2024:AD.2024.0579. [PMID: 39122450 DOI: 10.14336/ad.2024.0579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The process of aging, which involves progressive changes in the body over time, is closely associated with the development of age-related diseases. Cellular senescence is a pivotal hallmark and mechanism of the aging process. The accumulation of senescent cells can significantly contribute to the onset of age-related diseases, thereby compromising overall health. Conversely, the elimination of senescent cells enhances the body's regenerative and reparative capacity, thereby retarding the aging process. Here, we present a brief overview of 12 Hallmarks of aging and subsequently emphasize the potential of immune checkpoint blockade, innate immune cell therapy (including T cells, iNKT cells, macrophages, and NK cells), as well as CAR-T cell therapy for inducing and augmenting immune responses aimed at eliminating senescent cells. In addition to CAR-T cells, we also explore the possibility of engineered immune cells such as CAR-NK and CAR-M cells to eliminate senescent cells. In summary, immunotherapy, as an emerging strategy for the treatment of aging, offers new prospects for age-related research.
Collapse
Affiliation(s)
- Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chengyu Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zijun Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yunqing Hou
- LongmaTan District People's Hospital of Luzhou City, Luzhou 646600, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang 621000, China
| |
Collapse
|
18
|
Rampazzo Morelli N, Pipella J, Thompson PJ. Establishing evidence for immune surveillance of β-cell senescence. Trends Endocrinol Metab 2024; 35:576-585. [PMID: 38307810 DOI: 10.1016/j.tem.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024]
Abstract
Cellular senescence is a programmed state of cell cycle arrest that involves a complex immunogenic secretome, eliciting immune surveillance and senescent cell clearance. Recent work has shown that a subpopulation of pancreatic β-cells becomes senescent in the context of diabetes; however, it is not known whether these cells are normally subject to immune surveillance. In this opinion article, we advance the hypothesis that immune surveillance of β-cells undergoing a senescence stress response normally limits their accumulation during aging and that the breakdown of these mechanisms is a driver of senescent β-cell accumulation in diabetes. Elucidation and therapeutic activation of immune surveillance mechanisms in the pancreas holds promise for the improvement of approaches to target stressed senescent β-cells in the treatment of diabetes.
Collapse
Affiliation(s)
- Nayara Rampazzo Morelli
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jasmine Pipella
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Peter J Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
19
|
MacDonald T, Ryback B, da Silva Pereira JA, Wei S, Mendez B, Cai E, Ishikawa Y, Weir G, Bonner-Weir S, Kissler S, Yi P. Renalase inhibition regulates β cell metabolism to defend against acute and chronic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598322. [PMID: 38915698 PMCID: PMC11195134 DOI: 10.1101/2024.06.11.598322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Renalase (Rnls), annotated as an oxidase enzyme, is a GWAS gene associated with Type 1 Diabetes (T1D) risk. We previously discovered that Rnls inhibition delays diabetes onset in mouse models of T1D in vivo , and protects pancreatic β cells against autoimmune killing, ER and oxidative stress in vitro . The molecular biochemistry and functions of Rnls are entirely uncharted. Here we find that Rnls inhibition defends against loss of β cell mass and islet dysfunction in chronically stressed Akita mice in vivo . We used RNA sequencing, untargeted and targeted metabolomics and metabolic function experiments in mouse and human β cells and discovered a robust and conserved metabolic shift towards glycolysis, amino acid abundance and GSH synthesis to counter protein misfolding stress, in vitro . Our work illustrates a function for Rnls in mammalian cells, and suggests an axis by which manipulating intrinsic properties of β cells can rewire metabolism to protect against diabetogenic stress.
Collapse
|
20
|
Webster KL, Mirmira RG. Beta cell dedifferentiation in type 1 diabetes: sacrificing function for survival? Front Endocrinol (Lausanne) 2024; 15:1427723. [PMID: 38904049 PMCID: PMC11187278 DOI: 10.3389/fendo.2024.1427723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
The pathogeneses of type 1 and type 2 diabetes involve the progressive loss of functional beta cell mass, primarily attributed to cellular demise and/or dedifferentiation. While the scientific community has devoted significant attention to unraveling beta cell dedifferentiation in type 2 diabetes, its significance in type 1 diabetes remains relatively unexplored. This perspective article critically analyzes the existing evidence for beta cell dedifferentiation in type 1 diabetes, emphasizing its potential to reduce beta cell autoimmunity. Drawing from recent advancements in both human studies and animal models, we present beta cell identity as a promising target for managing type 1 diabetes. We posit that a better understanding of the mechanisms of beta cell dedifferentiation in type 1 diabetes is key to pioneering interventions that balance beta cell function and immunogenicity.
Collapse
Affiliation(s)
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
21
|
Mitra J, Dharmalingam P, Kodavati M, Guerrero EN, Rao KS, Garruto RM, Hegde ML. Endogenous TDP-43 mislocalization in a novel knock-in mouse model reveals DNA repair impairment, inflammation, and neuronal senescence. RESEARCH SQUARE 2024:rs.3.rs-3879966. [PMID: 38343852 PMCID: PMC10854316 DOI: 10.21203/rs.3.rs-3879966/v2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
TDP-43 mislocalization and aggregation are key pathological features of motor neuron diseases (MND) including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, transgenic hTDP-43 WT or ΔNLS-overexpression animal models mainly capture late-stages TDP-43 proteinopathy, and do not provide a complete understanding of early motor neuron-specific pathology during pre-symptomatic phases. We have now addressed this shortcoming by generating a new endogenous knock-in (KI) mouse model using a combination of CRISPR/Cas9 and FLEX Cre-switch strategy for the conditional expression of a mislocalized Tdp-43ΔNLS variant of mouse Tdp-43. This variant is either expressed conditionally in whole mice or specifically in the motor neurons. The mice exhibit loss of nuclear Tdp-43 concomitant with its cytosolic accumulation and aggregation in targeted cells, leading to increased DNA double-strand breaks (DSBs), signs of inflammation and DNA damage-associated cellular senescence. Notably, unlike WT Tdp43 which functionally interacts with Xrcc4 and DNA Ligase 4, the key DSB repair proteins in the non-homologous end-joining (NHEJ) pathway, the Tdp-43ΔNLS mutant sequesters them into cytosolic aggregates, exacerbating neuronal damage in mice brain. The mutant mice also exhibit myogenic degeneration in limb muscles and distinct motor deficits, consistent with the characteristics of MND. Our findings reveal progressive degenerative mechanisms in motor neurons expressing endogenous Tdp-43ΔNLS mutant, independent of TDP-43 overexpression or other confounding etiological factors. Thus, this unique Tdp-43 KI mouse model, which displays key molecular and phenotypic features of Tdp-43 proteinopathy, offers a significant opportunity to further characterize the early-stage progression of MND and also opens avenues for developing DNA repair-targeted approaches for treating TDP-43 pathology-linked neurodegenerative diseases.
Collapse
Affiliation(s)
- Joy Mitra
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Prakash Dharmalingam
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Manohar Kodavati
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Erika N. Guerrero
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - K. S. Rao
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation Deemed to be University, Green Fields, Vaddeswaram, Andhra Pradesh 522502, India
| | - Ralph M. Garruto
- Department of Anthropology, Binghamton University, State University of New York, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY 13902, USA
| | - Muralidhar L. Hegde
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
22
|
Mitra J, Dharmalingam P, Kodavati MM, Guerrero EN, Rao KS, Garruto R, Hegde ML. Endogenous TDP-43 mislocalization in a novel knock-in mouse model reveals DNA repair impairment, inflammation, and neuronal senescence. RESEARCH SQUARE 2024:rs.3.rs-3879966. [PMID: 38343852 PMCID: PMC10854316 DOI: 10.21203/rs.3.rs-3879966/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
TDP-43 mislocalization and aggregation are key pathological features of motor neuron diseases (MND) including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, transgenic hTDP-43 WT or ∆NLS-overexpression animal models mainly capture late-stages TDP-43 proteinopathy, and do not provide a complete understanding of early motor neuron-specific pathology during pre-symptomatic phases. We have now addressed this shortcoming by generating a new endogenous knock-in (KI) mouse model using a combination of CRISPR/Cas9 and FLEX Cre-switch strategy for the conditional expression of a mislocalized Tdp-43∆NLS variant of mouse Tdp-43. This variant is either expressed conditionally in whole mice or specifically in the motor neurons. The mice exhibit loss of nuclear Tdp-43 concomitant with its cytosolic accumulation and aggregation in targeted cells, leading to increased DNA double-strand breaks (DSBs), signs of inflammation and DNA damage-associated cellular senescence. Notably, unlike WT Tdp43 which functionally interacts with Xrcc4 and DNA Ligase 4, the key DSB repair proteins in the non-homologous end-joining (NHEJ) pathway, the Tdp-43∆NLS mutant sequesters them into cytosolic aggregates, exacerbating neuronal damage in mice brain. The mutant mice also exhibit myogenic degeneration in limb muscles and distinct motor deficits, consistent with the characteristics of MND. Our findings reveal progressive degenerative mechanisms in motor neurons expressing endogenous Tdp-43∆NLS mutant, independent of TDP-43 overexpression or other confounding etiological factors. Thus, this unique Tdp-43 KI mouse model, which displays key molecular and phenotypic features of Tdp-43 proteinopathy, offers a significant opportunity to further characterize the early-stage progression of MND and also opens avenues for developing DNA repair-targeted approaches for treating TDP-43 pathology-linked neurodegenerative diseases.
Collapse
|
23
|
Lee H, Thompson PJ, Engin F. Protocol for quantifying SA-β-gal activity as a measure of senescence in islets of a mouse model of type 1 diabetes. STAR Protoc 2024; 5:102923. [PMID: 38427571 PMCID: PMC10918323 DOI: 10.1016/j.xpro.2024.102923] [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: 11/30/2023] [Revised: 01/14/2024] [Accepted: 02/13/2024] [Indexed: 03/03/2024] Open
Abstract
A subpopulation of pancreatic beta cells becomes senescent during type 1 diabetes (T1D) progression, and removal of these populations protects against T1D in mice. Here, we present a protocol to measure senescence in murine pancreatic islet cells through analysis of senescence-associated β-galactosidase activity. We describe steps for staining with the fluorogenic substrate C12FDG and analysis by flow cytometry. Increased cell size is another marker of senescence and can also be concurrently measured in the same experiment. For complete details on the use and execution of this protocol, please refer to Lee et al.1 and Helman et al.2.
Collapse
Affiliation(s)
- Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA.
| | - Peter J Thompson
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB RT3 2N2, Canada
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, Wisconsin Institute for Discovery, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
24
|
Manji J, Pipella J, Brawerman G, Thompson PJ. Exploring Transcriptional Regulation of Beta Cell SASP by Brd4-Associated Proteins and Cell Cycle Control Protein p21. EPIGENOMES 2024; 8:10. [PMID: 38534794 DOI: 10.3390/epigenomes8010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Type 1 diabetes (T1D) is a metabolic disease resulting from progressive autoimmune destruction of insulin-producing pancreatic beta cells. Although the majority of beta cells are lost in T1D, a small subset undergoes senescence, a stress response involving growth arrest, DNA damage response, and activation of a senescence-associated secretory phenotype (SASP). SASP in beta cells of the nonobese diabetic (NOD) mouse model of T1D and primary human islets is regulated at the level of transcription by bromodomain extra-terminal (BET) proteins, but the mechanisms remain unclear. To explore how SASP is transcriptionally regulated in beta cells, we used the NOD beta cell line NIT-1 to model beta cell SASP and identified binding partners of BET protein Brd4 and explored the role of the cyclin-dependent kinase inhibitor p21. Brd4 interacted with a variety of proteins in senescent NIT-1 cells including subunits of the Ino80 chromatin remodeling complex, which was expressed in beta cells during T1D progression in NOD mice and in human beta cells of control, autoantibody-positive, and T1D donors as determined from single-cell RNA-seq data. RNAi knockdown of p21 during senescence in NIT-1 cells did not significantly impact viability or SASP. Taken together, these results suggest that Brd4 interacts with several protein partners during senescence in NIT-1 cells, some of which may play roles in SASP gene activation and that p21 is dispensable for the SASP in this beta cell model.
Collapse
Affiliation(s)
- Jasmine Manji
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Jasmine Pipella
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Gabriel Brawerman
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Peter J Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
25
|
Wenzlau JM, Peterson OJ, Vomund AN, DiLisio JE, Hohenstein A, Haskins K, Wan X. Mapping of a hybrid insulin peptide in the inflamed islet β-cells from NOD mice. Front Immunol 2024; 15:1348131. [PMID: 38455055 PMCID: PMC10917911 DOI: 10.3389/fimmu.2024.1348131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
There is accumulating evidence that pathogenic T cells in T1D recognize epitopes formed by post-translational modifications of β-cell antigens, including hybrid insulin peptides (HIPs). The ligands for several CD4 T-cell clones derived from the NOD mouse are HIPs composed of a fragment of proinsulin joined to peptides from endogenous β-cell granule proteins. The diabetogenic T-cell clone BDC-6.9 reacts to a fragment of C-peptide fused to a cleavage product of pro-islet amyloid polypeptide (6.9HIP). In this study, we used a monoclonal antibody (MAb) to the 6.9HIP to determine when and where HIP antigens are present in NOD islets during disease progression and with which immune cells they associate. Immunogold labeling of the 6.9HIP MAb and organelle-specific markers for electron microscopy were employed to map the subcellular compartment(s) in which the HIP is localized within β-cells. While the insulin B9-23 peptide was present in nearly all islets, the 6.9HIP MAb stained infiltrated islets only in NOD mice at advanced stages of T1D development. Islets co-stained with the 6.9HIP MAb and antibodies to mark insulin, macrophages, and dendritic cells indicate that 6.9HIP co-localizes within insulin-positive β-cells as well as intra-islet antigen-presenting cells (APCs). In electron micrographs, the 6.9HIP co-localized with granule structures containing insulin alone or both insulin and LAMP1 within β-cells. Exposing NOD islets to the endoplasmic reticulum (ER) stress inducer tunicamycin significantly increased levels of 6.9HIP in subcellular fractions containing crinosomes and dense-core granules (DCGs). This work demonstrates that the 6.9HIP can be visualized in the infiltrated islets and suggests that intra-islet APCs may acquire and present HIP antigens within islets.
Collapse
Affiliation(s)
- Janet M. Wenzlau
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Orion J. Peterson
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
- Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, United States
| | - Anthony N. Vomund
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
- Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, United States
| | - James E. DiLisio
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Anita Hohenstein
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kathryn Haskins
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Xiaoxiao Wan
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
- Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
26
|
Sun DY, Wu WB, Wu JJ, Shi Y, Xu JJ, Ouyang SX, Chi C, Shi Y, Ji QX, Miao JH, Fu JT, Tong J, Zhang PP, Zhang JB, Li ZY, Qu LF, Shen FM, Li DJ, Wang P. Pro-ferroptotic signaling promotes arterial aging via vascular smooth muscle cell senescence. Nat Commun 2024; 15:1429. [PMID: 38365899 PMCID: PMC10873425 DOI: 10.1038/s41467-024-45823-w] [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: 07/15/2023] [Accepted: 02/05/2024] [Indexed: 02/18/2024] Open
Abstract
Senescence of vascular smooth muscle cells (VSMCs) contributes to aging-related cardiovascular diseases by promoting arterial remodelling and stiffness. Ferroptosis is a novel type of regulated cell death associated with lipid oxidation. Here, we show that pro-ferroptosis signaling drives VSMCs senescence to accelerate vascular NAD+ loss, remodelling and aging. Pro-ferroptotic signaling is triggered in senescent VSMCs and arteries of aged mice. Furthermore, the activation of pro-ferroptotic signaling in VSMCs not only induces NAD+ loss and senescence but also promotes the release of a pro-senescent secretome. Pharmacological or genetic inhibition of pro-ferroptosis signaling, ameliorates VSMCs senescence, reduces vascular stiffness and retards the progression of abdominal aortic aneurysm in mice. Mechanistically, we revealed that inhibition of pro-ferroptotic signaling facilitates the nuclear-cytoplasmic shuttling of proliferator-activated receptor-γ and, thereby impeding nuclear receptor coactivator 4-ferrtin complex-centric ferritinophagy. Finally, the activated pro-ferroptotic signaling correlates with arterial stiffness in a human proof-of-concept study. These findings have significant implications for future therapeutic strategies aiming to eliminate vascular ferroptosis in senescence- or aging-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Di-Yang Sun
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wen-Bin Wu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jian-Jin Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Yu Shi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia-Jun Xu
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Shen-Xi Ouyang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Chi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Shi
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qing-Xin Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Hao Miao
- Department of Orthopedic Surgery/Spine Center, Changzheng Hospital Affiliated Hospital of Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jiang-Tao Fu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping-Ping Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jia-Bao Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Zhi-Yong Li
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Le-Feng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University/Second Military Medical University, Shanghai, China.
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Pei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China.
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China.
| |
Collapse
|
27
|
Tysoe O. β-cells protected from T1DM by early senescence programme. Nat Rev Endocrinol 2024; 20:1. [PMID: 38001397 DOI: 10.1038/s41574-023-00928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
|