1
|
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Sada Japp A, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Luning Prak ET, Kaestner KH, Naji A, Betts MR. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset. Nat Commun 2025; 16:4621. [PMID: 40383826 PMCID: PMC12086209 DOI: 10.1038/s41467-025-59626-0] [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: 09/24/2024] [Accepted: 04/25/2025] [Indexed: 05/20/2025] Open
Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. However, characterization of the immunological processes occurring in human pancreatic lymphatic tissues is lacking. Here, we conduct a comprehensive study of immune cells from pancreatic, mesenteric, and splenic lymphatic tissues of non-diabetic control (ND), β cell autoantibody-positive non-diabetic (AAb+), and T1D donors using flow cytometry and CITEseq. Compared to ND pancreas-draining lymph nodes (pLN), AAb+ and T1D donor pLNs display decreased CD4+ Treg and increased stem-like CD8+ T cell signatures, while only T1D donor pLNs exhibit naive T cell and NK cell differentiation. Mesenteric LNs have modulations only in CD4+ Tregs and naive cells, while splenocytes lack these perturbations. Further, T cell expression of activation markers and IL7 receptor correlate with T1D genetic risk. These results demonstrate tissue-restricted immune changes occur before and after T1D onset.
Collapse
Affiliation(s)
- Gregory J Golden
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Vincent H Wu
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jacob T Hamilton
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL, 32610, USA
| | - Alberto Sada Japp
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Chengyang Liu
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - M Betina Pampena
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - James J Knox
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jay S Gardner
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ali Naji
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Michael R Betts
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Añé-Kourí AL, Palomino JL, Lorenzo-Luaces P, Sanchez L, Ledon N, Pereira K, Hernandez JDLC, Suárez GM, García B, González A, Saavedra D, Lage A. Multivariate analysis of immunosenescence data in healthy humans and diverse diseases. FRONTIERS IN AGING 2025; 6:1568034. [PMID: 40308557 PMCID: PMC12040824 DOI: 10.3389/fragi.2025.1568034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025]
Abstract
Introduction Immunosenescence is a dynamic process, where both genetic and environmental factors account for the substantial inter-individual variability. This paper integrates all the data on immunosenescence markers generated in our laboratory and describes the differences and/or similarities between individuals based on their biological conditions (immunosenescence markers) and their associations with chronological age and health status. Materials and Methods The dataset consisted of immunological data from healthy donors, centenarians, patients diagnosed with chronic kidney disease, COVID-19 and non-small cell lung cancer (NSCLC), treatment-naïve or treated with platinum-based chemotherapy. To determine whether there are groups of immunologically different individuals despite their age or clinical condition, cluster analysis was performed. Canonical discriminant analysis was performed to determine which variables characterize each cluster. Results There are differences in the expression of immunosenescence markers between healthy subjects and patients diagnosed with different pathological conditions, regardless of their age. Meanwhile, the distribution of the clusters indicates the presence of two separate groups of healthy participants, one of them characterized by a high frequency of naïve lymphocytes, and the other with high expression of terminally differentiated lymphocyte subsets. Advanced NSCLC treatment-naïve patients were in the same cluster as a group of healthy subjects. Additionally, centenarians belong to a different cluster than healthy subjects, suggesting they might have a unique immune signature. Conclusion The distribution of clusters appears to be more appropriate than univariate associations of single markers for health and disease research. The present work reveals which immune markers are relevant in different physiological and pathological contexts and indicates the need for deeper studies on the biological age of the immune system.
Collapse
Affiliation(s)
- Ana Laura Añé-Kourí
- Clinical Research Direction, Center of Molecular Immunology, Havana, Cuba
- Biomedical Sciences Institute, Hasselt University, Hasselt, Belgium
| | | | | | - Lizet Sanchez
- Clinical Research Direction, Center of Molecular Immunology, Havana, Cuba
| | - Nuris Ledon
- Research Direction, Center of Molecular Immunology, Havana, Cuba
| | - Karla Pereira
- Research Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Gisela María Suárez
- Laboratory of Immunology, Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Beatriz García
- Clinical Research Direction, Center of Molecular Immunology, Havana, Cuba
| | - Amnely González
- Clinical Research Direction, Center of Molecular Immunology, Havana, Cuba
| | - Danay Saavedra
- Clinical Research Direction, Center of Molecular Immunology, Havana, Cuba
- Diabetes Research Institute, University of Miami, Miami, FL, United States
| | - Agustin Lage
- Clinical Research Direction, Center of Molecular Immunology, Havana, Cuba
| |
Collapse
|
3
|
Evans-Molina C, Oram RA. Type 1 diabetes presenting in adults: Trends, diagnostic challenges and unique features. Diabetes Obes Metab 2025. [PMID: 40230204 DOI: 10.1111/dom.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Type 1 diabetes (T1D) has been historically regarded as a childhood-onset disease; however, recent epidemiological data indicate that adult-onset T1D accounts for a substantial proportion of cases worldwide. There is evidence that adult-onset T1D is associated with the classic T1D triad of elevated genetic risk, the presence of islet-specific autoantibodies and progression to severe insulin deficiency. In this article, we review our understanding of the commonalities and differences between childhood and adult-onset T1D, and we highlight significant knowledge gaps in our understanding of the diagnosis, incidence, trajectory and treatment of adult-onset T1D. Compared to children, adults presenting with T1D exhibit differences in genetic risk, immunologic profiles and metabolic outcomes, including differences in the type and number of autoantibodies present, genetic associations and total genetic burden, rates of C-peptide decline, the persistence of C-peptide in long-duration disease and glycaemic control. In addition, obesity and metabolic syndrome are increasingly common in adults, which not only blurs the clinical distinction of adult-onset T1D from type 2 diabetes (T2D) but also likely contributes to differences in metabolic outcomes and rates of progression. Because T2D is so prevalent in the adult population, adult-onset T1D is misclassified as T2D in at least one in three cases, leading to delays in appropriate treatment. Current diagnostic tools, including autoantibody testing and C-peptide measurement, are underutilised or lack specificity in distinguishing adult-onset T1D from atypical T2D. Additionally, the impact of different responses to disease-modifying therapy between adults and children is unclear. Addressing these knowledge gaps requires expanded epidemiological studies, diverse patient registries and refined classification criteria to improve early detection and treatment strategies. A deeper understanding of adult-onset T1D will be critical to reduce the burden of misdiagnosis, lead to earlier diagnosis and treatment and optimise population-based screening approaches in this under-recognised population. PLAIN LANGUAGE SUMMARY: Type 1 diabetes (T1D) is an autoimmune disease that causes metabolic and nutritional complications due to the destruction of insulin-producing pancreatic β cells. T1D was formerly known as "juvenile diabetes" because it was assumed that most cases occurred in childhood; however, recent epidemiological data show that nearly half of all T1D cases are diagnosed in adulthood. Despite the high prevalence of adult-onset T1D, there are challenges with correctly diagnosing T1D in adulthood, and significant knowledge gaps remain regarding the incidence, trajectory, and treatment of adult-onset T1D. In this article, we summarize the current understanding of commonalities and differences between childhood and adult-onset T1D. Particularly, we highlight age-related differences in genetic risk, immunologic profiles, and metabolic outcomes and complications. Finally, we highlight key gaps in our understanding of adult-onset T1D that need to be addressed to reduce the burden of misdiagnosis and allow for better screening and treatment of T1D in adulthood.
Collapse
Affiliation(s)
- Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Richard A Oram
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
- The Academic Renal Unit, Royal Devon University Hospitals NHS Foundation Trust, Exeter, UK
| |
Collapse
|
4
|
Meyer M, Fourie C, van der Merwe H, Botha H, Engelbrecht AM. Targeting treatment resistance in cervical cancer: A new avenue for senolytic therapies. Adv Med Sci 2025; 70:33-43. [PMID: 39549742 DOI: 10.1016/j.advms.2024.11.001] [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: 04/05/2024] [Revised: 07/27/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
Cervical cancer poses a significant global health challenge, particularly impacting women in economically developing nations. This disparity stems from a combination of factors, including inadequate screening infrastructure and resource limitations. However, the foremost contributor is the widespread lack of awareness and limited accessibility to Human Papillomavirus (HPV) vaccination, which is a key preventative measure against cervical cancer development. Despite advancements in cervical cancer prevention, treatment resistance remains a major hurdle in achieving improved patient outcomes. Cellular senescence, specifically the senescence-associated secretory phenotype (SASP) and its bidirectional relationship with the immune system, has been implicated in resistance to conventional cervical cancer chemotherapy treatments. The exact mechanisms by which this state of growth arrest and the associated changes in immune regulation contribute to cervical cancer progression and the associated drug resistance are not entirely understood. This underscores the necessity for innovative strategies to address the prevalence of treatment-resistant cervical cancer, with one promising avenue being the utilisation of senolytics. Senolytics are agents that have promising efficacy in clearing senescent cells from tumour tissues, however neither the utilisation of senolytics for addressing senescence-induced treatment resistance nor the potential integration of immunotherapy as senolytic agents in cervical cancer treatment has been explored to date. This review provides a concise overview of the mechanisms underlying senescence induction and the pivotal role of the immune system in this process. Additionally, it explores various senolytic approaches that hold significant potential for advancing cervical cancer research.
Collapse
Affiliation(s)
- Madré Meyer
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Carla Fourie
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Haynes van der Merwe
- Department of Obstetrics and Gynaecology, Stellenbosch University Medical Campus, Cape Town, South Africa
| | - Hennie Botha
- Department of Obstetrics and Gynaecology, Stellenbosch University Medical Campus, Cape Town, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
5
|
Williams MD, Grace CR, Posgai AL, McGrail KM, Brusko MA, Haller MJ, Jacobsen L, Schatz D, Brusko TM, Atkinson M, Bacher R, Wasserfall CH. Serological markers of exocrine pancreatic function are differentially informative for distinguishing individuals progressing to type 1 diabetes. BMJ Open Diabetes Res Care 2025; 13:e004655. [PMID: 39755561 PMCID: PMC11749058 DOI: 10.1136/bmjdrc-2024-004655] [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: 10/03/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025] Open
Abstract
INTRODUCTION Altered serum levels of growth hormones, adipokines, and exocrine pancreas enzymes have been individually linked with type 1 diabetes (T1D). We collectively evaluated seven such biomarkers, combined with islet autoantibodies (AAb) and genetic risk score (GRS2), for their utility in predicting AAb/T1D status. RESEARCH DESIGN AND METHODS Cross-sectional serum samples (n=154 T1D, n=56 1AAb+, n=77 ≥2AAb+, n=256 AAb-) were assessed for IGF1, IGF2, adiponectin, leptin, amylase, lipase, and trypsinogen (n=543, age range 2.7-30.0 years) using random forest modeling. RESULTS GRS2, age, lipase, trypsinogen, and AAb against ZnT8, GAD65, and insulin were the most informative markers. Notably, these variables were differentially informative according to AAb/T1D status. Higher GRS2 (p<0.001) and lower lipase levels (p=0.002) favored ≥2AAb+ versus AAb- classification. AAb against ZnT8 (p<0.01), GAD65 (p=0.021), or insulin (p=0.01) each independently favored ≥2AAb+ versus 1AAb+ classification. Reduced trypsinogen (p<0.001) and increased lipase levels (p<0.001) favored recent-onset T1D versus ≥2AAb+ classification. CONCLUSIONS Among the serological markers tested, lipase and trypsinogen levels were the most informative for differentiating among clinical groups, with the utility of each enzyme varying according to GRS2 and AAb/T1D status. These data support exocrine pancreas enzymes as candidates for longitudinal follow-up.
Collapse
Affiliation(s)
- MacKenzie D Williams
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Catherine Ramsey Grace
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Kieran M McGrail
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Maigan A Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Michael J Haller
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Laura Jacobsen
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Desmond Schatz
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
6
|
Aljobaily N, Allard D, Perkins B, Raugh A, Galland T, Jing Y, Stephens WZ, Bettini ML, Hale JS, Bettini M. Autoimmune CD4 + T cells fine-tune TCF1 expression to maintain function and survive persistent antigen exposure during diabetes. Immunity 2024; 57:2583-2596.e6. [PMID: 39396521 PMCID: PMC11563894 DOI: 10.1016/j.immuni.2024.09.016] [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: 05/09/2024] [Revised: 07/21/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
Self-reactive T cells experience chronic antigen exposure but do not exhibit signs of exhaustion. Here, we investigated the mechanisms for sustained, functioning autoimmune CD4+ T cells despite chronic stimulation. Examination of T cell priming showed that CD4+ T cells activated in the absence of infectious signals retained TCF1 expression. At later time points and during blockade of new T cell recruitment, most islet-infiltrating autoimmune CD4+ T cells were TCF1+, although expression was reduced on a per T cell basis. The Tcf7 locus was epigenetically modified in circulating autoimmune CD4+ T cells, suggesting a pre-programmed de novo methylation of the locus in early stages of autoimmune CD4+ T cell differentiation. This mirrored the epigenetic profile of recently recruited CD4+CD62L+ T cells in the pancreas. Collectively, these data reveal a unique environment during autoimmune CD4+ T cell priming that allows T cells to fine-tune TCF1 expression and maintain long-term survival and function.
Collapse
Affiliation(s)
- Nouf Aljobaily
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Denise Allard
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Bryant Perkins
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Arielle Raugh
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Tessa Galland
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Yi Jing
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - W Zac Stephens
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Matthew L Bettini
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - J Scott Hale
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Maria Bettini
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
7
|
Coughlin BA, Christian B, Trombley B, Mohr S. Interleukin-1 receptor-dependent and -independent caspase-1 activity in retinal cells mediated by receptor interacting protein 2. Front Cell Dev Biol 2024; 12:1467799. [PMID: 39483336 PMCID: PMC11525982 DOI: 10.3389/fcell.2024.1467799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Inflammation and cell death play an important role in the pathogenesis of diabetic retinopathy. Previously we observed sustained activation of pro-inflammatory caspase-1 in retinas of diabetic animals and patients. In this study, we aimed to look at mechanisms underlying chronic caspase-1 activation in vitro and in vivo. Methods Non-diabetic and diabetic wild type and IL-1 receptor (IL-1R1) knockout mice were used for in vivo experiments. Diabetes was induced using STZ (streptozotocin). Human Müller cells were used for in vitro studies. Cells were treated with either 5 mM or 25 mM glucose or interleukin-1beta (IL-1β) in the presence or absence of IL-1 receptor antagonist (IL-1ra) or siRNA against RIP2 (receptor interacting protein-2) for up to 96 h. Outcome measurements to assess Müller cell functions included measurements of caspase-1 activity using a fluorescence peptide substrate, production of IL-1β by Elisa, and cell death using trypan blue exclusion assays. Results Our in vivo results demonstrate that caspase-1 activation progresses from an IL-1R1 independent mechanism at 10 weeks of diabetes to an IL-1R1 dependent mechanism at 20 weeks indicating that feedback through IL-1R1 is crucial for sustained caspase-1 activity in retinas of mice. A similar hyperglycemia-mediated caspase-1/IL-1β/IL-1R1 feedback signaling was detected in vitro in human Müller cells which was prevented by treatment with IL-1ra. Our data also indicate that hyperglycemia induces caspase-1 activation initially but IL-1β sustains caspase-1 activation via caspase-1/IL-1β/IL-1R1 feedback and we identified RIP2 as mediator for both hyperglycemia- and IL-1β-induced caspase-1 activation. Activation of caspase-1/IL-1β/IL-1R1 feedback signaling caused Müller cell death which was prevented by RIP2 knockdown. Discussion We conclude that any intervention in caspase-1/IL-1β/IL-1R1 feedback signaling presents novel therapeutic options for the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
| | | | | | - Susanne Mohr
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
8
|
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Japp AS, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Prak ETL, Kaestner KH, Naji A, Betts MR. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590798. [PMID: 39345402 PMCID: PMC11429609 DOI: 10.1101/2024.04.23.590798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. Studies of human T1D immunobiology have predominantly focused on circulating immune cells in the blood, while mouse models suggest diabetogenic lymphocytes primarily reside in pancreas-draining lymph nodes (pLN). A comprehensive study of immune cells in human T1D was conducted using pancreas draining lymphatic tissues, including pLN and mesenteric lymph nodes, and the spleen from non-diabetic control, β cell autoantibody positive non-diabetic (AAb+), and T1D organ donors using complementary approaches of high parameter flow cytometry and CITEseq. Immune perturbations suggestive of a proinflammatory environment were specific for T1D pLN and AAb+ pLN. In addition, certain immune populations correlated with high T1D genetic risk independent of disease state. These datasets form an extensive resource for profiling human lymphatic tissue immune cells in the context of autoimmunity and T1D.
Collapse
Affiliation(s)
- Gregory J Golden
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vincent H Wu
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jacob T Hamilton
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
| | - Alberto Sada Japp
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Maria Betina Pampena
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - James J Knox
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jay S Gardner
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael R Betts
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Lara-Aguilar V, Llamas-Adán M, Brochado-Kith Ó, Crespo-Bermejo C, Grande-García S, Arca-Lafuente S, de Los Santos I, Prado C, Alía M, Sainz-Pinós C, Fernández-Rodríguez A, Martín-Carbonero L, Madrid R, Briz V. Low-level HIV-1 viremia affects T-cell activation and senescence in long-term treated adults in the INSTI era. J Biomed Sci 2024; 31:80. [PMID: 39160510 PMCID: PMC11334306 DOI: 10.1186/s12929-024-01064-z] [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: 03/27/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Around 10% of people with HIV (PWH) exhibit a low-level viremia (LLV) under antiretroviral therapy (ART). However, its origin and clinical significance are largely unknown, particularly at viremias between 50 and 200 copies/mL and under modern ART based on integrase strand transfer inhibitors (INSTIs). Our aim was to characterize their poor immune response against HIV in comparison to individuals with suppressed viremia (SV) and non-HIV controls (NHC). METHODS Transversal observational study in 81 matched participants: 27 PWH with LLV, 27 PWH with SV, and 27 NHC. Activation (CD25, HLA-DR, and CD38) and senescence [CD57, PD1, and HAVCR2 (TIM3)] were characterized in peripheral T-cell subsets by spectral flow cytometry. 45 soluble biomarkers of systemic inflammation were evaluated by immunoassays. Differences in cell frequencies and plasma biomarkers among groups were evaluated by a generalized additive model for location, scale, and shape (GAMLSS) and generalized linear model (GLM) respectively, adjusted by age, sex at birth, and ART regimen. RESULTS The median age was 53 years and 77.8% were male. Compared to NHC, PWH showed a lower CD4+/CD8+ ratio and increased activation, senescence, and inflammation, highlighting IL-13 in LLV. In addition, LLV showed a downtrend in the frequency of CD8+ naive and effector memory (EM) type 1 compared to SV, along with higher activation and senescence in CD4+ and CD8+ EM and terminally differentiated effector memory RA+ (TEMRA) subpopulations. No significant differences in systemic inflammation were observed between PWH groups. CONCLUSION LLV between 50 and 200 copies/mL leads to reduced cytotoxic activity and T-cell dysfunction that could affect cytokine production, being unable to control and eliminate infected cells. The increase in senescence markers suggests a progressive loss of immunological memory and a reduction in the proliferative capacity of immune cells. This accelerated immune aging could lead to an increased risk of developing future comorbidities. These findings strongly advocate for heightened surveillance of these PWH to promptly identify potential future complications.
Collapse
Affiliation(s)
| | - Manuel Llamas-Adán
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Óscar Brochado-Kith
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
| | | | | | - Sonia Arca-Lafuente
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Ignacio de Los Santos
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
- La Princesa University Hospital, Madrid, Spain
| | - Carmen Prado
- Flow Cytometry Unit, Institute of Health Carlos III, Madrid, Spain
| | - Mario Alía
- Flow Cytometry Unit, Institute of Health Carlos III, Madrid, Spain
| | - Coral Sainz-Pinós
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
| | - Amanda Fernández-Rodríguez
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
| | - Luz Martín-Carbonero
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain
- La Paz University Hospital (IdiPAZ), Madrid, Spain
| | | | - Verónica Briz
- National Center of Microbiology, Institute of Health Carlos III, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Alexander M, Cho E, Gliozheni E, Salem Y, Cheung J, Ichii H. Pathology of Diabetes-Induced Immune Dysfunction. Int J Mol Sci 2024; 25:7105. [PMID: 39000211 PMCID: PMC11241249 DOI: 10.3390/ijms25137105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Diabetes is associated with numerous comorbidities, one of which is increased vulnerability to infections. This review will focus on how diabetes mellitus (DM) affects the immune system and its various components, leading to the impaired proliferation of immune cells and the induction of senescence. We will explore how the pathology of diabetes-induced immune dysfunction may have similarities to the pathways of "inflammaging", a persistent low-grade inflammation common in the elderly. Inflammaging may increase the likelihood of conditions such as rheumatoid arthritis (RA) and periodontitis at a younger age. Diabetes affects bone marrow composition and cellular senescence, and in combination with advanced age also affects lymphopoiesis by increasing myeloid differentiation and reducing lymphoid differentiation. Consequently, this leads to a reduced immune system response in both the innate and adaptive phases, resulting in higher infection rates, reduced vaccine response, and increased immune cells' senescence in diabetics. We will also explore how some diabetes drugs induce immune senescence despite their benefits on glycemic control.
Collapse
Affiliation(s)
- Michael Alexander
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Eric Cho
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Eiger Gliozheni
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Yusuf Salem
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Joshua Cheung
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Hirohito Ichii
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| |
Collapse
|
13
|
Honardoost MA, Adinatha A, Schmidt F, Ranjan B, Ghaeidamini M, Arul Rayan N, Gek Liang Lim M, Joanito I, Xiao Xuan Lin Q, Rajagopalan D, Qi Mok S, Hwang YY, Larbi A, Khor CC, Foo R, Boehm BO, Prabhakar S. Systematic immune cell dysregulation and molecular subtypes revealed by single-cell RNA-seq of subjects with type 1 diabetes. Genome Med 2024; 16:45. [PMID: 38539228 PMCID: PMC10976681 DOI: 10.1186/s13073-024-01300-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 01/30/2024] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a prototypic endocrine autoimmune disease resulting from an immune-mediated destruction of pancreatic insulin-secreting β cells. A comprehensive immune cell phenotype evaluation in T1DM has not been performed thus far at the single-cell level. METHODS In this cross-sectional analysis, we generated a single-cell transcriptomic dataset of peripheral blood mononuclear cells (PBMCs) from 46 manifest T1DM (stage 3) cases and 31 matched controls. RESULTS We surprisingly detected profound alterations in circulatory immune cells (1784 dysregulated genes in 13 immune cell types), far exceeding the count in the comparator systemic autoimmune disease SLE. Genes upregulated in T1DM were involved in WNT signaling, interferon signaling and migration of T/NK cells, antigen presentation by B cells, and monocyte activation. A significant fraction of these differentially expressed genes were also altered in T1DM pancreatic islets. We used the single-cell data to construct a T1DM metagene z-score (TMZ score) that distinguished cases and controls and classified patients into molecular subtypes. This score correlated with known prognostic immune markers of T1DM, as well as with drug response in clinical trials. CONCLUSIONS Our study reveals a surprisingly strong systemic dimension at the level of immune cell network in T1DM, defines disease-relevant molecular subtypes, and has the potential to guide non-invasive test development and patient stratification.
Collapse
Affiliation(s)
- Mohammad Amin Honardoost
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
- Cardiovascular Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Andreas Adinatha
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Florian Schmidt
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Bobby Ranjan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Maryam Ghaeidamini
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Nirmala Arul Rayan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Michelle Gek Liang Lim
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Ignasius Joanito
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Quy Xiao Xuan Lin
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Deepa Rajagopalan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Shi Qi Mok
- Integrated genomics platform, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, 138648, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, 138648, Singapore
| | - Chiea Chuen Khor
- Integrated genomics platform, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Roger Foo
- Cardiovascular Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, 138673, Singapore
| | - Bernhard Otto Boehm
- Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK.
| | - Shyam Prabhakar
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
14
|
Hardtke-Wolenski M, Landwehr-Kenzel S. Tipping the balance in autoimmunity: are regulatory t cells the cause, the cure, or both? Mol Cell Pediatr 2024; 11:3. [PMID: 38507159 PMCID: PMC10954601 DOI: 10.1186/s40348-024-00176-8] [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: 10/22/2023] [Accepted: 03/07/2024] [Indexed: 03/22/2024] Open
Abstract
Regulatory T cells (Tregs) are a specialized subgroup of T-cell lymphocytes that is crucial for maintaining immune homeostasis and preventing excessive immune responses. Depending on their differentiation route, Tregs can be subdivided into thymically derived Tregs (tTregs) and peripherally induced Tregs (pTregs), which originate from conventional T cells after extrathymic differentiation at peripheral sites. Although the regulatory attributes of tTregs and pTregs partially overlap, their modes of action, protein expression profiles, and functional stability exhibit specific characteristics unique to each subset. Over the last few years, our knowledge of Treg differentiation, maturation, plasticity, and correlations between their phenotypes and functions has increased. Genetic and functional studies in patients with numeric and functional Treg deficiencies have contributed to our mechanistic understanding of immune dysregulation and autoimmune pathologies. This review provides an overview of our current knowledge of Treg biology, discusses monogenetic Treg pathologies and explores the role of Tregs in various other autoimmune disorders. Additionally, we discuss novel approaches that explore Tregs as targets or agents of innovative treatment options.
Collapse
Affiliation(s)
- Matthias Hardtke-Wolenski
- Hannover Medical School, Department of Gastroenterology Hepatology, Infectious Diseases and Endocrinology, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
- University Hospital Essen, Institute of Medical Microbiology, University Duisburg-Essen, Hufelandstraße 55, Essen, 45122, Germany
| | - Sybille Landwehr-Kenzel
- Hannover Medical School, Department of Pediatric Pneumology, Allergology and Neonatology, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
| |
Collapse
|
15
|
Wu P, Song Y, Chen Z, Xia J, Zhou Y. Changes of B cell subsets in different types of diabetes and its effect on the progression of latent autoimmune diabetes in adults. Endocrine 2024; 83:624-635. [PMID: 37755622 DOI: 10.1007/s12020-023-03539-9] [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: 06/25/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
PURPOSE Developmental abnormalities in B cells is one of the key players in autoimmune diabetes, but little is known about its role in latent autoimmune diabetes in adults (LADA). This study aimed to investigate the distribution of B cell subsets in different types of diabetes and to analyze their correlations with other biochemical parameters. METHODS A total of 140 participants were prospectively enrolled from January 2021 to December 2022. Diabetes-related autoantibodies and laboratory indicators were tested. Flow cytometry was used to analyze the percentage of circulating B cell subsets and T follicular cells. The correlation of B cell subsets with different indicators was assessed by Spearman's correlation method. RESULTS We observed that the Naïve phenotype cells tended to be less frequent in patients with diabetes than in healthy controls. The frequency of plasmablasts (PB) and Breg cell-related phenotype (B10) were significantly higher in LADA. Notably, the percentage of PB was positively associated with levels of islet cell antibody (ICA) and insulin autoantibody (IAA), but inversely associated with fasting C-peptide (FCP), further indicating that PB may promote the destruction of β-cell in patients with diabetes. CONCLUSIONS This study showed that patients with LADA had significantly altered frequencies of B cell subsets, particularly in the naïve to memory B cell ratio. Our study provided valuable information on the distribution characteristics of B cell subsets in LADA and suggested the feasibility of B-cell targeted therapy in LADA patients.
Collapse
Affiliation(s)
- Peihao Wu
- Department of Clinical Laboratory, Women's Hospital, School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Yingxiang Song
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Zhuo Chen
- Department of Clinical Laboratory, Hangzhou Lin'an Third People's Hospital, Hangzhou, Zhejiang, 311311, China
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China.
| | - Yu Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China.
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang province, No.182 Tianmushan Road, Xihu District, Hangzhou, Zhejiang, 310063, China.
| |
Collapse
|
16
|
Lelarge V, Capelle R, Oger F, Mathieu T, Le Calvé B. Senolytics: from pharmacological inhibitors to immunotherapies, a promising future for patients' treatment. NPJ AGING 2024; 10:12. [PMID: 38321020 PMCID: PMC10847408 DOI: 10.1038/s41514-024-00138-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/17/2024] [Indexed: 02/08/2024]
Abstract
The involvement of cellular senescence in the initiation and propagation of diseases is clearly characterized, making the elimination of senescent cells essential to treat age-related diseases. The development of senolytic drugs demonstrated that targeting these cells limits the deterioration of patients' condition, by inducing apoptosis. Nevertheless, the first generations of senolytics which has been developed displayed their activities through specific mechanisms and demonstrated several limitations during clinical development. However, the rational to eliminate senescent cells remains evident, with the necessity to develop specific therapies in a context of diseases and tissues. The evolutions in the field of drug discovery open the way to a new generation of senolytic therapies, such as immunological approaches (CAR-T cells, Antibody-Drug Conjugated or vaccines), which require preliminary steps of research to identify markers specifically expressed on senescent cells, demonstrating promising specific effects. Currently, the preclinical development of these strategies appears more challenging to avoid strong side effects, but the expected results are commensurate with patients' hopes for treatments. In this review, we highlight the fact that the classical senolytic approach based on drug repurposing display limited efficacy and probably reached its limits in term of clinical development. The recent development of more complex therapies and the extension of interest in the domain of senescence in different fields of research allow to extend the possibility to discover powerful therapies. The future of age-related diseases treatment is linked to the development of new approaches based on cell therapy or immunotherapy to offer the best treatment for patients.
Collapse
Affiliation(s)
- V Lelarge
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - R Capelle
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - F Oger
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1283 - UMR 8199 - EGID, 59000, Lille, France
| | - T Mathieu
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
- Synlab, 60/62 Rue d'Hauteville, 75010, Paris, France
| | - B Le Calvé
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France.
| |
Collapse
|
17
|
Christen U, Pouzol L, Tunis M, Sassi A, Tondello C, Bayer M, Hintermann E, Strasser DS, Schuldes S, Mentzel U, Martinic MM. Combination treatment of a novel CXCR3 antagonist ACT-777991 with an anti-CD3 antibody synergistically increases persistent remission in experimental models of type 1 diabetes. Clin Exp Immunol 2023; 214:131-143. [PMID: 37458220 PMCID: PMC10714188 DOI: 10.1093/cei/uxad083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/29/2023] [Accepted: 07/15/2023] [Indexed: 12/18/2023] Open
Abstract
Treatment of patients with recent-onset type 1 diabetes with an anti-CD3 antibody leads to the transient stabilization of C-peptide levels in responder patients. Partial efficacy may be explained by the entry of islet-reactive T-cells spared by and/or regenerated after the anti-CD3 therapy. The CXCR3/CXCL10 axis has been proposed as a key player in the infiltration of autoreactive T cells into the pancreatic islets followed by the destruction of β cells. Combining the blockade of this axis using ACT-777991, a novel small-molecule CXCR3 antagonist, with anti-CD3 treatment may prevent further infiltration and β-cell damage and thus, preserve insulin production. The effect of anti-CD3 treatment on circulating T-cell subsets, including CXCR3 expression, in mice was evaluated by flow cytometry. Anti-CD3/ACT-777991 combination treatment was assessed in the virally induced RIP-LCMV-GP and NOD diabetes mouse models. Treatments started at disease onset. The effects on remission rate, blood glucose concentrations, insulitis, and plasma C-peptide were evaluated for the combination treatment and the respective monotherapies. Anti-CD3 treatment induced transient lymphopenia but spared circulating CXCR3+ T cells. Combination therapy in both mouse models synergistically and persistently reduced blood glucose concentrations, resulting in increased disease remission rates compared to each monotherapy. At the study end, mice in disease remission demonstrated reduced insulitis and detectable plasma C-peptide levels. When treatments were initiated in non-severely hyperglycemic NOD mice at diabetes onset, the combination treatment led to persistent disease remission in all mice. These results provide preclinical validation and rationale to investigate the combination of ACT-777991 with anti-CD3 for the treatment of patients with recent-onset diabetes.
Collapse
Affiliation(s)
- Urs Christen
- Pharmazentrum Frankfurt, Goethe University Frankfurt, Germany
| | - Laetitia Pouzol
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Mélanie Tunis
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Anna Sassi
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | | | - Monika Bayer
- Pharmazentrum Frankfurt, Goethe University Frankfurt, Germany
| | | | - Daniel S Strasser
- Translational Biomarkers Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Sabrina Schuldes
- Project Management Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Ulrich Mentzel
- Pharmacology and Preclinical Development Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Marianne M Martinic
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| |
Collapse
|