1
|
Vaillant L, Akhter W, Nakhle J, Simon M, Villalba M, Jorgensen C, Vignais ML, Hernandez J. The role of mitochondrial transfer in the suppression of CD8 + T cell responses by Mesenchymal stem cells. Stem Cell Res Ther 2024; 15:394. [PMID: 39497203 PMCID: PMC11536934 DOI: 10.1186/s13287-024-03980-1] [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: 06/20/2024] [Accepted: 10/04/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND . CD8+ Cytotoxic T lymphocytes play a key role in the pathogenesis of autoimmune diseases and clinical conditions such as graft versus host disease and graft rejection. Mesenchymal Stromal Cells (MSCs) are multipotent cells with tissue repair and immunomodulatory capabilities. Since they are able to suppress multiple pathogenic immune responses, MSCs have been proposed as a cellular therapy for the treatment of immune-mediated diseases. However, the mechanisms underlying their immunosuppressive properties are not yet fully understood. MSCs have the remarkable ability to sense tissue injury and inflammation and respond by donating their own mitochondria to neighboring cells. Whether mitochondrial transfer has any role in the repression of CD8+ responses is unknown. METHODS AND RESULTS . We have utilized CD8+ T cells from Clone 4 TCR transgenic mice that differentiate into effector cells upon activation in vitro and in vivo to address this question. Allogeneic bone marrow derived MSCs, co-cultured with activated Clone 4 CD8+ T cells, decreased their expansion, the production of the effector cytokine IFNγ and their diabetogenic potential in vivo. Notably, we found that during this interaction leading to suppression, MSCs transferred mitochondria to CD8+ T cells as evidenced by FACS and confocal microscopy. Transfer of MSC mitochondria to Clone 4 CD8+ T cells also resulted in decreased expansion and production of IFNγ upon activation. These effects overlapped and were additive with those of prostaglandin E2 secreted by MSCs. Furthermore, preventing mitochondrial transfer in co-cultures diminished the ability of MSCs to inhibit IFNγ production. Finally, we demonstrated that both MSCs and MSC mitochondria downregulated T-bet and Eomes expression, key transcription factors for CTL differentiation, on activated CD8+ T cells. CONCLUSION . In this report we showed that MSCs are able to interact with CD8+ T cells and transfer them their mitochondria. Mitochondrial transfer contributed to the global suppressive effect of MSCs on CD8+ T cell activation by downregulating T-bet and Eomes expression resulting in impaired IFNγ production of activated CD8+ T cells.
Collapse
Affiliation(s)
- Loic Vaillant
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Waseem Akhter
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Jean Nakhle
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
- IGMM, Université de Montpellier, CNRS, Montpellier, France
| | - Matthieu Simon
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Martin Villalba
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Christian Jorgensen
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- CHU Montpellier, Montpellier, France
| | - Marie-Luce Vignais
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Javier Hernandez
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France.
| |
Collapse
|
2
|
Desai V, Shaikhsurab MZ, Varghese N, Ashtekar H. Molecular docking and network pharmacology study on active compounds of Cyprus rotundus for the treatment of diabetes mellitus. In Silico Pharmacol 2024; 12:98. [PMID: 39498162 PMCID: PMC11531456 DOI: 10.1007/s40203-024-00273-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/11/2024] [Indexed: 11/07/2024] Open
Abstract
Background Diabetes Mellitus (DM) is a complex metabolic disorder with increasing global prevalence, necessitating the exploration of novel therapeutic strategies. Cyprus rotundus, a medicinal plant with a long history of traditional use, has shown promising potential in managing DM. Aim of the study This study aims to elucidate the mechanism of action of active components of C. rotundus in managing DM using a combination of network pharmacology and molecular docking approaches. Materials and methods The active compounds of C. rotundus were identified through IMPPAT and CHEBI database mining. Subsequently, compound-target are taken from swiss target prediction and SEA. Collection of DM-related targets is done through DisGeNET and TTD database. After identifying both the targets, common targets were evaluated through venny 2.1.0. by constructing venn diagram. To elucidate the potential targets of these compounds, a protein-protein interaction network was constructed by utilizing STRING database. Through network analysis, we identified key targets and pathways involved in the pathogenesis of DM and targeted by the active components of C. rotundus. Furthermore, molecular docking was performed to explore the binding affinity and interactions between the active compounds and their target proteins. Results This, reveal that the 12 active components of C. rotundus exert their therapeutic effects on DM through multiple mechanisms, there are 141 common target genes between C. rotundus and DM. Enrichment of the KEGG pathway mainly involves in the AGE-RAGE signaling pathway in diabetic complications, Type II DM pathway. Top 10 genes were regulated by C. rotundus in DM, including MMP9, PTGS2, CASP3, CD4, EGFR, STAT3, PPARG, AKT1, NFKB1 and MAPK3. Molecular docking analysis further validates the strong binding affinity between the active compounds and their target proteins, providing insights into their mode of action at the molecular level. Conclusions This study provides a systematic understanding of the mechanism of action of C. rotundus in managing DM, offering a basis for further experimental validation and drug development.
Collapse
Affiliation(s)
- Vishakha Desai
- Department of Pharmaceutics, Rajiv Gandhi University of Health sciences, Maratha Mandal College of Pharmacy, Belgaum, 590001 Karnataka India
| | - Mohammad Ziyad Shaikhsurab
- Department of Pharmacology, Rajiv Gandhi University of Health sciences, Maratha Mandal College of Pharmacy, Belgaum, 590001 Karnataka India
| | - Nimmy Varghese
- Department of Pharmacology, NGSM Institute of Pharmaceutical SciencesNitte (Deemed to be university), Mangalore, 575018 Karnataka India
| | - Harsha Ashtekar
- Department of Pharmacology, NGSM Institute of Pharmaceutical SciencesNitte (Deemed to be university), Mangalore, 575018 Karnataka India
| |
Collapse
|
3
|
Rojas M, Acosta-Ampudia Y, Heuer LS, Zang W, M Monsalve D, Ramírez-Santana C, Anaya JM, M Ridgway W, A Ansari A, Gershwin ME. Antigen-specific T cells and autoimmunity. J Autoimmun 2024; 148:103303. [PMID: 39141985 DOI: 10.1016/j.jaut.2024.103303] [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: 07/01/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Autoimmune diseases (ADs) showcase the intricate balance between the immune system's protective functions and its potential for self-inflicted damage. These disorders arise from the immune system's erroneous targeting of the body's tissues, resulting in damage and disease. The ability of T cells to distinguish between self and non-self-antigens is pivotal to averting autoimmune reactions. Perturbations in this process contribute to AD development. Autoreactive T cells that elude thymic elimination are activated by mimics of self-antigens or are erroneously activated by self-antigens can trigger autoimmune responses. Various mechanisms, including molecular mimicry and bystander activation, contribute to AD initiation, with specific triggers and processes varying across the different ADs. In addition, the formation of neo-epitopes could also be implicated in the emergence of autoreactivity. The specificity of T cell responses centers on the antigen recognition sequences expressed by T cell receptors (TCRs), which recognize peptide fragments displayed by major histocompatibility complex (MHC) molecules. The assortment of TCR gene combinations yields a diverse array of T cell populations, each with distinct affinities for self and non-self antigens. However, new evidence challenges the traditional notion that clonal expansion solely steers the selection of higher-affinity T cells. Lower-affinity T cells also play a substantial role, prompting the "two-hit" hypothesis. High-affinity T cells incite initial responses, while their lower-affinity counterparts perpetuate autoimmunity. Precision treatments that target antigen-specific T cells hold promise for avoiding widespread immunosuppression. Nevertheless, detection of such antigen-specific T cells remains a challenge, and multiple technologies have been developed with different sensitivities while still harboring several drawbacks. In addition, elements such as human leukocyte antigen (HLA) haplotypes and validation through animal models are pivotal for advancing these strategies. In brief, this review delves into the intricate mechanisms contributing to ADs, accentuating the pivotal role(s) of antigen-specific T cells in steering immune responses and disease progression, as well as the novel strategies for the identification of antigen-specific cells and their possible future use in humans. Grasping the mechanisms behind ADs paves the way for targeted therapeutic interventions, potentially enhancing treatment choices while minimizing the risk of systemic immunosuppression.
Collapse
Affiliation(s)
- Manuel Rojas
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA; Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Luke S Heuer
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - Weici Zang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | | | - William M Ridgway
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - Aftab A Ansari
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
4
|
Espinosa-Carrasco G, Chiu E, Scrivo A, Zumbo P, Dave A, Betel D, Kang SW, Jang HJ, Hellmann MD, Burt BM, Lee HS, Schietinger A. Intratumoral immune triads are required for immunotherapy-mediated elimination of solid tumors. Cancer Cell 2024; 42:1202-1216.e8. [PMID: 38906155 PMCID: PMC11413804 DOI: 10.1016/j.ccell.2024.05.025] [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: 07/01/2023] [Revised: 03/11/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Tumor-specific CD8+ T cells are frequently dysfunctional and unable to halt tumor growth. We investigated whether tumor-specific CD4+ T cells can be enlisted to overcome CD8+ T cell dysfunction within tumors. We find that the spatial positioning and interactions of CD8+ and CD4+ T cells, but not their numbers, dictate anti-tumor responses in the context of adoptive T cell therapy as well as immune checkpoint blockade (ICB): CD4+ T cells must engage with CD8+ T cells on the same dendritic cell during the effector phase, forming a three-cell-type cluster (triad) to license CD8+ T cell cytotoxicity and cancer cell elimination. When intratumoral triad formation is disrupted, tumors progress despite equal numbers of tumor-specific CD8+ and CD4+ T cells. In patients with pleural mesothelioma treated with ICB, triads are associated with clinical responses. Thus, CD4+ T cells and triads are required for CD8+ T cell cytotoxicity during the effector phase and tumor elimination.
Collapse
Affiliation(s)
| | - Edison Chiu
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, and Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sung Wook Kang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Hee-Jin Jang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Matthew D Hellmann
- Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA; Division of Thoracic Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
5
|
Riaz F, Wei P, Pan F. PPARs at the crossroads of T cell differentiation and type 1 diabetes. Front Immunol 2023; 14:1292238. [PMID: 37928539 PMCID: PMC10623333 DOI: 10.3389/fimmu.2023.1292238] [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: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
T-cell-mediated autoimmune type 1 diabetes (T1D) is characterized by the immune-mediated destruction of pancreatic beta cells (β-cells). The increasing prevalence of T1D poses significant challenges to the healthcare system, particularly in countries with struggling economies. This review paper highlights the multifaceted roles of Peroxisome Proliferator-Activated Receptors (PPARs) in the context of T1D, shedding light on their potential as regulators of immune responses and β-cell biology. Recent research has elucidated the intricate interplay between CD4+ T cell subsets, such as Tregs and Th17, in developing autoimmune diseases like T1D. Th17 cells drive inflammation, while Tregs exert immunosuppressive functions, highlighting the delicate balance crucial for immune homeostasis. Immunotherapy has shown promise in reinstating self-tolerance and restricting the destruction of autoimmune responses, but further investigations are required to refine these therapeutic strategies. Intriguingly, PPARs, initially recognized for their role in lipid metabolism, have emerged as potent modulators of inflammation in autoimmune diseases, particularly in T1D. Although evidence suggests that PPARs affect the β-cell function, their influence on T-cell responses and their potential impact on T1D remains largely unexplored. It was noted that PPARα is involved in restricting the transcription of IL17A and enhancing the expression of Foxp3 by minimizing its proteasomal degradation. Thus, antagonizing PPARs may exert beneficial effects in regulating the differentiation of CD4+ T cells and preventing T1D. Therefore, this review advocates for comprehensive investigations to delineate the precise roles of PPARs in T1D pathogenesis, offering innovative therapeutic avenues that target both the immune system and pancreatic function. This review paper seeks to bridge the knowledge gap between PPARs, immune responses, and T1D, providing insights that may revolutionize the treatment landscape for this autoimmune disorder. Moreover, further studies involving PPAR agonists in non-obese diabetic (NOD) mice hold promise for developing novel T1D therapies.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Ping Wei
- Department of Otolaryngology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
6
|
Espinosa-Carrasco G, Scrivo A, Zumbo P, Dave A, Betel D, Hellmann M, Burt BM, Lee HS, Schietinger A. Intratumoral immune triads are required for adoptive T cell therapy-mediated elimination of solid tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547423. [PMID: 37461721 PMCID: PMC10349998 DOI: 10.1101/2023.07.03.547423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Tumor-reactive CD8 T cells found in cancer patients are frequently dysfunctional, unable to halt tumor growth. Adoptive T cell transfer (ACT), the administration of large numbers of in vitro-generated cytolytic tumor-reactive CD8 T cells, is an important cancer immune therapy being pursued. However, a limitation of ACT is that transferred CD8 T cells often rapidly lose effector function, and despite exciting results in certain malignancies, few ACT clinical trials have shown responses in solid tumors. Here, we developed preclinical cancer mouse models to investigate if and how tumor-specific CD4 T cells can be enlisted to overcome CD8 T cell dysfunction in the setting of ACT. In situ confocal microscopy of color-coded cancer cells, tumor-specific CD8 and CD4 T cells, and antigen presenting cells (APC), combined with functional studies, revealed that the spatial positioning and interactions of CD8 and CD4 T cells, but not their numbers, dictates ACT efficacy and anti-tumor responses. We uncover a new role of antigen-specific CD4 T cells in addition to the known requirement for CD4 T cells during priming/activation of naïve CD8 T cells. CD4 T cells must co-engage with CD8 T cells and APC cross-presenting CD8- and CD4-tumor antigens during the effector phase, forming a three-cell-cluster (triad), to license CD8 T cell cytotoxicity and mediate cancer cell elimination. Triad formation transcriptionally and epigenetically reprogram CD8 T cells, prevent T cell dysfunction/exhaustion, and ultimately lead to the elimination of large established tumors and confer long-term protection from recurrence. When intratumoral triad formation was disrupted, adoptively transferred CD8 T cells could not be reprogrammed, and tumors progressed despite equal numbers of tumor-infiltrating CD8 and CD4 T cells. Strikingly, the formation of CD4 T cell::CD8 T cell::APC triads in tumors of patients with lung cancers treated with immune checkpoint blockade was associated with clinical responses, but not CD4::APC dyads or overall numbers of CD8 or CD4 T cells, demonstrating the importance of triads in non-ACT settings in humans. Our work uncovers intratumoral triads as a key requirement for anti-tumor immunity and a new role for CD4 T cells in CD8 T cell cytotoxicity and cancer cell eradication.
Collapse
Affiliation(s)
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, and Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doron Betel
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew Hellmann
- Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| |
Collapse
|
7
|
Rai U, Senapati D, Arora MK. Insights on the role of anti-inflammatory and immunosuppressive agents in the amelioration of diabetes. Diabetol Int 2023; 14:134-144. [PMID: 37090130 PMCID: PMC10113422 DOI: 10.1007/s13340-022-00607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Diabetes is a major health problem worldwide. It is a chronic metabolic disorder that produces overt hyperglycemic condition that occurs either when the pancreas does not produce enough insulin due to excessive destruction of pancreatic β-cells (type 1 diabetes) or due to development of insulin resistance (type 2 diabetes). An autoimmune condition known as type 1 diabetes (T1D) results in the targeted immune death of β-cells that produce insulin. The only available treatment for T1D at the moment is the lifelong use of insulin. Multiple islet autoantibody positivity is used to diagnose T1D. There are four standard autoantibodies observed whose presence shows the development of T1D: antibodies against insulin, glutamic acid decarboxylase (GAD65), zinc T8 transporter (ZnT8), and tyrosine phosphatase-like protein (ICA512). In type 2 diabetes (T2D), an inflammatory response precipitates as a consequence of the immune response to high blood glucose level along with the presence of inflammation mediators produced by macrophages and adipocytes in fat tissue. The slow and chronic inflammatory condition of adipose tissue produces insulin resistance leading to increased stress on pancreatic β-cells to produce more insulin to compensate for the insulin resistance. Thus, this stress condition exacerbates the apoptosis of β-cells leading to insufficient production of insulin, resulting in hyperglycemia which signifies late stage T2D. Therefore, the therapeutic utilization of immunosuppressive agents may be a better alternative over the use of insulin and oral hypoglycemic agents for the treatment of T1D and T2D, respectively. This review enlightens the immune intervention for the prevention and amelioration of T1D and T2D in humans with main focus on the antigen-specific immune suppressive therapy.
Collapse
Affiliation(s)
- Uddipak Rai
- School of Pharmaceutical and Population Health Informatics, DIT University, 248009, Dehradun, Uttarakhand India
| | - Dhirodatta Senapati
- School of Pharmaceutical and Population Health Informatics, DIT University, 248009, Dehradun, Uttarakhand India
| | - Mandeep Kumar Arora
- School of Pharmaceutical and Population Health Informatics, DIT University, 248009, Dehradun, Uttarakhand India
| |
Collapse
|
8
|
Dludla PV, Mabhida SE, Ziqubu K, Nkambule BB, Mazibuko-Mbeje SE, Hanser S, Basson AK, Pheiffer C, Kengne AP. Pancreatic β-cell dysfunction in type 2 diabetes: Implications of inflammation and oxidative stress. World J Diabetes 2023; 14:130-146. [PMID: 37035220 PMCID: PMC10075035 DOI: 10.4239/wjd.v14.i3.130] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/26/2022] [Accepted: 02/28/2023] [Indexed: 03/15/2023] Open
Abstract
Insulin resistance and pancreatic β-cell dysfunction are major pathological mechanisms implicated in the development and progression of type 2 diabetes (T2D). Beyond the detrimental effects of insulin resistance, inflammation and oxidative stress have emerged as critical features of T2D that define β-cell dysfunction. Predominant markers of inflammation such as C-reactive protein, tumor necrosis factor alpha, and interleukin-1β are consistently associated with β-cell failure in preclinical models and in people with T2D. Similarly, important markers of oxidative stress, such as increased reactive oxygen species and depleted intracellular antioxidants, are consistent with pancreatic β-cell damage in conditions of T2D. Such effects illustrate a pathological relationship between an abnormal inflammatory response and generation of oxidative stress during the progression of T2D. The current review explores preclinical and clinical research on the patho-logical implications of inflammation and oxidative stress during the development of β-cell dysfunction in T2D. Moreover, important molecular mechanisms and relevant biomarkers involved in this process are discussed to divulge a pathological link between inflammation and oxidative stress during β-cell failure in T2D. Underpinning the clinical relevance of the review, a systematic analysis of evidence from randomized controlled trials is covered, on the potential therapeutic effects of some commonly used antidiabetic agents in modulating inflammatory makers to improve β-cell function.
Collapse
Affiliation(s)
- Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
| | - Sihle E Mabhida
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town 7505, South Africa
| | - Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | | | - Sidney Hanser
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga 0727, South Africa
| | - Albert Kotze Basson
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town 7505, South Africa
| | - Andre Pascal Kengne
- Department of Medicine, University of Cape Town, Cape Town 7500, South Africa
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg 7505, South Africa
| |
Collapse
|
9
|
Akhter W, Nakhle J, Vaillant L, Garcin G, Le Saout C, Simon M, Crozet C, Djouad F, Jorgensen C, Vignais ML, Hernandez J. Transfer of mesenchymal stem cell mitochondria to CD4 + T cells contributes to repress Th1 differentiation by downregulating T-bet expression. Stem Cell Res Ther 2023; 14:12. [PMID: 36694226 PMCID: PMC9875419 DOI: 10.1186/s13287-022-03219-x] [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: 05/09/2022] [Accepted: 12/08/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSCs) are multipotent cells with strong tissue repair and immunomodulatory properties. Due to their ability to repress pathogenic immune responses, and in particular T cell responses, they show therapeutic potential for the treatment of autoimmune diseases, organ rejection and graft versus host disease. MSCs have the remarkable ability to export their own mitochondria to neighboring cells in response to injury and inflammation. However, whether mitochondrial transfer occurs and has any role in the repression of CD4+ Th1 responses is unknown. METHODS AND RESULTS In this report we have utilized CD4+ T cells from HNT TCR transgenic mice that develop Th1-like responses upon antigenic stimulation in vitro and in vivo. Allogeneic bone marrow-derived MSCs reduced the diabetogenic potential of HNT CD4+ T cells in vivo in a transgenic mouse model of disease. In co-culture experiments, we have shown that MSCs were able to reduce HNT CD4+ T cell expansion, expression of key effector markers and production of the effector cytokine IFNγ after activation. This was associated with the ability of CD4+ T cells to acquire mitochondria from MSCs as evidenced by FACS and confocal microscopy. Remarkably, transfer of isolated MSC mitochondria to CD4+ T cells resulted in decreased T cell proliferation and IFNγ production. These effects were additive with those of prostaglandin E2 secreted by MSCs. Finally, we demonstrated that both co-culture with MSCs and transfer of isolated MSC mitochondria prevent the upregulation of T-bet, the master Th1 transcription factor, on activated CD4+ T cells. CONCLUSION The present study demonstrates that transfer of MSC mitochondria to activated CD4+ T cells results in the suppression of Th1 responses in part by downregulating T-bet expression. Furthermore, our studies suggest that MSC mitochondrial transfer might represent a general mechanism of MSC-dependent immunosuppression.
Collapse
Affiliation(s)
- Waseem Akhter
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France
| | - Jean Nakhle
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France ,grid.121334.60000 0001 2097 0141IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France ,grid.121334.60000 0001 2097 0141IGMM, CNRS, Université de Montpellier, Montpellier, France
| | - Loïc Vaillant
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France
| | - Geneviève Garcin
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France
| | - Cécile Le Saout
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France
| | - Matthieu Simon
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France
| | - Carole Crozet
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France ,grid.121334.60000 0001 2097 0141INM, INSERM, Université de Montpellier, Montpellier, France
| | - Farida Djouad
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France
| | - Christian Jorgensen
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France ,grid.157868.50000 0000 9961 060XCHU Montpellier, Montpellier, France
| | - Marie-Luce Vignais
- grid.121334.60000 0001 2097 0141Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295 Montpellier, France ,grid.121334.60000 0001 2097 0141IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Javier Hernandez
- Institute for Regenerative Medicine and Biotherapy (IRMB), INSERM U1183, Université de Montpellier, 34295, Montpellier, France.
| |
Collapse
|
10
|
Valta M, Yoshihara M, Einarsdottir E, Pahkuri S, Ezer S, Katayama S, Knip M, Veijola R, Toppari J, Ilonen J, Kere J, Lempainen J. Viral infection-related gene upregulation in monocytes in children with signs of β-cell autoimmunity. Pediatr Diabetes 2022; 23:703-713. [PMID: 35419920 PMCID: PMC9545759 DOI: 10.1111/pedi.13346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/24/2022] [Accepted: 04/08/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The pathogenesis of type 1 diabetes (T1D) is associated with genetic predisposition and immunological changes during presymptomatic disease. Differences in immune cell subset numbers and phenotypes between T1D patients and healthy controls have been described; however, the role and function of these changes in the pathogenesis is still unclear. Here we aimed to analyze the transcriptomic landscapes of peripheral blood mononuclear cells (PBMCs) during presymptomatic disease. METHODS Transcriptomic differences in PBMCs were compared between cases positive for islet autoantibodies and autoantibody negative controls (9 case-control pairs) and further in monocytes and lymphocytes separately in autoantibody positive subjects and control subjects (25 case-control pairs). RESULTS No significant differential expression was found in either data set. However, when gene set enrichment analysis was performed, the gene sets "defence response to virus" (FDR <0.001, ranking 2), "response to virus" (FDR <0.001, ranking 3) and "response to type I interferon" (FDR = 0.002, ranking 12) were enriched in the upregulated genes among PBMCs in cases. Upon further analysis, this was also seen in monocytes in cases (FDR = 0.01, ranking 2; FDR = 0.04, ranking 3 and FDR = 0.02, ranking 1, respectively) but not in lymphocytes. CONCLUSION Gene set enrichment analysis of children with T1D-associated autoimmunity revealed changes in pathways relevant for virus infection in PBMCs, particularly in monocytes. Virus infections have been repeatedly implicated in the pathogenesis of T1D. These results support the viral hypothesis by suggesting altered immune activation of viral immune pathways in monocytes during diabetes.
Collapse
Affiliation(s)
- Milla Valta
- Immunogenetics Laboratory, Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Masahito Yoshihara
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene TechnologyKTH‐Royal Institute of TechnologySolnaSweden
| | - Sirpa Pahkuri
- Immunogenetics Laboratory, Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Sini Ezer
- Stem Cells and Metabolism Research ProgramUniversity of Helsinki, and Folkhälsan Research CenterHelsinkiFinland
| | - Shintaro Katayama
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden,Stem Cells and Metabolism Research ProgramUniversity of Helsinki, and Folkhälsan Research CenterHelsinkiFinland
| | - Mikael Knip
- Pediatric Research Center, Children's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland,Research Program for Clinical and Molecular MetabolismFaculty of Medicine, University of HelsinkiHelsinkiFinland,Folkhälsan Research CenterHelsinkiFinland,Department of PediatricsTampere University HospitalTampereFinland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, MRC OuluOulu University Hospital and University of OuluOuluFinland
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrative Physiology and PharmacologyUniversity of TurkuTurkuFinland,Department of PediatricsUniversity of Turku and Turku University HospitalTurkuFinland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Juha Kere
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden,Stem Cells and Metabolism Research ProgramUniversity of Helsinki, and Folkhälsan Research CenterHelsinkiFinland
| | - Johanna Lempainen
- Immunogenetics Laboratory, Institute of BiomedicineUniversity of TurkuTurkuFinland,Department of PediatricsUniversity of Turku and Turku University HospitalTurkuFinland,Clinical MicrobiologyTurku University HospitalTurkuFinland
| |
Collapse
|
11
|
Michau A, Lafont C, Bargi-Souza P, Kemkem Y, Guillou A, Ravier MA, Bertrand G, Varrault A, Fiordelisio T, Hodson DJ, Mollard P, Schaeffer M. Metabolic Stress Impairs Pericyte Response to Optogenetic Stimulation in Pancreatic Islets. Front Endocrinol (Lausanne) 2022; 13:918733. [PMID: 35813647 PMCID: PMC9259887 DOI: 10.3389/fendo.2022.918733] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic islets are highly vascularized micro-organs ensuring whole body glucose homeostasis. Islet vascular cells play an integral part in sustaining adequate insulin release by beta cells. In particular, recent studies have demonstrated that islet pericytes regulate local blood flow velocity and are required for maintenance of beta cell maturity and function. In addition, increased metabolic demand accompanying obesity alters islet pericyte morphology. Here, we sought to explore the effects of metabolic stress on islet pericyte functional response to stimulation in a mouse model of type 2 diabetes, directly in the pancreas in vivo . We found that high fat diet induced islet pericyte hypertrophy without alterations in basal local blood flow. However, optogenetic stimulation of pericyte activity revealed impaired islet vascular responses, despite increased expression of genes encoding proteins directly or indirectly involved in cell contraction. These findings suggest that metabolic stress impinges upon islet pericyte function, which may contribute to beta cell failure during T2D.
Collapse
Affiliation(s)
- Aurélien Michau
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Chrystel Lafont
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Paula Bargi-Souza
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- Department of Physiology and Biophysics of the Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Yasmine Kemkem
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Guillou
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Magalie A. Ravier
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Gyslaine Bertrand
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Annie Varrault
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Tatiana Fiordelisio
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- Laboratorio de Neuroendocrinología Comparada, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David J. Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Patrice Mollard
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- Centre de Biologie Structurale, CNRS UMR 5048, INSERM U1054, Univ Montpellier, Montpellier, France
- *Correspondence: Marie Schaeffer,
| |
Collapse
|
12
|
The Emerging Roles of T Helper Cell Subsets and Cytokines in Severe Neutrophilic Asthma. Inflammation 2021; 45:1007-1022. [PMID: 34825300 DOI: 10.1007/s10753-021-01598-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/01/2021] [Accepted: 11/07/2021] [Indexed: 12/11/2022]
Abstract
Neutrophilic asthma (NA) is a severe type of steroid resistant asthma, and so far the immune mechanisms underlying NA are not clear. In this article, we performed a comprehensive assessment of Th-cell subsets and cytokines in severe NA patients. A total of 13 healthy individuals and 31 severe asthma patients were enrolled in this study. Refractory asthma patients were defined as those with eosinophilic asthma (EA, accounted for 32% of asthmatic patients) or NA (68%) according to sputum neutrophil/eosinophil counts or blood eosinophils. Th-cell subsets in peripheral blood mononuclear cells (PBMCs) were analyzed by flow cytometry, and cytokines were detected by cytometric bead array (CBA). The results showed significant differences were observed in Th-cell phenotypes, where the number of Th1 cells were reduced and the numbers of Th2 cells were increased in NA and EA groups, respectively, when compared with healthy controls. Th17 cells were not strongly associated with severe neutrophilic asthma. The frequencies of mucosal-associated invariant T (MAIT) cells were strikingly reduced in severe asthma patients, especially in the NA group. This NA group also showed increased levels of IL-17A, IL-17F, TNF-α, and IL-6 in serum and increased levels of IL-17A, IL-17F, IFN-γ, TNF-α, IL-1β, IL-5, IL-6, and IL-8 in sputum. In addition, sputum IL-6 was positively correlated with TNF-α, IFN-γ, IL-17A, and IL-8. Our results uncovered a controversial role for Th17 cells, which were reduced in severe asthma patients. Severe neutrophilic asthma was associated with a striking deficiency of MAIT cells and high pro-inflammatory cytokine levels.
Collapse
|
13
|
Rodriguez-Calvo T, Johnson JD, Overbergh L, Dunne JL. Neoepitopes in Type 1 Diabetes: Etiological Insights, Biomarkers and Therapeutic Targets. Front Immunol 2021; 12:667989. [PMID: 33953728 PMCID: PMC8089389 DOI: 10.3389/fimmu.2021.667989] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms underlying type 1 diabetes (T1D) pathogenesis remain largely unknown. While autoantibodies to pancreatic beta-cell antigens are often the first biological response and thereby a useful biomarker for identifying individuals in early stages of T1D, their role in T1D pathogenesis is not well understood. Recognition of these antigenic targets by autoreactive T-cells plays a pathological role in T1D development. Recently, several beta-cell neoantigens have been described, indicating that both neoantigens and known T1D antigens escape central or peripheral tolerance. Several questions regarding the mechanisms by which tolerance is broken in T1D remain unanswered. Further delineating the timing and nature of antigenic responses could allow their use as biomarkers to improve staging, as targets for therapeutic intervention, and lead to a better understanding of the mechanisms leading to loss of tolerance. Multiple factors that contribute to cellular stress may result in the generation of beta-cell derived neoepitopes and contribute to autoimmunity. Understanding the cellular mechanisms that induce beta-cells to produce neoantigens has direct implications on development of therapies to intercept T1D disease progression. In this perspective, we will discuss evidence for the role of neoantigens in the pathogenesis of T1D, including antigenic responses and cellular mechanisms. We will additionally discuss the pathways leading to neoepitope formation and the cross talk between the immune system and the beta-cells in this regard. Ultimately, delineating the timing of neoepitope generation in T1D pathogenesis will determine their role as biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Teresa Rodriguez-Calvo
- Institute of Diabetes Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Munich, Germany
| | - James D. Johnson
- Diabetes Research Group, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Lut Overbergh
- Laboratory Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Jessica L. Dunne
- Janssen Research and Development, LLC, Raritan, NJ, United States
| |
Collapse
|
14
|
Thomas R, Oh J, Wang W, Su DM. Thymic atrophy creates holes in Treg-mediated immuno-regulation via impairment of an antigen-specific clone. Immunology 2021; 163:478-492. [PMID: 33786850 DOI: 10.1111/imm.13333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/14/2022] Open
Abstract
Age-related thymic atrophy results in reduced output of naïve conventional T (Tcon) cells. However, its impact on regulatory T (Treg) cells is insufficiently understood. Given evidence that thymic Treg (tTreg) cell generation is enhanced in the aged, atrophy thymus and that the aged periphery accumulates peripheral Treg (pTreg) cells, we asked why these Treg cells are unable to effectively attenuate increased autoreactivity-induced chronic inflammation in the elderly. We designed a mock-self-antigen chimera mouse model, in which membrane-bound ovalbumin (mOVA) transgenic mice, bearing a FoxN1-floxed gene for induction of conditional thymic atrophy, received OVA-specific (OT-II) T-cell receptor (TCR) transgenic progenitor cells. The chimeric mice with thymic atrophy exhibited a significant decrease in OVA-specific tTreg and pTreg cells but not polyclonal (pan)-Treg cells. These OVA-specific pTreg cells were significantly less able to suppress OVA-specific stimulation-induced proliferation in vitro and exhibited lower FoxP3 expression. Additionally, we conducted preliminary TCR repertoire diversity sequencing for Treg cells among recent thymic emigrants (RTEs) from RagGFP -FoxP3RFP dual-reporter mice and observed a trend for decreased diversity in mice with thymic atrophy compared to littermates with normal thymus. These data indicate that although the effects of age-related thymic atrophy do not affect pan-Treg generation, certain tissue-specific Treg clones may experience abnormal agonist selection. This, combined with enhanced pan-pTreg cells, may greatly contribute to age-related chronic inflammation, even in the absence of acute autoimmune disease in the elderly.
Collapse
Affiliation(s)
- Rachel Thomas
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Jiyoung Oh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weikan Wang
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Dong-Ming Su
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
15
|
Hu W, Song X, Yu H, Sun J, Wang H, Zhao Y. Clinical Translational Potentials of Stem Cell-Derived Extracellular Vesicles in Type 1 Diabetes. Front Endocrinol (Lausanne) 2021; 12:682145. [PMID: 35095751 PMCID: PMC8789747 DOI: 10.3389/fendo.2021.682145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific disease characterized by the deficiency of insulin caused by the autoimmune destruction of pancreatic islet β cells. Stem cell-based therapies play essential roles in immunomodulation and tissue regeneration, both of which hold great promise for treating many autoimmune dysfunctions. However, their clinical translational potential has been limited by ethical issues and cell transplant rejections. Exosomes are small extracellular vesicles (EVs) released by almost all types of cells, performing a variety of cell functions through the delivery of their molecular contents such as proteins, DNAs, and RNAs. Increasing evidence suggests that stem cell-derived EVs exhibit similar functions as their parent cells, which may represent novel therapeutic agents for the treatment of autoimmune diseases including T1D. In this review, we summarize the current research progresses of stem cell-derived EVs for the treatment of T1D.
Collapse
Affiliation(s)
- Wei Hu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Xiang Song
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Haibo Yu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Jingyu Sun
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ, United States
| | - Hongjun Wang
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ, United States
| | - Yong Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
- Throne Biotechnologies Inc., Paramus, NJ, United States
- *Correspondence: Yong Zhao,
| |
Collapse
|
16
|
Bender C, Rajendran S, von Herrath MG. New Insights Into the Role of Autoreactive CD8 T Cells and Cytokines in Human Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:606434. [PMID: 33469446 PMCID: PMC7813992 DOI: 10.3389/fendo.2020.606434] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/20/2020] [Indexed: 12/31/2022] Open
Abstract
Since the establishment of the network for pancreatic organ donors with diabetes (nPOD), we have gained unprecedented insight into the pathology of human type 1 diabetes. Many of the pre-existing "dogmas", mostly derived from studies of animal models and sometimes limited human samples, have to be revised now. For example, we have learned that autoreactive CD8 T cells are present even in healthy individuals within the exocrine pancreas. Furthermore, their "attraction" to islets probably relies on beta-cell intrinsic events, such as the over-expression of MHC class I and resulting presentation of autoantigens such as (prepro)insulin. In addition, we are discovering other signs of beta-cell dysfunction, possibly at least in part due to stress, such as the over-expression of certain cytokines. This review summarizes the latest developments focusing on cytokines and autoreactive CD8 T cells in human type 1 diabetes pathogenesis.
Collapse
|
17
|
Baharlou H, Canete NP, Cunningham AL, Harman AN, Patrick E. Mass Cytometry Imaging for the Study of Human Diseases-Applications and Data Analysis Strategies. Front Immunol 2019; 10:2657. [PMID: 31798587 PMCID: PMC6868098 DOI: 10.3389/fimmu.2019.02657] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/28/2019] [Indexed: 01/09/2023] Open
Abstract
High parameter imaging is an important tool in the life sciences for both discovery and healthcare applications. Imaging Mass Cytometry (IMC) and Multiplexed Ion Beam Imaging (MIBI) are two relatively recent technologies which enable clinical samples to be simultaneously analyzed for up to 40 parameters at subcellular resolution. Importantly, these "Mass Cytometry Imaging" (MCI) modalities are being rapidly adopted for studies of the immune system in both health and disease. In this review we discuss, first, the various applications of MCI to date. Second, due to the inherent challenge of analyzing high parameter spatial data, we discuss the various approaches that have been employed for the processing and analysis of data from MCI experiments.
Collapse
Affiliation(s)
- Heeva Baharlou
- The Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Nicolas P. Canete
- The Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Ellis Patrick
- The Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Italiya KS, Basak M, Mazumdar S, Sahel DK, Shrivastava R, Chitkara D, Mittal A. Scalable Self-Assembling Micellar System for Enhanced Oral Bioavailability and Efficacy of Lisofylline for Treatment of Type-I Diabetes. Mol Pharm 2019; 16:4954-4967. [DOI: 10.1021/acs.molpharmaceut.9b00833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Kishan S. Italiya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Moumita Basak
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Samrat Mazumdar
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Deepak K. Sahel
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Richa Shrivastava
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani, Rajasthan 333031, India
| |
Collapse
|
19
|
Abraham TS, Flickinger JC, Waldman SA, Snook AE. TCR Retrogenic Mice as a Model To Map Self-Tolerance Mechanisms to the Cancer Mucosa Antigen GUCY2C. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:1301-1310. [PMID: 30642983 PMCID: PMC6363846 DOI: 10.4049/jimmunol.1801206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/13/2018] [Indexed: 01/21/2023]
Abstract
Characterizing self-tolerance mechanisms and their failure is critical to understand immune homeostasis, cancer immunity, and autoimmunity. However, examination of self-tolerance mechanisms has relied primarily on transgenic mice expressing TCRs targeting well-characterized, but nonphysiologic, model Ags, such as OVA and hemagglutinin. Identifying TCRs directed against bona fide self-antigens is made difficult by the extraordinary diversity of TCRs and the low prevalence of Ag-specific clones (<10-100 naive cells per organism), limiting dissection of tolerance mechanisms restricting immunity to self-proteins. In this study, we isolated and characterized TCRs recognizing the intestinal epithelial cell receptor and colorectal cancer Ag GUCY2C to establish a model to study self-antigen-specific tolerance mechanisms. GUCY2C-specific CD4+ effector T cells were isolated from immunized, nontolerant Gucy2c -/- mice. Next-generation sequencing identified GUCY2C-specific TCRs, which were engineered into CD4+ T cells in vitro to confirm TCR recognition of GUCY2C. Further, the generation of "retrogenic" mice by reconstitution with TCR-transduced hematopoietic stem cells resulted in normal CD4+ T cell development, responsiveness to immunization, and GUCY2C-induced tolerance in recipient mice, recapitulating observations in conventional models. This retrogenic model can be employed to define self-tolerance mechanisms restricting T and B cell responses to GUCY2C to optimize colorectal cancer immunotherapy without autoimmunity.
Collapse
Affiliation(s)
- Tara S Abraham
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| | - John C Flickinger
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
20
|
Espinosa-Carrasco G, Le Saout C, Fontanaud P, Michau A, Mollard P, Hernandez J, Schaeffer M. Integrin β1 Optimizes Diabetogenic T Cell Migration and Function in the Pancreas. Front Immunol 2018; 9:1156. [PMID: 29904378 PMCID: PMC5990596 DOI: 10.3389/fimmu.2018.01156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/08/2018] [Indexed: 01/08/2023] Open
Abstract
T cell search behavior is dictated by their need to encounter their specific antigen to eliminate target cells. However, mechanisms controlling effector T cell motility are highly tissue-dependent. Specifically, how diabetogenic T cells encounter their target beta cells in dispersed islets throughout the pancreas (PA) during autoimmune diabetes remains unclear. Using intra-vital 2-photon microscopy in a mouse model of diabetes, we found that CXCR3 chemokine downregulated CD8+ T cell motility specifically within islets, promoting effector cell confinement to their target sites. By contrast, T cell velocity and directionality in the exocrine tissue were enhanced along blood vessels and extracellular matrix fibers. This guided migration implicated integrin-dependent interactions, since integrin blockade impaired exocrine T cell motility. In addition, integrin β1 blockade decreased CD4+ T cell effector phenotype specifically in the PA. Thus, we unveil an important role for integrins in the PA during autoimmune diabetes that may have important implications for the design of new therapies.
Collapse
Affiliation(s)
- Gabriel Espinosa-Carrasco
- INSERM U1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Montpellier, France.,Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Cécile Le Saout
- INSERM U1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Montpellier, France
| | - Pierre Fontanaud
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Aurélien Michau
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Patrice Mollard
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Javier Hernandez
- INSERM U1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
21
|
Ahrends T, Borst J. The opposing roles of CD4 + T cells in anti-tumour immunity. Immunology 2018; 154:582-592. [PMID: 29700809 PMCID: PMC6050207 DOI: 10.1111/imm.12941] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy focuses mainly on anti-tumour activity of CD8+ cytotoxic T lymphocytes (CTLs). CTLs can directly kill all tumour cell types, provided they carry recognizable antigens. However, CD4+ T cells also play important roles in anti-tumour immunity. CD4+ T cells can either suppress or promote the anti-tumour CTL response, either in secondary lymphoid organs or in the tumour. In this review, we highlight opposing mechanisms of conventional and regulatory T cells at both sites. We outline how current cancer immunotherapy strategies affect both subsets and how selective modulation of each subset is important to maximize the clinical response of cancer patients.
Collapse
Affiliation(s)
- Tomasz Ahrends
- Division of Tumour Biology and ImmunologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jannie Borst
- Division of Tumour Biology and ImmunologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|