1
|
Casso-Chapa B, González NAV, Le NT, Palaskas NL, Nead KT, Eutsey LP, Samanthapudi VSK, Osborn AM, Lee J, Mejia G, Hoang O, Lin SH, Deswal A, Herrmann J, Wang G, Kirkland JL, Krishnan S, Wehrens XH, Chini EN, Yusuf SW, Iliescu CA, Jain A, Burks JK, Seeley E, Lorenzi PL, Chau KM, Mendoza KCO, Grumbach IM, Brookes PS, Hanssen NM, de Winther MP, Yvan-Charvet L, Kotla S, Schadler K, Abe JI. Reevaluating Anti-Inflammatory Therapy: Targeting Senescence to Balance Anti-Cancer Efficacy and Vascular Disease. Arterioscler Thromb Vasc Biol 2025; 45:372-385. [PMID: 39817327 PMCID: PMC11864897 DOI: 10.1161/atvbaha.124.319870] [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: 06/18/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025]
Abstract
Modulating immune function is a critical strategy in cancer and atherosclerosis treatments. For cancer, boosting or maintaining the immune system is crucial to prevent tumor growth. However, in vascular disease, mitigating immune responses can decrease inflammation and slow atherosclerosis progression. Anti-inflammatory therapy, therefore, presents a unique dilemma for cancer survivors: while it may decrease cardiovascular risk, it might also promote cancer growth and metastasis by suppressing the immune response. Senescence presents a potentially targetable solution to this challenge; senescence increases the risk of both cancer therapy resistance and vascular disease. Exercise, notably, shows promise in delaying this premature senescence, potentially improving cancer outcomes and lowering vascular disease risk post-treatment. This review focuses on the long-term impact of cancer therapies on vascular health. We underscore the importance of modulating senescence to balance cancer treatment's effectiveness and its vascular impact, and we emphasize investigating the role of exercise-mediated suppression of senescence in improving cancer survivorship.
Collapse
Affiliation(s)
- Bernardo Casso-Chapa
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Norma Alicia Vazquez González
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kevin T. Nead
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lydia P. Eutsey
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Cancer Center Support Grant & Extramural Research Development, UT MD Anderson Cancer Center, Houston, TX
| | | | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonghae Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gilbert Mejia
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Oanh Hoang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xander H.T. Wehrens
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cezar A. Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jared K. Burks
- Department of Leukemia, Division of Center Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Erin Seeley
- Department of Chemistry, University of Texas at Austin, Austin, Texas, USA
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khanh M. Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Keila Carolina Ostos Mendoza
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | | | - Paul S. Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Nordin M.J. Hanssen
- Department of (Experimental) Vascular and Internal Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Immunology and Infectious Diseases (AII), Inflammatory Diseases Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keri Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Abbott V, Housden BE, Houldsworth A. Could immunotherapy and regulatory T cells be used therapeutically to slow the progression of Alzheimer's disease? Brain Commun 2025; 7:fcaf092. [PMID: 40078868 PMCID: PMC11896979 DOI: 10.1093/braincomms/fcaf092] [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: 07/16/2024] [Revised: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease and other cognitive impairments are a growing problem in the healthcare world with the ageing population. There are currently no effective treatments available; however, it has been suggested that targeting neuroinflammation may be a successful approach in slowing the progression of neurodegeneration. Reducing the destructive hyperinflammatory pathology to maintain homeostasis in neural tissue is a promising option to consider. This review explores the mechanisms behind neuroinflammation and the effectiveness of immunotherapy in slowing the progression of cognitive decline in patients with Alzheimer's disease. The key components of neuroinflammation in Alzheimer's disease researched are microglia, astrocytes, cytokines and CD8+ effector T cells. The role of oxidative stress on modulating regulatory T cells and some of the limitations of regulatory T cell-based therapies are also explored. Increasing regulatory T cells can decrease activation of microglia, proinflammatory cytokines and astrocytes; however, it can also increase levels of inflammatory cytokines. There is a complex network of regulatory T cell interactions that reduce Alzheimer's disease pathology, which is not fully understood. Exploring the current literature, further research into the use of immunotherapy in Alzheimer's disease is vital to determine the potential of these techniques; however, there is sufficient evidence to suggest that increasing regulatory T cells count does prevent Alzheimer's disease symptoms and pathology in patients with Alzheimer's disease. Some exciting innovative therapies are muted to explore in the future. The function of regulatory T cells in the presence of reactive oxygen species and oxidative stress should be investigated further in patients with neurogenerative disorders to ascertain if combination therapies could reduce oxidative stress while also enhancing regulatory T cells function. Could methods of immunotherapy infuse exogenous functional Tregs or enhance the immune environment in favour of endogenous regulatory T cells differentiation, thus reducing neuroinflammation in neurodegenerative pathology, inhibiting the progression of Alzheimer's disease?
Collapse
Affiliation(s)
- Victoria Abbott
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| | - Benjamin E Housden
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Annwyne Houldsworth
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| |
Collapse
|
3
|
Khatami SH, Alehossein P, Ehtiati S, Zarei T, Salmani F, Bagherzadeh S, Razmafrooz M, Rajabibazl M, Halimi A, Shahmohammadi MR, Jouibari MF, Tafakhori A, Karima S. Therapeutic Efficacy of Intermittent Ketogenesis in Modulating Adenosine Metabolism, Immune Response, and Seizure Severity in Refractory Temporal Lobe Epilepsy: A Pilot Human Study. Inflammation 2025:10.1007/s10753-025-02264-x. [PMID: 39920557 DOI: 10.1007/s10753-025-02264-x] [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: 12/08/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Temporal lobe epilepsy (TLE) is a common neurological disorder characterized by recurrent seizures originating in the temporal lobe, often affecting patients' physical, cognitive, and social well-being. Despite the availability of antiseizure medication (ASMs), approximately 30% of TLE patients exhibit drug-resistant seizures, emphasizing the need for alternative therapeutic approaches. Ketogenic diets, known for their anticonvulsant effects, have shown promise in managing drug-resistant epilepsy. However, their demanding high-fat, low-carbohydrate regimens pose significant adherence challenges. Medium-chain triglyceride (MCT) offers a viable alternative by inducing ketosis periodically without the need for continuous dietary restrictions. This study evaluated seizure severity, biochemical markers, and immune-related factors in TLE patients. The intervention group received neuro-Capridin caprylate and caprate (n-CAP), while the control group did not. Significant findings included increased plasma ATP and adenosine levels in the treatment group, along with higher expression of ADORA1 and CD73 and reduced expression of ADK. Corresponding protein changes were observed, with increased CD73 and decreased ADK levels. Caprylate and Caprate also elevated regulatory T cells and reduced proinflammatory cytokines (TNF-α, IL-6, IL-1β). These changes were associated with significant reductions in seizure severity and frequency. Intermittent ketogenesis through the consumption of Caprylate and Caprate effectively reduced seizures and improved immune and metabolic markers in drug-resistant TLE patients. These findings highlight its potential as a complementary therapy, warranting further exploration of its long-term impact and underlying molecular mechanisms.
Collapse
Affiliation(s)
- Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Parsa Alehossein
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Ehtiati
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Tayebe Zarei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Farzaneh Salmani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Sadegh Bagherzadeh
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Razmafrooz
- Iranian Center of Neurological Research, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Aram Halimi
- Research Center for Social Determinants of Health, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Faghih Jouibari
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.
| |
Collapse
|
4
|
Nassar M, Nassar O, Abosheaishaa H, Misra A. Comparative outcomes of systemic diseases in people with type 2 diabetes, or obesity alone treated with and without GLP-1 receptor agonists: a retrospective cohort study from the Global Collaborative Network : Author list. J Endocrinol Invest 2025; 48:483-497. [PMID: 39302577 DOI: 10.1007/s40618-024-02466-4] [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: 06/26/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are increasingly used to manage type 2 diabetes (T2D) and obesity. Despite their recognized benefits in glycemic control and weight management, their impact on broader systemic has been less explored. OBJECTIVE This study aimed to evaluate the impact of GLP-1RAs on a variety of systemic diseases in people with T2D or obesity. METHODS We conducted a retrospective cohort study using data from the Global Collaborative Network, accessed through the TriNetX analytics platform. The study comprised two primary groups: individuals with T2D and those with obesity. Each group was further divided into subgroups based on whether they received GLP-1RA treatment or not. Data were analyzed over more than a 5-year follow-up period, comparing incidences of systemic diseases; systemic lupus erythematosus (SLE), systemic sclerosis (SS), rheumatoid arthritis (RA), ulcerative colitis (UC), crohn's disease (CD), alzheimer's disease (AD), parkinson's disease (PD), dementia, bronchial asthma (BA), osteoporosis, and several cancers. RESULTS In the T2D cohorts, GLP-1RA treatment was associated with significantly lower incidences of several systemic and metabolic conditions as compared to those without GLP-1RA, specifically, dementia (Risk Difference (RD): -0.010, p < 0.001), AD (RD: -0.003, p < 0.001), PD (RD: -0.002, p < 0.001), and pancreatic cancer (RD: -0.003, p < 0.001). SLE and SS also saw statistically significant reductions, though the differences were minor in magnitude (RD: -0.001 and - 0.000 respectively, p < 0.001 for both). Conversely, BA a showed a slight increase in risk (RD: 0.002, p < 0.001). CONCLUSIONS GLP-1RAs demonstrate potential benefits in reducing the risk of several systemic conditions in people with T2D or obesity. Further prospective studies are needed to confirm these effects fully and understand the mechanisms.
Collapse
Affiliation(s)
- Mahmoud Nassar
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Omar Nassar
- Williamsville East High School, Buffalo, NY, USA
| | - Hazem Abosheaishaa
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anoop Misra
- Fortis-C-DOC Centre of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India
- National Diabetes, Obesity and Cholesterol Foundation (N-DOC), New Delhi, India
- Diabetes Foundation (India) (DFI) India, New Delhi, India
| |
Collapse
|
5
|
Li S, Li S, Meng L, Gao R, Liu H, Li M. Immunopathogenesis and immunotherapy of diabetes-associated periodontitis. Clin Oral Investig 2025; 29:44. [PMID: 39755848 DOI: 10.1007/s00784-024-06141-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: 08/02/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
OBJECTIVES This paper aims to review the immunopathogenesis of Diabetes-associated periodontitis (DPD) and to propose a description of the research progress of drugs with potential clinical value from an immunotherapeutic perspective. MATERIALS AND METHODS A comprehensive literature search was conducted in PubMed, MEDLINE, Embase, Web of Science, Scopus and the Cochrane Library. Inclusion criteria were studies on the association between diabetes and periodontitis using the Boolean operator "AND" for association between diabetes and periodontitis, with no time or language restrictions. Search terms included diabetes mellitus, periodontitis, immunopathogenesis, specific immunity, non-specific immunity, flora, estrogen, pharmacological treatment, immunotherapy. RESULTS Alterations in the subgingival flora environment in a hyperglycemic environment elicit an immune response. Overactivity/suppression of nonspecific immune cells and impaired cellular defenses trigger specific immune responses. Epigenetics as well as female hormones also play a role. There is already a small amount of clinical evidence for the role of metronidazole, subantimicrobial doses of doxycycline, minocycline hydrochloride, and metformin in the treatment of DPD. Some preclinical studies have also accumulated experimental evidence for the improved effects of vitamin D3 and other drugs on DPD. CONCLUSIONS The development of diabetic periodontitis is immunologically linked to a state of immune imbalance and therefore holds great promise for the use of immunotherapeutic drugs. CLINICAL SIGNIFICANCE Immunotherapy with drugs along with periodontal nonsurgical treatment could provide ideas for DPD treatment based on the immunopathogenesis of DPD.
Collapse
Affiliation(s)
- Shuangshuang Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
- Department of Stomatology, Shengli Oilfield Central Hospital, Dongying, Shandong, China
| | - Shuang Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, Shandong, China
| | - Lingxiao Meng
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, Shandong, China
| | - Ruihan Gao
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, Shandong, China
| | - Hongrui Liu
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China.
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, Shandong, China.
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China.
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
6
|
Halabitska I, Petakh P, Lushchak O, Kamyshna I, Oksenych V, Kamyshnyi O. Metformin in Antiviral Therapy: Evidence and Perspectives. Viruses 2024; 16:1938. [PMID: 39772244 PMCID: PMC11680154 DOI: 10.3390/v16121938] [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: 12/08/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Metformin, a widely used antidiabetic medication, has emerged as a promising broad-spectrum antiviral agent due to its ability to modulate cellular pathways essential for viral replication. By activating AMPK, metformin depletes cellular energy reserves that viruses rely on, effectively limiting the replication of pathogens such as influenza, HIV, SARS-CoV-2, HBV, and HCV. Its role in inhibiting the mTOR pathway, crucial for viral protein synthesis and reactivation, is particularly significant in managing infections caused by HIV, CMV, and EBV. Furthermore, metformin reduces oxidative stress and reactive oxygen species (ROS), which are critical for replicating arboviruses such as Zika and dengue. The drug also regulates immune responses, cellular differentiation, and inflammation, disrupting the life cycle of HPV and potentially other viruses. These diverse mechanisms suppress viral replication, enhance immune system functionality, and contribute to better clinical outcomes. This multifaceted approach highlights metformin's potential as an adjunctive therapy in treating a wide range of viral infections.
Collapse
Affiliation(s)
- Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Voli Square, 1, 46001 Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, 88017 Uzhhorod, Ukraine
| | - Oleh Lushchak
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| |
Collapse
|
7
|
Feng P, Yang Q, Luo L, Guan Z, Fu J, Zhao M, Meng W, Wan S, He J, Li Z, Wang G, Sun G, Dong Z, Yang M. Vps34 sustains Treg cell survival and function via regulating intracellular redox homeostasis. Cell Death Differ 2024; 31:1519-1533. [PMID: 39117783 PMCID: PMC11519664 DOI: 10.1038/s41418-024-01353-y] [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: 08/13/2023] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
The survival and suppressive function of regulatory T (Treg) cells rely on various intracellular metabolic and physiological processes. Our study demonstrates that Vps34 plays a critical role in maintaining Treg cell homeostasis and function by regulating cellular metabolic activities. Disruption of Vps34 in Treg cells leads to spontaneous fatal systemic autoimmune disorder and multi-tissue inflammatory damage, accompanied by a reduction in the number of Treg cells, particularly eTreg cells with highly immunosuppressive activity. Mechanistically, the poor survival of Vps34-deficient Treg cells is attributed to impaired endocytosis, intracellular vesicular trafficking and autophagosome formation, which further results in enhanced mitochondrial respiration and excessive ROS production. Removal of excessive ROS can effectively rescue the death of Vps34-deficient Treg cells. Functionally, acute deletion of Vps34 within established Treg cells enhances anti-tumor immunity in a malignant melanoma model by boosting T-cell-mediated anti-tumor activity. Overall, our results underscore the pivotal role played by Vps34 in orchestrating Treg cell homeostasis and function towards establishing immune homeostasis and tolerance.
Collapse
Affiliation(s)
- Peiran Feng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
| | - Liang Luo
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Zerong Guan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Jiamin Fu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Mingyue Zhao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Wanqing Meng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Shuo Wan
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Junming He
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Zhizhong Li
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Hefei, Anhui, China.
| | - Meixiang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China.
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China.
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. Institute of Laboratory Animal Science, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
8
|
Bostick JW, Connerly TJ, Thron T, Needham BD, de Castro Fonseca M, Kaddurah-Daouk R, Knight R, Mazmanian SK. The microbiome shapes immunity in a sex-specific manner in mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593011. [PMID: 38766238 PMCID: PMC11100721 DOI: 10.1101/2024.05.07.593011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
INTRODUCTION : Preclinical studies reveal that the microbiome broadly affects immune responses and deposition and/or clearance of amyloid-beta (Aβ) in mouse models of Alzheimer's disease (AD). Whether the microbiome shapes central and peripheral immune profiles in AD models remains unknown. METHODS : We examined adaptive immune responses in two mouse models containing AD- related genetic predispositions (3xTg and 5xFAD) in the presence or absence of the microbiome. RESULTS : T and B cells were altered in brain-associated and systemic immune tissues between genetic models and wildtype mice, with earlier signs of immune activity in females. Systemic immune responses were modulated by the microbiome and differed by sex. Further, the absence of a microbiome in germ-free mice resulted in reduced cognitive deficits, primarily in females. DISCUSSION : These data reveal sexual dimorphism in early signs of immune activity and microbiome effects, and highlight an interesting interaction between sex and the microbiome in mouse models of AD.
Collapse
|
9
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [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] [Indexed: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Park SM, Oh YH, Lim GH, Yun GH, Kim KB, An JH, Seo KW, Youn HY. Deferoxamine preconditioning of canine stem cell derived extracellular vesicles alleviates inflammation in an EAE mouse model through STAT3 regulation. Sci Rep 2024; 14:19273. [PMID: 39164295 PMCID: PMC11335858 DOI: 10.1038/s41598-024-68853-2] [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: 02/25/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs), specifically those preconditioned with deferoxamine (DFO) in canine adipose tissue-derived MSCs (cAT-MSCs), were explored for treating autoimmune diseases. This study assessed the effects of DFO-preconditioned EVs (EVDFO) in an experimental autoimmune encephalomyelitis (EAE) mouse model. cAT-MSCs were treated with DFO for 48 h, after which EVs were isolated. EAE mice received intranasal EV or EVDFO treatments and were euthanized following histopathologic analysis; RNA and protein expression levels were measured. Histologically, EV and EVDFO groups showed a significant reduction in inflammatory cell infiltration and demyelination. Immunofluorescence revealed increased CD206 and Foxp3 expression, indicating elevated M2 macrophages and regulatory T (Treg) cells, particularly in the EVDFO group. Treg cells also notably increased in the spleen of EVDFO -treated mice. STAT3 and pSTAT3 proteins were upregulated in the EAE groups compared to the naïve group. However, following EV treatment, STAT3 expression decreased compared to the EAE group, whereas pSTAT3 expression was similar in both the EV and EAE groups. In conclusion, EVDFO treatment resulted in reduced STAT3 expression, suggesting its role in T cell regulation and the potential of EVDFO in modulating the STAT3 pathway for reducing inflammation more effectively than non-preconditioned EVs.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/transplantation
- STAT3 Transcription Factor/metabolism
- Mice
- Dogs
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
- Deferoxamine/pharmacology
- Deferoxamine/therapeutic use
- Mesenchymal Stem Cells/metabolism
- Inflammation/pathology
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
- Su-Min Park
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yong-Hun Oh
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ga-Hyun Lim
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ga-Hee Yun
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyung-Bo Kim
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ju-Hyun An
- Department of Veterinary Emergency and Critical Care Medicine and Institute of Veterinary Science, College of Veterinary Medicine, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Kyung-Won Seo
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine and Research Institute for Veterinary Science, Department of Clinical Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
11
|
Gill JS, Bansal B, Guo K, Huang F, Singh H, Hur J, Khan N, Mathur R. Mitochondrial Oxidative Stress Regulates FOXP3+ T-Cell Activity and CD4-Mediated Inflammation in Older Adults with Frailty. Int J Mol Sci 2024; 25:6235. [PMID: 38892421 PMCID: PMC11173216 DOI: 10.3390/ijms25116235] [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: 04/23/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
In healthy older adults, the immune system generally preserves its response and contributes to a long, healthy lifespan. However, rapid deterioration in immune regulation can lead to chronic inflammation, termed inflammaging, which accelerates pathological aging and diminishes the quality of life in older adults with frailty. A significant limitation in current aging research is the predominant focus on comparisons between young and older populations, often overlooking the differences between healthy older adults and those experiencing pathological aging. Our study elucidates the intricate immunological dynamics of the CD4/Treg axis in frail older adults compared to comparable age-matched healthy older adults. By utilizing publicly available RNA sequencing and single-cell RNA sequencing (scRNAseq) data from peripheral blood mononuclear cells (PBMCs), we identified a specific Treg cell subset and transcriptional landscape contributing to the dysregulation of CD4+ T-cell responses. We explored the molecular mechanisms underpinning Treg dysfunction, revealing that Tregs from frail older adults exhibit reduced mitochondrial protein levels, impairing mitochondrial oxidative phosphorylation. This impairment is driven by the TNF/NF-kappa B pathway, leading to cumulative inflammation. Further, we gained a deeper understanding of the CD4/Treg axis by predicting the effects of gene perturbations on cellular signaling networks. Collectively, these findings highlight the age-related relationship between mitochondrial dysfunction in the CD4/Treg axis and its role in accelerating aging and frailty in older adults. Targeting Treg dysfunction offers a critical basis for developing tailored therapeutic strategies aimed at improving the quality of life in older adults.
Collapse
Affiliation(s)
- Jappreet Singh Gill
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
- Department of Biomedical Engineering, School of Electrical Engineering and Computer Sciences, University of North Dakota, Grand Forks, ND 58292, USA
| | - Benu Bansal
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
- Department of Biomedical Engineering, School of Electrical Engineering and Computer Sciences, University of North Dakota, Grand Forks, ND 58292, USA
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
| | - Kai Guo
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fang Huang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
| | - Harpreet Singh
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
| | - Nadeem Khan
- Department of Oral Biology, University of Florida, Gainsville, FL 32603, USA;
| | - Ramkumar Mathur
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
| |
Collapse
|
12
|
Larson AC, Doty KR, Solheim JC. The double life of a chemotherapy drug: Immunomodulatory functions of gemcitabine in cancer. Cancer Med 2024; 13:e7287. [PMID: 38770637 PMCID: PMC11106691 DOI: 10.1002/cam4.7287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024] Open
Abstract
Although the development of immunotherapies has been revolutionary in the treatment of several cancers, many cancer types remain unresponsive to immune-based treatment and are largely managed by chemotherapy drugs. However, chemotherapeutics are not infallible and are frequently rendered ineffective as resistance develops from prolonged exposure. Recent investigations have indicated that some chemotherapy drugs have additional functions beyond their normative cytotoxic capacity and are in fact immune-modifying agents. Of the pharmaceuticals with identified immune-editing properties, gemcitabine is well-studied and of interest to clinicians and scientists alike. Gemcitabine is a chemotherapy drug approved for the treatment of multiple cancers, including breast, lung, pancreatic, and ovarian. Because of its broad applications, relatively low toxicity profile, and history as a favorable combinatory partner, there is promise in the recharacterization of gemcitabine in the context of the immune system. Such efforts may allow the identification of suitable immunotherapeutic combinations, wherein gemcitabine can be used as a priming agent to improve immunotherapy efficacy in traditionally insensitive cancers. This review looks to highlight documented immunomodulatory abilities of one of the most well-known chemotherapy agents, gemcitabine, relating to its influence on cells and proteins of the immune system.
Collapse
Affiliation(s)
- Alaina C. Larson
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kenadie R. Doty
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Joyce C. Solheim
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Biochemistry & Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Pathology, Microbiology, & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
13
|
Li QH, Zhao QY, Yang WJ, Jiang AF, Ren CE, Meng YH. Beyond Immune Balance: The Pivotal Role of Decidual Regulatory T Cells in Unexplained Recurrent Spontaneous Abortion. J Inflamm Res 2024; 17:2697-2710. [PMID: 38707955 PMCID: PMC11070170 DOI: 10.2147/jir.s459263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more consecutive pregnancy failures, which brings tremendous stress to women of childbearing age and seriously affects family well-being. However, the reason in about 50% of cases remains unknown and is defined as unexplained recurrent spontaneous abortion (URSA). The immunological perspective in URSA has attracted widespread attention in recent years. The embryo is regarded as a semi-allogeneic graft to the mother. A successful pregnancy requires transition to an immune environment conducive to embryo survival at the maternal-fetal interface. As an important member of regulatory immunity, regulatory T (Treg) cells play a key role in regulating immune tolerance at the maternal-fetal interface. This review will focus on the phenotypic plasticity and lineage stability of Treg cells to illustrate its relationship with URSA.
Collapse
Affiliation(s)
- Qing-Hui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Qiu-Yan Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Wei-Jing Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Ai-Fang Jiang
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Chun-E Ren
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Yu-Han Meng
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| |
Collapse
|
14
|
Zhang YS, Chen YQ. Dysfunctional regulatory T cell: May be an obstacle to immunotherapy in cardiovascular diseases. Biomed Pharmacother 2024; 173:116359. [PMID: 38430633 DOI: 10.1016/j.biopha.2024.116359] [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: 12/06/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Inflammatory responses are linked to cardiovascular diseases (CVDs) in various forms. Tregs, members of CD4+ T cells, play important roles in regulating immune system and suppressing inflammatory response, thus contributing to maintaining immune homeostasis. However, Tregs exert their powerful suppressive function relying on the stable phenotype and function. The stability of Tregs primarily depends on the FOXP3 (Forkhead box P3) expression and epigenetic regulation. Although Tregs are quite stable under physiological conditions, prolonged exposure to inflammatory cues, Tregs may lose suppressive function and require proinflammatory phenotype, namely plastic Tregs or ex-Tregs. There are extensive researches have established the beneficial role of Tregs in CVDs. Nevertheless, the potential risks of dysfunctional Tregs lack deep research. Anti-inflammatory and immunological modulation have been hotspots in the treatment of CVDs. Tregs are appealing because of their crucial role in resolving inflammation and promoting tissue repair. If alleviating inflammatory response through modulating Tregs could be a new therapeutic strategy for CVDs, the next step to consider is how to prevent the formation of dysfunctional Tregs or reverse detrimental Tregs to normal phenotype.
Collapse
Affiliation(s)
- Yu-Sha Zhang
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Ya-Qin Chen
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Hunan, China.
| |
Collapse
|
15
|
Martinez HA, Koliesnik I, Kaber G, Reid JK, Nagy N, Barlow G, Falk BA, Medina CO, Hargil A, Zihsler S, Vlodavsky I, Li JP, Pérez-Cruz M, Tang SW, Meyer EH, Wrenshall LE, Lord JD, Garcia KC, Palmer TD, Steinman L, Nepom GT, Wight TN, Bollyky PL, Kuipers HF. Regulatory T cells use heparanase to access IL-2 bound to extracellular matrix in inflamed tissue. Nat Commun 2024; 15:1564. [PMID: 38378682 PMCID: PMC10879116 DOI: 10.1038/s41467-024-45012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 01/08/2024] [Indexed: 02/22/2024] Open
Abstract
Although FOXP3+ regulatory T cells (Treg) depend on IL-2 produced by other cells for their survival and function, the levels of IL-2 in inflamed tissue are low, making it unclear how Treg access this critical resource. Here, we show that Treg use heparanase (HPSE) to access IL-2 sequestered by heparan sulfate (HS) within the extracellular matrix (ECM) of inflamed central nervous system tissue. HPSE expression distinguishes human and murine Treg from conventional T cells and is regulated by the availability of IL-2. HPSE-/- Treg have impaired stability and function in vivo, including in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Conversely, endowing monoclonal antibody-directed chimeric antigen receptor (mAbCAR) Treg with HPSE enhances their ability to access HS-sequestered IL-2 and their ability to suppress neuroinflammation in vivo. Together, these data identify a role for HPSE and the ECM in immune tolerance, providing new avenues for improving Treg-based therapy of autoimmunity.
Collapse
Affiliation(s)
- Hunter A Martinez
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ievgen Koliesnik
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacqueline K Reid
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Nadine Nagy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Graham Barlow
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ben A Falk
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Carlos O Medina
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Aviv Hargil
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Svenja Zihsler
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Magdiel Pérez-Cruz
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sai-Wen Tang
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Everett H Meyer
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lucile E Wrenshall
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - James D Lord
- Translational Research Program, Benaroya Research Institute, Seattle, WA, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Theo D Palmer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Gerald T Nepom
- Immune Tolerance Network, Benaroya Research Institute, Seattle, WA, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Paul L Bollyky
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Hedwich F Kuipers
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada.
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
16
|
Shan J, Shi R, Hazra R, Hu X. Regulatory T lymphocytes in traumatic brain injury. Neurochem Int 2024; 173:105660. [PMID: 38151109 PMCID: PMC10872294 DOI: 10.1016/j.neuint.2023.105660] [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: 10/30/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/29/2023]
Abstract
Traumatic brain injury (TBI) presents a significant global health challenge with no effective therapies developed to date. Regulatory T lymphocytes (Tregs) have recently emerged as a potential therapy due to their critical roles in maintaining immune homeostasis, reducing inflammation, and promoting brain repair. Following TBI, fluctuations in Treg populations and shifts in their functionality have been noted. However, the precise impact of Tregs on the pathophysiology of TBI remains unclear. In this review, we discuss recent advances in understanding the intricate roles of Tregs in TBI and other brain diseases. Increased knowledge about Tregs may facilitate their future application as an immunotherapy target for TBI treatment.
Collapse
Affiliation(s)
- Jiajing Shan
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ruyu Shi
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Rimi Hazra
- Department of Medicine, Pittsburgh Heart Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
17
|
Eshraghisamani R, Facciuolo A, Harman-McKenna V, Illanes O, De Buck J. Immunogenicity and efficacy of an oral live-attenuated vaccine for bovine Johne's disease. Front Immunol 2024; 14:1307621. [PMID: 38283338 PMCID: PMC10810994 DOI: 10.3389/fimmu.2023.1307621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/11/2023] [Indexed: 01/30/2024] Open
Abstract
Mycobacterium avium subsp. paratuberculosis (MAP), the etiological agent of Johne's disease (JD) in ruminants, establishes a prolonged and often lifelong enteric infection. The implementation of control measures for bovine JD has faced obstacles due to the considerable expenses involved in disease surveillance and hindered by unreliable and inadequate diagnostic tests, emphasizing the need for an effective vaccine that can stimulate mucosal immunity in the gastrointestinal tract. Previous investigations have demonstrated that deletion of the BacA gene in MAP produces an attenuated strain that can transiently colonize the calf small intestine while retaining its capacity to stimulate systemic immune responses similar to wildtype MAP strains. This study assessed the efficacy of the BacA gene deletion MAP strain, referred to as the BacA vaccine, when administered orally to young calves. The research aimed to evaluate its effectiveness in controlling MAP intestinal infection and to investigate the immune responses elicited by mucosal vaccination. The study represents the first evaluation of an enteric modified live MAP vaccine in the context of an oral MAP challenge in young calves. Oral immunization with BacA reduced MAP colonization specifically in the ileum and ileocecal valve. This partially protective immune response was associated with an increased frequency of CD4+ and CD8+ T cells with a pro-inflammatory phenotype (IFNγ+/TNFα+) in vaccinated animals. Moreover, re-stimulated PBMCs from vaccinated animals showed increased expression of IFNγ, IP-10, IL-2, and IL-17 at 10- and 12-weeks post challenge. Furthermore, immunophenotyping of blood leukocytes revealed that vaccinated calves had increased levels of T cells expressing cell-surface markers consistent with long-term central memory. Overall, our findings provide new insights into the development and immunogenicity of a modified live MAP vaccine against bovine JD, demonstrating oral vaccination can stimulate host immune responses that can be protective against enteric MAP infection.
Collapse
Affiliation(s)
| | - Antonio Facciuolo
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Oscar Illanes
- College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Jeroen De Buck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
18
|
Paterson CW, Fay KT, Chen CW, Klingensmith NJ, Gutierrez MB, Liang Z, Coopersmith CM, Ford ML. CTLA-4 Checkpoint Inhibition Improves Sepsis Survival in Alcohol-Exposed Mice. Immunohorizons 2024; 8:74-88. [PMID: 38226924 PMCID: PMC10835704 DOI: 10.4049/immunohorizons.2300060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Chronic alcohol use increases morbidity and mortality in the setting of sepsis. Both chronic alcohol use and sepsis are characterized by immune dysregulation, including overexpression of T cell coinhibitory molecules. We sought to characterize the role of CTLA-4 during sepsis in the setting of chronic alcohol exposure using a murine model of chronic alcohol ingestion followed by cecal ligation and puncture. Results indicated that CTLA-4 expression is increased on CD4+ T cells isolated from alcohol-drinking septic mice as compared with either alcohol-drinking sham controls or water-drinking septic mice. Moreover, checkpoint inhibition of CTLA-4 improved sepsis survival in alcohol-drinking septic mice, but not water-drinking septic mice. Interrogation of the T cell compartments in these animals following pharmacologic CTLA-4 blockade, as well as following conditional Ctla4 deletion in CD4+ T cells, revealed that CTLA-4 deficiency promoted the activation and proliferation of effector regulatory T cells and the generation of conventional effector memory CD4+ T cells. These data highlight an important role for CTLA-4 in mediating mortality during sepsis in the setting of chronic alcohol exposure and may inform future approaches to develop targeted therapies for this patient population.
Collapse
Affiliation(s)
- Cameron W. Paterson
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
- Lieutenant, Medical Corps, Naval Reserve Officer Training Corp, United States Navy, Atlanta, GA
| | - Katherine T. Fay
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Ching-Wen Chen
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Nathan J. Klingensmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Melissa B. Gutierrez
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Zhe Liang
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Craig M. Coopersmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Mandy L. Ford
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta GA
| |
Collapse
|
19
|
Deng X, Wang L, Zhai Y, Liu Q, Du F, Zhang Y, Zhao W, Wu T, Tao Y, Deng J, Cao Y, Hao P, Ren J, Shen Y, Yu Z, Zheng Y, Zhang H, Wang H. RIPK1 plays a crucial role in maintaining regulatory T-Cell homeostasis by inhibiting both RIPK3- and FADD-mediated cell death. Cell Mol Immunol 2024; 21:80-90. [PMID: 38082146 PMCID: PMC10757712 DOI: 10.1038/s41423-023-01113-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 11/13/2023] [Indexed: 01/01/2024] Open
Abstract
Regulatory T (Treg) cells play an essential role in maintaining immune balance across various physiological and pathological conditions. However, the mechanisms underlying Treg homeostasis remain incompletely understood. Here, we report that RIPK1 is crucial for Treg cell survival and homeostasis. We generated mice with Treg cell-specific ablation of Ripk1 and found that these mice developed fatal systemic autoimmunity due to a dramatic reduction in the Treg cell compartment caused by excessive cell death. Unlike conventional T cells, Treg cells with Ripk1 deficiency were only partially rescued from cell death by blocking FADD-dependent apoptosis. However, simultaneous removal of both Fadd and Ripk3 completely restored the homeostasis of Ripk1-deficient Treg cells by blocking two cell death pathways. Thus, our study highlights the critical role of RIPK1 in regulating Treg cell homeostasis by controlling both apoptosis and necroptosis, thereby providing novel insights into the mechanisms of Treg cell homeostasis.
Collapse
Affiliation(s)
- Xiaoxue Deng
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lingxia Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yunze Zhai
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qiuyue Liu
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Fengxue Du
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Zhang
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenxing Zhao
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tingtao Wu
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yiwen Tao
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jie Deng
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200433, China
| | - Yongbing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200433, China
| | - Pei Hao
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiazi Ren
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yunli Shen
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zuoren Yu
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Haikun Wang
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
20
|
Wu C, Jiang ML, Pang T, Zhang CJ. T Cell Subsets and Immune Homeostasis. Methods Mol Biol 2024; 2782:39-63. [PMID: 38622391 DOI: 10.1007/978-1-0716-3754-8_3] [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] [Indexed: 04/17/2024]
Abstract
T cells are a heterogeneous group of cells that can be classified into different subtypes according to different classification methods. The body's immune system has a highly complex and effective regulatory network that allows for the relative stability of immune system function. Maintaining proper T cell homeostasis is essential for promoting protective immunity and limiting autoimmunity and tumor formation. Among the T cell family members, more and more T cell subsets have gradually been characterized. In this chapter, we summarize the functions of some key T cell subsets and their impact on immune homeostasis.
Collapse
Affiliation(s)
- Chuyu Wu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Mei-Ling Jiang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Cun-Jin Zhang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Reißing J, Berres M, Strnad P, Wree A, Inzaugarat ME, Trautwein C, Bruns T, Zimmermann HW. Th2 Cell Activation in Chronic Liver Disease Is Driven by Local IL33 and Contributes to IL13-Dependent Fibrogenesis. Cell Mol Gastroenterol Hepatol 2023; 17:517-538. [PMID: 38158122 PMCID: PMC10882164 DOI: 10.1016/j.jcmgh.2023.12.011] [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: 05/20/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND & AIMS Type 2 immune responses contribute to liver fibrosis in parasite infections, but their role in other liver diseases is less well understood. Here, we aimed at unravelling mechanisms involved in T helper 2 (Th2) T-cell polarization, activation, and recruitment in human liver fibrosis and cirrhosis. METHODS Tissues, cells, and serum from human livers were analyzed using quantitative reverse-transcription polymerase chain reaction, enzyme-linked immunosorbent assay, fluorescence in situ hybridization, immunostaining, flow cytometry, and various functional in vitro assays. Cellular interactions and soluble mediators involved in T-cell polarization and recruitment were studied, as well as their effect on hepatic stellate cell (HSC) activation, proliferation, and extracellular matrix synthesis. RESULTS In human liver fibrosis, a stage-dependent increase in Th2-related transcription factors, Th2 cytokines, and trans-acting T-cell-specific transcription factor-expressing T cells was observed, and was highest in cirrhotic livers. The alarmin interleukin (IL)33 was found to be increased in livers and sera from patients with cirrhosis, to act as a chemotactic agent for Th2 cells, and to induce type 2 polarization of CD4+ T cells. Oval cells, liver sinusoidal endothelial cells, intrahepatic macrophages, and migrating monocytes were identified as sources of IL33. IL33-activated T cells, but not IL33 alone, induced HSC activation, as shown by Ki67 and α-smooth muscle actin staining, increased collagen type I alpha 1 chain messenger RNA expression, and wound healing assays. The profibrotic effect of IL33-activated T cells was contact-independent and could be antagonized using monoclonal antibodies against IL13. CONCLUSION In patients with chronic liver disease, the alarmin IL33 promotes the recruitment and activation of CD4+ T cells with Th2-like properties, which activate paracrine HSC in an IL13-dependent manner and promotes fibrogenesis.
Collapse
Affiliation(s)
- Johanna Reißing
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Marie Berres
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Alexander Wree
- Department of Gastroenterology/Hepatology, Campus Virchow Klinikum, Charité Campus Mitte, Charité University Medicine Berlin, Berlin, Germany
| | - Maria Eugenia Inzaugarat
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Henning Wolfgang Zimmermann
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.
| |
Collapse
|
22
|
Funk MC, Gleixner JG, Heigwer F, Vonficht D, Valentini E, Aydin Z, Tonin E, Del Prete S, Mahara S, Throm Y, Hetzer J, Heide D, Stegle O, Odom DT, Feldmann A, Haas S, Heikenwalder M, Boutros M. Aged intestinal stem cells propagate cell-intrinsic sources of inflammaging in mice. Dev Cell 2023; 58:2914-2929.e7. [PMID: 38113852 DOI: 10.1016/j.devcel.2023.11.013] [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: 08/16/2022] [Revised: 05/03/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023]
Abstract
Low-grade chronic inflammation is a hallmark of ageing, associated with impaired tissue function and disease development. However, how cell-intrinsic and -extrinsic factors collectively establish this phenotype, termed inflammaging, remains poorly understood. We addressed this question in the mouse intestinal epithelium, using mouse organoid cultures to dissect stem cell-intrinsic and -extrinsic sources of inflammaging. At the single-cell level, we found that inflammaging is established differently along the crypt-villus axis, with aged intestinal stem cells (ISCs) strongly upregulating major histocompatibility complex class II (MHC-II) genes. Importantly, the inflammaging phenotype was stably propagated by aged ISCs in organoid cultures and associated with increased chromatin accessibility at inflammation-associated loci in vivo and ex vivo, indicating cell-intrinsic inflammatory memory. Mechanistically, we show that the expression of inflammatory genes is dependent on STAT1 signaling. Together, our data identify that intestinal inflammaging in mice is promoted by a cell-intrinsic mechanism, stably propagated by ISCs, and associated with a disbalance in immune homeostasis.
Collapse
Affiliation(s)
- Maja C Funk
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Jan G Gleixner
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), Division of Computational Genomics and Systems Genetics, 69120 Heidelberg, Germany; Genome Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69117 Heidelberg, Germany
| | - Florian Heigwer
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany; Department of Life Sciences and Engineering, University of Applied Sciences Bingen, 55411 Bingen am Rhein, Germany
| | - Dominik Vonficht
- Faculty of Biosciences, Heidelberg University, 69117 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine, (HI-STEM gGmbH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), Division of Stem Cells and Cancer, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Erica Valentini
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Zeynep Aydin
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Elena Tonin
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Stefania Del Prete
- German Cancer Research Center (DKFZ), Division Regulatory Genomics and Cancer Evolution, 69120 Heidelberg, Germany
| | - Sylvia Mahara
- German Cancer Research Center (DKFZ), Junior Research Group Mechanisms of Genome Control, 69120 Heidelberg, Germany
| | - Yannick Throm
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Jenny Hetzer
- German Cancer Research Center (DKFZ), Division Chronic Inflammation and Cancer, 69120 Heidelberg, Germany
| | - Danijela Heide
- German Cancer Research Center (DKFZ), Division Chronic Inflammation and Cancer, 69120 Heidelberg, Germany
| | - Oliver Stegle
- German Cancer Research Center (DKFZ), Division of Computational Genomics and Systems Genetics, 69120 Heidelberg, Germany; Genome Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Duncan T Odom
- German Cancer Research Center (DKFZ), Division Regulatory Genomics and Cancer Evolution, 69120 Heidelberg, Germany
| | - Angelika Feldmann
- German Cancer Research Center (DKFZ), Junior Research Group Mechanisms of Genome Control, 69120 Heidelberg, Germany
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, (HI-STEM gGmbH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), Division of Stem Cells and Cancer, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, 10115 Berlin, Germany
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division Chronic Inflammation and Cancer, 69120 Heidelberg, Germany; M3 Research Center, Medical Faculty Tübingen, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
23
|
Rutkowska-Zapała M, Grabowska A, Lenart M, Kluczewska A, Szaflarska A, Kobylarz K, Pituch-Noworolska A, Siedlar M. Transcriptome profiling of regulatory T cells from children with transient hypogammaglobulinemia of infancy. Clin Exp Immunol 2023; 214:275-288. [PMID: 37936298 PMCID: PMC10719223 DOI: 10.1093/cei/uxad116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/26/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023] Open
Abstract
Transient hypogammaglobulinemia of infancy (THI) is one of the most common forms of hypogammaglobulinemia in the early childhood. THI is usually associated with chronic, recurrent bacterial and viral infections, life-threatening in some cases, yet its pathogenesis is still largely unknown. As our previous findings indicated the possible role of Treg cells in the pathomechanism of THI, the aim of the current study was to investigate gene expression profile of Treg cells isolated from THI patients. The transcriptome-wide gene profiling was performed using microarray technology on THI patients in two time-points: during (THI-1), and in resolution phase (THI-2) of hypogammaglobulinemia. As a result, a total of 1086 genes were differentially expressed in THI-1 patients, when compared to THI-2 as well as control group. Among them, 931 were up- and 155 downregulated, and part of them encodes genes important for Treg lymphocyte biology and function, i.e. transcription factors/cofactors that regulate FOXP3 expression. Thus, we postulate that Treg cells isolated from THI patients during hypogammaglobulinemia display enhanced suppressor transcriptome signature. Treg expression profile of THI children after normalization of Ig levels largely resembles the results obtained in healthy control group, suggesting THI Treg transcriptome seems to return to that observed in healthy children. Taken together, we suggest that THI pathomechanism is associated not only with transiently elevated Treg cell numbers, but also with their enhanced regulatory/inhibitory functions. These findings expand our knowledge of human Treg cells and may be useful for the future diagnosis or management of THI.
Collapse
Affiliation(s)
- Magdalena Rutkowska-Zapała
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Agnieszka Grabowska
- Department of Medical Genetics, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Marzena Lenart
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Anna Kluczewska
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Anna Szaflarska
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Krzysztof Kobylarz
- Department of Anesthesiology and Intensive Care, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Anna Pituch-Noworolska
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Wielicka, Krakow, Poland
| |
Collapse
|
24
|
Lao P, Chen J, Tang L, Zhang J, Chen Y, Fang Y, Fan X. Regulatory T cells in lung disease and transplantation. Biosci Rep 2023; 43:BSR20231331. [PMID: 37795866 PMCID: PMC10611924 DOI: 10.1042/bsr20231331] [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: 08/07/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/06/2023] Open
Abstract
Pulmonary disease can refer to the disease of the lung itself or the pulmonary manifestations of systemic diseases, which are often connected to the malfunction of the immune system. Regulatory T (Treg) cells have been shown to be important in maintaining immune homeostasis and preventing inflammatory damage, including lung diseases. Given the increasing amount of evidence linking Treg cells to various pulmonary conditions, Treg cells might serve as a therapeutic strategy for the treatment of lung diseases and potentially promote lung transplant tolerance. The most potent and well-defined Treg cells are Foxp3-expressing CD4+ Treg cells, which contribute to the prevention of autoimmune lung diseases and the promotion of lung transplant rejection. The protective mechanisms of Treg cells in lung disease and transplantation involve multiple immune suppression mechanisms. This review summarizes the development, phenotype and function of CD4+Foxp3+ Treg cells. Then, we focus on the therapeutic potential of Treg cells in preventing lung disease and limiting lung transplant rejection. Furthermore, we discussed the possibility of Treg cell utilization in clinical applications. This will provide an overview of current research advances in Treg cells and their relevant application in clinics.
Collapse
Affiliation(s)
- Peizhen Lao
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jingyi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Longqian Tang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jiwen Zhang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuxi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuyin Fang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Xingliang Fan
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| |
Collapse
|
25
|
Alfar R, Napoleon JV, Shahriar I, Finnell R, Walchle C, Johnson A, Low PS. Selective reprogramming of regulatory T cells in solid tumors can strongly enhance or inhibit tumor growth. Front Immunol 2023; 14:1274199. [PMID: 37928524 PMCID: PMC10623129 DOI: 10.3389/fimmu.2023.1274199] [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: 08/08/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
Folate receptor delta (FRδ) has been used as a biomarker for regulatory T cells (Tregs), because its expression is limited to Tregs and ovum. Although FRδ is unable to bind folate, we have used molecular docking software to identify a folate congener that binds FRδ with high affinity and have exploited this FRδ-specific ligand to target attached drugs (imaging agents, immune activators, and immune suppressors) specifically to Tregs in murine tumor xenografts. Analysis of treated tumors demonstrates that targeting of a Toll-like receptor 7 agonist inhibits Treg expression of FOXP3, PD-1, CTLA4, and HELIOS, resulting in 40-80% reduction in tumor growth and repolarization of other tumor-infiltrating immune cells to more inflammatory phenotypes. Targeting of the immunosuppressive drug dexamethasone, in contrast, promotes enhanced tumor growth and shifts the tumor-infiltrating immune cells to more anti-inflammatory phenotypes. Since Tregs comprise <1% of cells in the tumor masses examined, and since the targeted drugs are not internalized by cancer cells, these data demonstrate that Tregs exert a disproportionately large effect on tumor growth. Because the targeted drug did not bind to Tregs or other immune cells in healthy tissues, the data demonstrate that the immunosuppressive properties of Tregs in tumors can be manipulated without causing systemic toxicities associated with global reprogramming of the immune system.
Collapse
Affiliation(s)
- Rami Alfar
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - John V. Napoleon
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Imrul Shahriar
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Richard Finnell
- Departments of Molecular and Cellular Biology, Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Cole Walchle
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Austin Johnson
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Philip S. Low
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
26
|
Petakh P, Kamyshna I, Kamyshnyi A. Unveiling the potential pleiotropic effects of metformin in treating COVID-19: a comprehensive review. Front Mol Biosci 2023; 10:1260633. [PMID: 37881440 PMCID: PMC10595158 DOI: 10.3389/fmolb.2023.1260633] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023] Open
Abstract
This review article explores the potential of metformin, a medication commonly used for type 2 diabetes, as an antiviral and anti-inflammatory agent in the context of coronavirus disease 2019 (COVID-19). Metformin has demonstrated inhibitory effects on the growth of SARS-CoV-2 in cell culture models and has shown promising results in reducing viral load and achieving undetectable viral levels in clinical trials. Additionally, metformin exhibits anti-inflammatory properties by reducing the production of pro-inflammatory cytokines and modulating immune cell function, which may help prevent cytokine storms associated with severe COVID-19. The drug's ability to regulate the balance between pro-inflammatory Th17 cells and anti-inflammatory Treg cells suggests its potential in mitigating inflammation and restoring T cell functionality. Furthermore, metformin's modulation of the gut microbiota, particularly changes in bacterial taxa and the production of short-chain fatty acids, may contribute to its therapeutic effects. The interplay between metformin, bile acids, the gut microbiome, glucagon-like peptide-1 secretion, and glycemic control has implications for the management of diabetes and potential interventions in COVID-19. By refreshing the current evidence, this review highlights the potential of metformin as a therapeutic option in the management of COVID-19, while also exploring its effects on the gut microbiome and immunometabolism.
Collapse
Affiliation(s)
- Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
27
|
Kumar A, Singh VK, Tiwari R, Madhukar P, Rajneesh, Kumar S, Gautam V, Engwerda C, Sundar S, Kumar R. Post kala-azar dermal leishmaniasis in the Indian sub-continent: challenges and strategies for elimination. Front Immunol 2023; 14:1236952. [PMID: 37638047 PMCID: PMC10451093 DOI: 10.3389/fimmu.2023.1236952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Visceral leishmaniasis (VL) is a severe and often fatal form of leishmaniasis caused by Leishmania donovani in the Indian sub-continent. Post Kala-azar Dermal Leishmaniasis (PKDL) is a late cutaneous manifestation of VL, typically occurring after apparent cure of VL, but sometimes even without a prior history of VL in India. PKDL serves as a significant yet neglected reservoir of infection and plays a crucial role in the transmission of the disease, posing a serious threat to the VL elimination program in the Indian sub-continent. Therefore, the eradication of PKDL should be a priority within the current VL elimination program aimed at achieving a goal of less than 1 case per 10,000 in the population at the district or sub-district levels of VL endemic areas. To accomplish this, a comprehensive understanding of the pathogenesis of PKDL is essential, as well as developing strategies for disease management. This review provides an overview of the current status of diagnosis and treatment options for PKDL, highlighting our current knowledge of the immune responses underlying disease development and progression. Additionally, the review discusses the impact of PKDL on elimination programs and propose strategies to overcome this challenge and achieve the goal of elimination. By addressing the diagnostic and therapeutic gaps, optimizing surveillance and control measures, and implementing effective intervention strategies, it is possible to mitigate the burden of PKDL and facilitate the successful elimination of VL in the Indian sub-continent.
Collapse
Affiliation(s)
- Awnish Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rahul Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Prasoon Madhukar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajneesh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shashi Kumar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Christian Engwerda
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
28
|
Bernardini F, Nusca A, Coletti F, La Porta Y, Piscione M, Vespasiano F, Mangiacapra F, Ricottini E, Melfi R, Cavallari I, Ussia GP, Grigioni F. Incretins-Based Therapies and Their Cardiovascular Effects: New Game-Changers for the Management of Patients with Diabetes and Cardiovascular Disease. Pharmaceutics 2023; 15:1858. [PMID: 37514043 PMCID: PMC10386670 DOI: 10.3390/pharmaceutics15071858] [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: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Atherosclerosis is the leading cause of death worldwide, especially in patients with type 2 diabetes mellitus (T2D). GLP-1 receptor agonists and DPP-4 inhibitors were demonstrated to play a markedly protective role for the cardiovascular system beyond their glycemic control. Several cardiovascular outcome trials (CVOT) reported the association between using these agents and a significant reduction in cardiovascular events in patients with T2D and a high cardiovascular risk profile. Moreover, recent evidence highlights a favorable benefit/risk profile in myocardial infarction and percutaneous coronary revascularization settings. These clinical effects result from their actions on multiple molecular mechanisms involving the immune system, platelets, and endothelial and vascular smooth muscle cells. This comprehensive review specifically concentrates on these cellular and molecular processes mediating the cardiovascular effects of incretins-like molecules, aiming to improve clinicians' knowledge and stimulate a more extensive use of these drugs in clinical practice as helpful cardiovascular preventive strategies.
Collapse
Affiliation(s)
- Federico Bernardini
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Annunziata Nusca
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Federica Coletti
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Ylenia La Porta
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Mariagrazia Piscione
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Francesca Vespasiano
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Fabio Mangiacapra
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Elisabetta Ricottini
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Rosetta Melfi
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Ilaria Cavallari
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Gian Paolo Ussia
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| | - Francesco Grigioni
- Unit of Cardiac Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy
| |
Collapse
|
29
|
Wang Y, Wang C, Shen L, Xu D. The Role of Regulatory T Cells in Heart Repair After Myocardial Infarction. J Cardiovasc Transl Res 2023:10.1007/s12265-022-10290-5. [PMID: 37347425 DOI: 10.1007/s12265-022-10290-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/04/2022] [Indexed: 06/23/2023]
Abstract
Myocardial infarction (MI) remains one of the leading causes of death worldwide. Inflammation and immune responses after MI are of significance to the adverse cardiac remodeling. Regulatory T cells (Tregs) play an important role in suppressing the immune response and thus benefit the post-MI remodeling. After MI, damaged cardiomyocytes may be replaced by scar tissue, leading to systolic and diastolic dysfunction and subsequently adverse remodeling. In this review, we provide an overview of the function and possible mechanisms of Tregs in post-MI heart repair. Specifically, after the occurrence of MI, Tregs infiltrated to peri-infarcted myocardium through CCR5 pathway, CXCR4-CXCL12 axis, and Hippo pathway. Normal functional Tregs can reduce the size of the MI area, improve heart function, and ameliorate myocardial remodeling by inhibiting proinflammatory cells accumulation, changing the proportion of macrophages phenotypes, improving myocardial fibrosis, protecting myocardial cells, and promoting angiogenesis. Eventually, Functional Tregs recruited into the heart can improve MI outcomes. Therefore, targeted therapies with Tregs might provide a promising approach to the treatment of MI remodeling.
Collapse
Affiliation(s)
- Yishu Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China
| | - Chunfang Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China
| | - Li Shen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China.
| |
Collapse
|
30
|
Ahuja SK, Manoharan MS, Lee GC, McKinnon LR, Meunier JA, Steri M, Harper N, Fiorillo E, Smith AM, Restrepo MI, Branum AP, Bottomley MJ, Orrù V, Jimenez F, Carrillo A, Pandranki L, Winter CA, Winter LA, Gaitan AA, Moreira AG, Walter EA, Silvestri G, King CL, Zheng YT, Zheng HY, Kimani J, Blake Ball T, Plummer FA, Fowke KR, Harden PN, Wood KJ, Ferris MT, Lund JM, Heise MT, Garrett N, Canady KR, Abdool Karim SS, Little SJ, Gianella S, Smith DM, Letendre S, Richman DD, Cucca F, Trinh H, Sanchez-Reilly S, Hecht JM, Cadena Zuluaga JA, Anzueto A, Pugh JA, Agan BK, Root-Bernstein R, Clark RA, Okulicz JF, He W. Immune resilience despite inflammatory stress promotes longevity and favorable health outcomes including resistance to infection. Nat Commun 2023; 14:3286. [PMID: 37311745 PMCID: PMC10264401 DOI: 10.1038/s41467-023-38238-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 04/17/2023] [Indexed: 06/15/2023] Open
Abstract
Some people remain healthier throughout life than others but the underlying reasons are poorly understood. Here we hypothesize this advantage is attributable in part to optimal immune resilience (IR), defined as the capacity to preserve and/or rapidly restore immune functions that promote disease resistance (immunocompetence) and control inflammation in infectious diseases as well as other causes of inflammatory stress. We gauge IR levels with two distinct peripheral blood metrics that quantify the balance between (i) CD8+ and CD4+ T-cell levels and (ii) gene expression signatures tracking longevity-associated immunocompetence and mortality-associated inflammation. Profiles of IR metrics in ~48,500 individuals collectively indicate that some persons resist degradation of IR both during aging and when challenged with varied inflammatory stressors. With this resistance, preservation of optimal IR tracked (i) a lower risk of HIV acquisition, AIDS development, symptomatic influenza infection, and recurrent skin cancer; (ii) survival during COVID-19 and sepsis; and (iii) longevity. IR degradation is potentially reversible by decreasing inflammatory stress. Overall, we show that optimal IR is a trait observed across the age spectrum, more common in females, and aligned with a specific immunocompetence-inflammation balance linked to favorable immunity-dependent health outcomes. IR metrics and mechanisms have utility both as biomarkers for measuring immune health and for improving health outcomes.
Collapse
Affiliation(s)
- Sunil K Ahuja
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Muthu Saravanan Manoharan
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Grace C Lee
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, 4001, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Justin A Meunier
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
| | - Nathan Harper
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
| | - Alisha M Smith
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Marcos I Restrepo
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Anne P Branum
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Matthew J Bottomley
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX1 2JD, UK
- Oxford Kidney Unit, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
| | - Fabio Jimenez
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Andrew Carrillo
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Lavanya Pandranki
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Caitlyn A Winter
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Lauryn A Winter
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Alvaro A Gaitan
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Alvaro G Moreira
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Elizabeth A Walter
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Guido Silvestri
- Department of Pathology, Emory University School of Medicine & Emory National Primate Research Center, Atlanta, GA, 30322, USA
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- National Resource Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Hong-Yi Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- National Resource Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Joshua Kimani
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - T Blake Ball
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Keith R Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Paul N Harden
- Oxford Kidney Unit, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX1 2JD, UK
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jennifer M Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Mark T Heise
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Kristen R Canady
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, 4001, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Susan J Little
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sara Gianella
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Davey M Smith
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, 92161, USA
| | - Scott Letendre
- Department of Medicine, University of California, La Jolla, CA, 92093, USA
| | - Douglas D Richman
- San Diego Center for AIDS Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Monserrato, 09042, Italy
- Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, 07100, Italy
| | - Hanh Trinh
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Sandra Sanchez-Reilly
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Joan M Hecht
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Jose A Cadena Zuluaga
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Antonio Anzueto
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jacqueline A Pugh
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Brian K Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | | | - Robert A Clark
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| | - Jason F Okulicz
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Department of Medicine, Infectious Diseases Service, Brooke Army Medical Center, San Antonio, TX, 78234, USA
| | - Weijing He
- VA Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, TX, 78229, USA
| |
Collapse
|
31
|
Huang QQ, Hang Y, Doyle R, Mao Q, Fang D, Pope RM. Mechanisms regulating the loss of Tregs in HUPO mice that develop spontaneous inflammatory arthritis. iScience 2023; 26:106734. [PMID: 37216119 PMCID: PMC10193230 DOI: 10.1016/j.isci.2023.106734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/07/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
T regulatory cells (Tregs) are a potential therapeutic target in many autoimmune diseases including rheumatoid arthritis (RA). The mechanisms responsible for the maintenance of Tregs in chronic inflammatory conditions such as RA are poorly understood. We employed our mouse model of RA in which, the following deletion of Flice-like inhibitory protein in CD11c+ cells, CD11c-FLIP-KO (HUPO) mice develop spontaneous, progressive, erosive arthritis, with reduced Tregs, and the adoptive transfer of Tregs ameliorates the arthritis. HUPO thymic Treg development was normal, but peripheral of Treg Foxp3 was diminished mediated by reduction of dendritic cells and interleukin-2 (IL-2). During chronic inflammatory arthritis Tregs fail to maintain Foxp3, leading to non-apoptotic cell death and conversion to CD4+CD25+Foxp3- cells. Treatment with IL-2 increased Tregs and ameliorated the arthritis. In summary, reduced dendritic cells and IL-2 in the milieu of chronic inflammation, contribute to Treg instability, promoting HUPO arthritis progression, and suggesting a therapeutic approach in RA.
Collapse
Affiliation(s)
- Qi-Quan Huang
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60091, USA
| | - Yiwei Hang
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60091, USA
| | - Renee Doyle
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60091, USA
| | - Qinwen Mao
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Deyu Fang
- Departments of Pathology and Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60091, USA
| | - Richard M. Pope
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60091, USA
| |
Collapse
|
32
|
Mei D, Zhang T, Liu R, Wang P, Hu L, Xu L, Ge J, Zhang X, Wang H, Xue Z, Liang F, Yu Q, Wei W, Zhang L. hIgD-Fc-Ig fusion protein regulates T cell functions by inhibiting TCR signaling pathway in adjuvant arthritis rats. Int Immunopharmacol 2023; 119:110154. [PMID: 37062257 DOI: 10.1016/j.intimp.2023.110154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
This study aimed to investigate the effect of hIgD-Fc-Ig on TCR-Lck-Erk activated by IgD in adjuvant arthritis (AA) rats. Wistar rats were divided into the normal, AA model, hIgD-Fc-Ig (1 mg/kg, 3 mg/kg and 9 mg/kg) and Etanercept (3 mg/kg) groups. The overall index of AA rats was measured every 3 days. The pathologic examination of knee joints and the proliferation of the spleen and thymus of AA rats were detected by H&E staining and CCK-8. The blood flow signal of knee joints of experimental rats was examined by US. The articular bone injury was detected by X-ray. The changes in PBMCs and spleen T cell subsets were detected by flow cytometry. The expression of CD3ε, p-Lck, p-Zap70, Ras, and p-Erk in rat spleens was detected by immunofluorescence and WB. Rat spleen T cells or Jurkat cells treated by IgD to observe the effect of hIgD-Fc-Ig on TCR and its downstream protein expression. The results showed that hIgD-Fc-Ig had a therapeutic effect on AA rats by reducing the secondary inflammation, improving pathological changes. hIgD-Fc-Ig can reduce the ratio of Th cells of PBMCs of AA rats, the ratio of Th, Th1, Th17 cells and increase the ratio of Th2, Treg cells of AA rat spleens. hIgD-Fc-Ig could down-regulate the expression of CD3ε, p-Lck, p-Zap70, Ras, p-Erk in vivo or in vitro. In conclusion, hIgD-Fc-Ig could alleviate the symptoms of AA rats and regulate T cells through TCR-Lck-Erk signaling pathway and maybe a new promising biological agent for RA.
Collapse
Affiliation(s)
- Dan Mei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Tianjing Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Ruijin Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Pan Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Ling Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Li Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Jinru Ge
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Ziyang Xue
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Faqin Liang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Qianqian Yu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
33
|
Yuan Y, Zhang H, Gu Q, Xu X, Yu R, Huang H. Analysis of Th-cell subsets in local and systemic environments from experimental periodontitis rats. Mol Oral Microbiol 2023; 38:83-92. [PMID: 35863754 DOI: 10.1111/omi.12376] [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: 12/16/2021] [Revised: 05/11/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The objective of this study was to explore the effect of periodontitis on Th-cell subsets in local and systemic environments. METHODS A total of 32 male Sprague-Dawley rats were randomly divided into periodontitis and control groups. Silk ligatures were applied to the mandibular first (M1) molars in the periodontitis group. Inflammation and alveolar bone loss around the M1 molars were analyzed by histological staining and microcomputed tomography. The mRNA expression of interferon-γ (IFN-γ), interleukin 4 (IL-4), IL-17, and IL-10 in the gingiva was measured by qRT-PCR. The proportions of Th1, Th2, Th17, and Treg cells in the submandibular lymph nodes, peripheral blood, and jaw bone marrow were tested using flow cytometry. RESULTS More inflammatory cells and alveolar bone resorption were found in the periodontitis group, with upregulated mRNA expression of IFN-γ, IL-17, and IL-10. The proportion of Th1 and Th17 cells was significantly elevated in submandibular lymph nodes, and the proportion of Th1, Th2, and Th17 cells was significantly elevated in peripheral blood, while the proportion of Th1, Th17, and Treg cells was significantly elevated in jaw bone marrow in the periodontitis group. CONCLUSION This study suggests that periodontitis affects the differentiation of Th-cell subsets in both local and systemic environments, resulting in an increased proportion of proinflammatory cells.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hongming Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Qinhua Gu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinrui Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runping Yu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Huang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
34
|
Lopes C, Almeida TC, Pimentel-Nunes P, Dinis-Ribeiro M, Pereira C. Linking dysbiosis to precancerous stomach through inflammation: Deeper than and beyond imaging. Front Immunol 2023; 14:1134785. [PMID: 37063848 PMCID: PMC10102473 DOI: 10.3389/fimmu.2023.1134785] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Upper gastrointestinal endoscopy is considered the gold standard for gastric lesions detection and surveillance, but it is still associated with a non-negligible rate of missing conditions. In the Era of Personalized Medicine, biomarkers could be the key to overcome missed lesions or to better predict recurrence, pushing the frontier of endoscopy to functional endoscopy. In the last decade, microbiota in gastric cancer has been extensively explored, with gastric carcinogenesis being associated with progressive dysbiosis. Helicobacter pylori infection has been considered the main causative agent of gastritis due to its interference in disrupting the acidic environment of the stomach through inflammatory mediators. Thus, does inflammation bridge the gap between gastric dysbiosis and the gastric carcinogenesis cascade and could the microbiota-inflammation axis-derived biomarkers be the answer to the unmet challenge of functional upper endoscopy? To address this question, in this review, the available evidence on the role of gastric dysbiosis and chronic inflammation in precancerous conditions of the stomach is summarized, particularly targeting the nuclear factor-κB (NF-κB), toll-like receptors (TLRs) and cyclooxygenase-2 (COX-2) pathways. Additionally, the potential of liquid biopsies as a non-invasive source and the clinical utility of studied biomarkers is also explored. Overall, and although most studies offer a mechanistic perspective linking a strong proinflammatory Th1 cell response associated with, but not limited to, chronic infection with Helicobacter pylori, promising data recently published highlights not only the diagnostic value of microbial biomarkers but also the potential of gastric juice as a liquid biopsy pushing forward the concept of functional endoscopy and personalized care in gastric cancer early diagnosis and surveillance.
Collapse
Affiliation(s)
- Catarina Lopes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- CINTESIS – Center for Health Technology and Services Research, University of Porto, Porto, Portugal
- ICBAS-UP – Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Tatiana C. Almeida
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Pedro Pimentel-Nunes
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
- Department of Gastroenterology, Unilabs, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Gastroenterology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Carina Pereira
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- CINTESIS – Center for Health Technology and Services Research, University of Porto, Porto, Portugal
- *Correspondence: Carina Pereira,
| |
Collapse
|
35
|
Shen C, Song Y, Fan W, Guo X, Li J, Zhao R. Changes in expression levels of immune cells and inflammatory cytokines in pre-eclampsia patients before and after delivery. J Reprod Immunol 2023; 156:103812. [PMID: 36708687 DOI: 10.1016/j.jri.2023.103812] [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: 09/21/2022] [Revised: 01/01/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVE By analyze the expression levels of immune cells and cytokines secreted by preeclampsia patients before and after delivery. Furthermore, to determine the inflammatory and immunological mechanism responsible for preeclampsia, to provide better future prevention and treatment. METHODS Twenty-one preeclampsia-diagnosed pregnant women were involved in this study as an observation group. Twenty pregnant women with no history of PE, age-matched, were added to the control group. Thirty NP women, age-matched were included as the reference. The levels of cytokines secreted by T, B, NK and Treg immune cells and Th1, Th2 and Th17 cells were detected before and after delivery. RESULTS The number of CD3 + cells in women without preeclampsia before delivery was not statistically different from that in PE patients, but the number of CD4 + and CD8 + cells was lower than that in women without preeclampsia. After delivery, CD4 + cells were larger than those in pregnant women without preeclampsia, while CD8 + cells were smaller than those in pregnant women without preeclampsia. The level of cytokines secreted by Th1 in pregnant women without preeclampsia before delivery was higher than that in PE patients. The levels of cytokines produced by Th1, Th2 and Th17 cells before and after delivery were higher in pregnant women without preeclampsia than in NP women, while Th2 and Th17 were in PE group. CONCLUSION PE patients may reduce CD4 + cells and CD8 + cells, and down-regulate the level of cytokines (especially TNF-β) secreted by Th1 cells to reduce the maternal rejection of embryos, thereby reducing the risk of premature delivery, which is a protective adaptive compensation mechanism in favor of embryos.
Collapse
Affiliation(s)
- Cuihua Shen
- Kunming Maternity and Child Care Hospital Kunming, China
| | - Ying Song
- Kunming Maternity and Child Care Hospital Kunming, China
| | - Wei Fan
- Kunming Maternity and Child Care Hospital Kunming, China
| | - Xiaojuan Guo
- Kunming Maternity and Child Care Hospital Kunming, China
| | - Jia Li
- Kunming Maternity and Child Care Hospital Kunming, China
| | - Renbin Zhao
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Yunnan Provincial Hematopathy Hospital, Kunming, China.
| |
Collapse
|
36
|
Martinez HA, Koliesnik I, Kaber G, Reid JK, Nagy N, Barlow G, Falk BA, Medina CO, Hargil A, Vlodavsky I, Li JP, Pérez-Cruz M, Tang SW, Meyer EH, Wrenshall LE, Lord JD, Garcia KC, Palmer TD, Steinman L, Nepom GT, Wight TN, Bollyky PL, Kuipers HF. FOXP3 + regulatory T cells use heparanase to access IL-2 bound to ECM in inflamed tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.26.529772. [PMID: 36909599 PMCID: PMC10002643 DOI: 10.1101/2023.02.26.529772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
FOXP3+ regulatory T cells (Treg) depend on exogenous IL-2 for their survival and function, but circulating levels of IL-2 are low, making it unclear how Treg access this critical resource in vivo. Here, we show that Treg use heparanase (HPSE) to access IL-2 sequestered by heparan sulfate (HS) within the extracellular matrix (ECM) of inflamed central nervous system tissue. HPSE expression distinguishes human and murine Treg from conventional T cells and is regulated by the availability of IL-2. HPSE-/- Treg have impaired stability and function in vivo, including the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Conversely, endowing Treg with HPSE enhances their ability to access HS-sequestered IL-2 and their tolerogenic function in vivo. Together, these data identify novel roles for HPSE and the ECM in immune tolerance, providing new avenues for improving Treg-based therapy of autoimmunity.
Collapse
Affiliation(s)
- Hunter A Martinez
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Ievgen Koliesnik
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Gernot Kaber
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Jacqueline K Reid
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary; Calgary, Canada
| | - Nadine Nagy
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Graham Barlow
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Ben A Falk
- Matrix Biology Program, Benaroya Research Institute; Seattle, USA
| | - Carlos O Medina
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Aviv Hargil
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Israel Vlodavsky
- Tumor Integrated Cancer Center, Technion-Israel Institute of Technology; Haifa, Israel
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University; Uppsala, Finland
| | - Magdiel Pérez-Cruz
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Sai-Wen Tang
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Everett H Meyer
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Lucile E Wrenshall
- Department of Surgery, Boonshoft School of Medicine, Wright State University; Dayton, USA
| | - James D Lord
- Translational Research Program, Benaroya Research Institute; Seattle, USA
| | - K Christopher Garcia
- Department of Molecular & Cellular Physiology, Stanford University; Stanford, USA
| | - Theo D Palmer
- Department of Neurosurgery, Stanford University School of Medicine; Stanford, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine; Stanford, USA
| | - Gerald T Nepom
- Immune Tolerance Network, Benaroya Research Institute; Seattle, USA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute; Seattle, USA
| | - Paul L Bollyky
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
| | - Hedwich F Kuipers
- Department of Medicine, Stanford University School of Medicine; Stanford, USA
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary; Calgary, Canada
| |
Collapse
|
37
|
Rodriguez-Sevilla JJ, Adema V, Garcia-Manero G, Colla S. Emerging treatments for myelodysplastic syndromes: Biological rationales and clinical translation. Cell Rep Med 2023; 4:100940. [PMID: 36787738 PMCID: PMC9975331 DOI: 10.1016/j.xcrm.2023.100940] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
Myelodysplastic syndromes (MDSs) are a heterogeneous group of clonal hematopoietic stem cell disorders characterized by myeloid dysplasia, peripheral blood cytopenias, and increased risk of progression to acute myeloid leukemia (AML). The standard of care for patients with MDS is hypomethylating agent (HMA)-based therapy; however, nearly 50% of patients have no response to the treatment. Patients with MDS in whom HMA therapy has failed have a dismal prognosis and no approved second-line therapy options, so enrollment in clinical trials of experimental agents represents these patients' only chance for improved outcomes. A better understanding of the molecular and biological mechanisms underpinning MDS pathogenesis has enabled the development of new agents that target molecular alterations, cell death regulators, signaling pathways, and immune regulatory proteins in MDS. Here, we review novel therapies for patients with MDS in whom HMA therapy has failed, with an emphasis on the biological rationale for these therapies' development.
Collapse
Affiliation(s)
| | - Vera Adema
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
38
|
Lee WH, Kim GE, Hong KJ, Kim HS, Lee GR. Insulin Receptor Substrate 1 Signaling Inhibits Foxp3 Expression and Suppressive Functions in Treg Cells through the mTORC1 Pathway. Int J Mol Sci 2023; 24:2551. [PMID: 36768873 PMCID: PMC9917118 DOI: 10.3390/ijms24032551] [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: 12/13/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Regulatory T (Treg) cells play an important role in immune homeostasis by inhibiting cells within the innate and adaptive immune systems; therefore, the stability and immunosuppressive function of Treg cells need to be maintained. In this study, we found that the expression of insulin receptor substrate 1 (IRS1) by Treg cells was lower than that by conventional CD4 T cells. IRS1-overexpressing Treg cells showed the downregulated expression of FOXP3, as well as Treg signature markers CD25 and CTLA4. IRS1-overexpressing Treg cells also showed diminished immunosuppressive functions in an in vitro suppression assay. Moreover, IRS1-overexpressing Treg cells were unable to suppress the pathogenic effects of conventional T cells in a transfer-induced colitis model. IRS1 activated the mTORC1 signaling pathway, a negative regulator of Treg cells. Moreover, IRS1 destabilized Treg cells by upregulating the expression of IFN-γ and Glut1. Thus, IRS1 acts as a negative regulator of Treg cells by downregulating the expression of FOXP3 and disrupting stability.
Collapse
Affiliation(s)
| | | | | | | | - Gap Ryol Lee
- Department of Life Science, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
39
|
Li Q, Lu J, Li J, Zhang B, Wu Y, Ying T. Antibody-based cancer immunotherapy by targeting regulatory T cells. Front Oncol 2023; 13:1157345. [PMID: 37182149 PMCID: PMC10174253 DOI: 10.3389/fonc.2023.1157345] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Regulatory T cells (Tregs) are among the most abundant suppressive cells, which infiltrate and accumulate in the tumor microenvironment, leading to tumor escape by inducing anergy and immunosuppression. Their presence has been correlated with tumor progression, invasiveness and metastasis. Targeting tumor-associated Tregs is an effective addition to current immunotherapy approaches, but it may also trigger autoimmune diseases. The major limitation of current therapies targeting Tregs in the tumor microenvironment is the lack of selective targets. Tumor-infiltrating Tregs express high levels of cell surface molecules associated with T-cell activation, such as CTLA4, PD-1, LAG3, TIGIT, ICOS, and TNF receptor superfamily members including 4-1BB, OX40, and GITR. Targeting these molecules often attribute to concurrent depletion of antitumor effector T-cell populations. Therefore, novel approaches need to improve the specificity of targeting Tregs in the tumor microenvironment without affecting peripheral Tregs and effector T cells. In this review, we discuss the immunosuppressive mechanisms of tumor-infiltrating Tregs and the status of antibody-based immunotherapies targeting Tregs.
Collapse
Affiliation(s)
- Quanxiao Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Baohong Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Tianlei Ying, ; Yanling Wu,
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Tianlei Ying, ; Yanling Wu,
| |
Collapse
|
40
|
Chen J, Mei A, Wei Y, Li C, Qian H, Min X, Yang H, Dong L, Rao X, Zhong J. GLP-1 receptor agonist as a modulator of innate immunity. Front Immunol 2022; 13:997578. [PMID: 36569936 PMCID: PMC9772276 DOI: 10.3389/fimmu.2022.997578] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a 30-amino acid hormone secreted by L cells in the distal ileum, colon, and pancreatic α cells, which participates in blood sugar regulation by promoting insulin release, reducing glucagon levels, delaying gastric emptying, increasing satiety, and reducing appetite. GLP-1 specifically binds to the glucagon-like peptide-1 receptor (GLP-1R) in the body, directly stimulating the secretion of insulin by pancreatic β-cells, promoting proliferation and differentiation, and inhibiting cell apoptosis, thereby exerting a glycemic lowering effect. The glycemic regulating effect of GLP-1 and its analogues has been well studied in human and murine models in the circumstance of many diseases. Recent studies found that GLP-1 is able to modulate innate immune response in a number of inflammatory diseases. In the present review, we summarize the research progression of GLP-1 and its analogues in immunomodulation and related signal pathways.
Collapse
Affiliation(s)
- Jun Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, China
| | - Aihua Mei
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunlei Li
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, China
| | - Hang Qian
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoquan Rao
- Department of Cardiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
41
|
Mousstaaid A, Fatemi SA, Elliott KEC, Levy AW, Miller WW, Gerard PD, Alqhtani AH, Peebles ED. Effects of the In Ovo and Dietary Supplementation of L-Ascorbic Acid on the Growth Performance, Inflammatory Response, and Eye L-Ascorbic Acid Concentrations in Ross 708 Broiler Chickens. Animals (Basel) 2022; 12:2573. [PMID: 36230314 PMCID: PMC9559629 DOI: 10.3390/ani12192573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
Effects of the dietary and in ovo administration of L-ascorbic acid (L-AA) on the performance, plasma nitric oxide, and eye L-AA concentrations of Ross 708 broilers were investigated. At 17 days of incubation, live embryonated hatching eggs were randomly assigned to a non-injected or sham-injected (100 μL of saline) control group, or a group injected with either 12 or 25 mg of L-AA suspended in 100 μL of saline. Chicks received a commercial diet with or without 200 mg/kg of supplemental L-AA and were randomly assigned to each of 6 replicate floor pens in each in ovo injection-dietary treatment combination. Weekly live performance variables through 14 days of post hatch age (doa) and the eye weights in both sexes at 0, 7, and 14 doa were determined. At 0 and 14 doa, plasma nitric oxide levels and eye L-AA concentrations of one bird of each sex in each pen were determined. Dietary supplemental L-AA decreased feed intake and growth between 0 and 7 doa, but from 8 to 14 doa; all birds fed supplemental L-AA had a lower feed conversion ratio. At 14 doa, male chicks had higher eye L-AA concentrations and lower plasma nitric oxide levels when treated in ovo with 12 mg of L-AA. In conclusion, dietary L-AA may be used to improve feed conversion in the second week of broiler post hatch growth. However, the in ovo administration of 12 mg of L-AA can increase male eye L-AA concentrations and is effective in reducing their general inflammatory response.
Collapse
Affiliation(s)
- Ayoub Mousstaaid
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| | | | | | | | - William Wadd Miller
- Advanced Animal Eye Care, 3308 Old West Point Road, Starkville, MS 39759, USA
| | - Patrick D. Gerard
- Department of Mathematical Sciences, Clemson University, Clemson, SC 29634, USA
| | | | - Edgar David Peebles
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| |
Collapse
|
42
|
Cui MY, Yi X, Cao ZZ, Zhu DX, Wu J. Targeting Strategies for Aberrant Lipid Metabolism Reprogramming and the Immune Microenvironment in Esophageal Cancer: A Review. JOURNAL OF ONCOLOGY 2022; 2022:4257359. [PMID: 36106333 PMCID: PMC9467784 DOI: 10.1155/2022/4257359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022]
Abstract
Esophageal cancer is of high importance to occurrence, development, and treatment resistance. As evidenced by recent studies, pathways (e.g., Wnt/β-catenin, AMPK, and Hippo) are critical to the proliferation, differentiation, and self-renewal of esophageal cancer. In addition, the above pathways play a certain role in regulating esophageal cancer and act as potential therapeutic targets. Over the past few years, the function of lipid metabolism in controlling tumor cells and immune cells has aroused extensive attention. It has been reported that there are intricate interactions between lipid metabolism reprogramming between immune and esophageal cancer cells, whereas molecular mechanisms should be studied in depth. Immune cells have been commonly recognized as a vital player in the esophageal cancer microenvironment, having complex crosstalk with cancer cells. It is increasingly evidenced that the function of immune cells in the tumor microenvironment (TME) is significantly correlated with abnormal lipid metabolism. In this review, the latest findings in lipid metabolism reprogramming in TME are summarized, and the above findings are linked to esophageal cancer progression. Aberrant lipid metabolism and associated signaling pathways are likely to serve as a novel strategy to treat esophageal cancer through lipid metabolism reprogramming.
Collapse
Affiliation(s)
- Meng-Ying Cui
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xing Yi
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Zhen-Zhen Cao
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Dan-Xia Zhu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jun Wu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
43
|
Taheri M, Roustapour S, Gholipour M, Hussen BM, Eslami S, Ghafouri-Fard S, Sayad A. Analysis of expression of regulatory T cell related lncRNAs in inflammatory demyelinating polyneuropathies. Int Immunopharmacol 2022; 112:109188. [PMID: 36041257 DOI: 10.1016/j.intimp.2022.109188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/05/2022]
Abstract
Long non-coding RNAs that regulate function of regulatory T cells can affect pathoetiology of autoimmune disorders, such as inflammatory demyelinating polyneuropathies. In the current case-control study, we compared expression of four of these lncRNAs, namely FLICR, NEST, RMRP and TH2-LCR between patients with inflammatory demyelinating polyneuropathies and healthy subjects. Expressions of RMRP, NEST and FLICR were higher in total patients compared with controls. However, there was no significant difference in their expressions between acute and chronic demyelinating polyneuropathies. In addition, interaction of gender and disease factors had significant effect on expression levels of RMRP and TH2-LCR genes in subgroups. RMRR was superior to other lncRNAs in terms of AUC, sensitivity and specificity values in total patients and both subgroups of patients. This lncRNA could separate total patients, female patients and male patients from corresponding controls with AUC values (±SD) of 0.9 ± 0.03, 0.86 ± 0.07 and 0.93 ± 0.03, respectively. FLICR ranked second in this regard, since it could separate total patients, female patients and male patients from corresponding controls with AUC values (±SD) of 0.81 ± 0.03, 0.72 ± 0.07 and 0.87 ± 0.04, respectively. Therefore, our study provides evidence for participation of regulatory T cells-related lncRNAs in the pathoetiology of inflammatory demyelinating polyneuropathies.
Collapse
Affiliation(s)
- Mohammad Taheri
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | | | - Mahdi Gholipour
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq; Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region, Iraq
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran; Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Mohd Idris RA, Mussa A, Ahmad S, Al-Hatamleh MAI, Hassan R, Tengku Din TADAA, Wan Abdul Rahman WF, Lazim NM, Boer JC, Plebanski M, Mohamud R. The Effects of Tamoxifen on Tolerogenic Cells in Cancer. BIOLOGY 2022; 11:1225. [PMID: 36009853 PMCID: PMC9405160 DOI: 10.3390/biology11081225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
Tamoxifen (TAM) is the most prescribed selective estrogen receptor modulator (SERM) to treat hormone-receptor-positive breast cancer patients and has been used for more than 20 years. Its role as a hormone therapy is well established; however, the potential role in modulating tolerogenic cells needs to be better clarified. Infiltrating tumor-microenvironment-regulatory T cells (TME-Tregs) are important as they serve a suppressive function through the transcription factor Forkhead box P3 (Foxp3). Abundant studies have suggested that Foxp3 regulates the expression of several genes (CTLA-4, PD-1, LAG-3, TIM-3, TIGIT, TNFR2) involved in carcinogenesis to utilize its tumor suppressor function through knockout models. TAM is indirectly concomitant via the Cre/loxP system by allowing nuclear translocation of the fusion protein, excision of the floxed STOP cassette and heritable expression of encoding fluorescent protein in a cohort of cells that express Foxp3. Moreover, TAM administration in breast cancer treatment has shown its effects directly through MDSCs by the enrichment of its leukocyte populations, such as NK and NKT cells, while it impairs the differentiation and activation of DCs. However, the fundamental mechanisms of the reduction of this pool by TAM are unknown. Here, we review the vital effects of TAM on Tregs for a precise mechanistic understanding of cancer immunotherapies.
Collapse
Affiliation(s)
- Ros Akmal Mohd Idris
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Ali Mussa
- Haematology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, Omdurman P.O. Box 382, Sudan
| | - Suhana Ahmad
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Mohammad A. I. Al-Hatamleh
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Rosline Hassan
- Haematology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | | | - Wan Faiziah Wan Abdul Rahman
- Pathology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Norhafiza Mat Lazim
- Otorhinolaryngology Department-Head & Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Jennifer C. Boer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Rohimah Mohamud
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| |
Collapse
|
45
|
Alrumaihi F. The Multi-Functional Roles of CCR7 in Human Immunology and as a Promising Therapeutic Target for Cancer Therapeutics. Front Mol Biosci 2022; 9:834149. [PMID: 35874608 PMCID: PMC9298655 DOI: 10.3389/fmolb.2022.834149] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
An important hallmark of the human immune system is to provide adaptive immunity against pathogens but tolerance toward self-antigens. The CC-chemokine receptor 7 (CCR7) provides a significant contribution in guiding cells to and within lymphoid organs and is important for acquiring immunity and tolerance. The CCR7 holds great importance in establishing thymic architecture and function and naïve and regulatory T-cell homing in the lymph nodes. Similarly, the receptor is a key regulator in cancer cell migration and the movement of dendritic cells. This makes the CCR7 an important receptor as a drug and prognostic marker. In this review, we discussed several biological roles of the CCR7 and its importance as a drug and prognostic marker.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
46
|
Cervantes O, Talavera IC, Every E, Coler B, Li M, Li A, Li H, Adams Waldorf K. Role of hormones in the pregnancy and sex-specific outcomes to infections with respiratory viruses. Immunol Rev 2022; 308:123-148. [PMID: 35373371 PMCID: PMC9189035 DOI: 10.1111/imr.13078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/13/2023]
Abstract
Pregnant women infected with pathogenic respiratory viruses, such as influenza A viruses (IAV) and coronaviruses, are at higher risk for mortality, hospitalization, preterm birth, and stillbirth. Several factors are likely to contribute to the susceptibility of pregnant individuals to severe lung disease including changes in pulmonary physiology, immune defenses, and effector functions of some immune cells. Pregnancy is also a physiologic state characterized by higher levels of multiple hormones that may impact the effector functions of immune cells, such as progesterone, estrogen, human chorionic gonadotropin, prolactin, and relaxin. Each of these hormones acts to support a tolerogenic immune state of pregnancy, which helps prevent fetal rejection, but may also contribute to an impaired antiviral response. In this review, we address the unique role of adaptive and innate immune cells in the control of pathogenic respiratory viruses and how pregnancy and specific hormones can impact their effector actions. We highlight viruses with sex-specific differences in infection outcomes and why pregnancy hormones may contribute to fetal protection but aid the virus at the expense of the mother's health.
Collapse
Affiliation(s)
- Orlando Cervantes
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Irene Cruz Talavera
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Emma Every
- University of Washington School of Medicine, Spokane, Washington, United States of America
| | - Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, United States of America
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Department of Biological Sciences, Columbia University, New York City, New York, United States of America
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Case Western Reserve, Cleveland, Ohio, United States of America
| | - Hanning Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
47
|
A novel lipidic peptide with potential to promote balanced effector-regulatory T cell responses. Sci Rep 2022; 12:11185. [PMID: 35778468 PMCID: PMC9249808 DOI: 10.1038/s41598-022-15455-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/23/2022] [Indexed: 11/08/2022] Open
Abstract
T cell-dendritic cell (DC) interactions contribute to reciprocal stimulation leading to DC maturation that results in production of interleukin-12 (IL-12) and interferon-gamma (IFN-γ). Both cytokines have been implicated in autoimmune diseases while being necessary for effective immune responses against foreign antigens. We describe a lipidic peptide, designated IK14004, that modifies crosstalk between T cells and DCs resulting in suppression of IL-12p40/IFN-γ production. T cell production of interleukin-2 (IL-2) and IFN-γ is uncoupled and IL-12p70 production is enhanced. IK14004 induces expression of activating co-receptors in CD8+ T cells and increases the proportion of Foxp3-expressing CD4+ T regulatory cells. The potential for IK14004 to impact on signalling pathways required to achieve a balanced immune response upon stimulation of DCs and T cells is highlighted. This novel compound provides an opportunity to gain further insights into the complexity of T cell-DC interactions relevant to autoimmunity associated with malignancies and may have therapeutic benefit.
Collapse
|
48
|
New insights for regulatory T cell in lupus nephritis. Clin Exp Rheumatol 2022; 21:103134. [PMID: 35690245 DOI: 10.1016/j.autrev.2022.103134] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022]
Abstract
Lupus nephritis (LN) is a complicated autoimmune disease marked by out-of-balance of immunological reactivity and immune tolerance. With the advance of immunotherapy in human disease, regulatory T (Treg) cells serve a crucial function in immune tolerance regulation and are characterized with suppression function as one of the most important research hotspots for autoimmunity diseases. In recent years, Treg cells have shown the robust potential for treatment to autoimmunity diseases like type I diabetic mellitus and rheumatoid arthritis. However, Treg cell therapy is poorly understood for LN patients. This review aims to summarize new insights for Treg-targeting techniques in LN patients. The current data regarding the biology features of Treg cells in LN patients is discussed. The propotion of Treg cells in LN patients have contradictory results regarding the use of different molecular markers. Forkhead box protein 3 (FOXP3) are hallmarks for control function of Treg cells. Treg cells can directly or indirectly target T cells and B cells by playing supressive role. The molecular targets for Treg cells in LN patients includes gene variants, miRNAs, and inflammatory related factors. Based on the current knowledge of Treg cell biology, several therapeutic strategies could be used to treat LN: cell transplantation, low dose IL-2 treatment, drugs target the balance of Treg and type 17 T helper (Th17) cells, and Chinese medicine.
Collapse
|
49
|
Tumor-Associated Regulatory T Cells in Non-Small-Cell Lung Cancer: Current Advances and Future Perspectives. J Immunol Res 2022; 2022:4355386. [PMID: 35497874 PMCID: PMC9054468 DOI: 10.1155/2022/4355386] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most threatening malignant tumors to human health, with the overall 5-year survival rate being less than 30%. Regulatory T cells (Tregs), a functional subset of T cells, maintain immunologic immunological self-tolerance and homeostasis. Accumulating evidence has uncovered their implicated roles in various cancers in recent years. In NSCLC, they are associated with staging, therapeutic efficacy, and prognosis by infiltrating in tissues and thereby attenuating immunologic anticancer effects in patients. Tumor-associated Tregs display distinct immune signatures in NSCLC compared to thymus-derived Tregs, playing an important role in remodeling the tumor microenvironment (TME). Targeting Tregs has become a novel direction for NSCLC patients, such as disrupting their immune-suppressive functions, blocking their trafficking into tumors, and inhibiting their development and/or activation. This review is aimed at elucidating the molecular mechanisms of tumor-associated Tregs in NSCLC and providing therapeutic targets relevant to Tregs.
Collapse
|
50
|
Traxinger B, Vick SC, Woodward-Davis A, Voillet V, Erickson JR, Czartoski J, Teague C, Prlic M, Lund JM. Mucosal viral infection induces a regulatory T cell activation phenotype distinct from tissue residency in mouse and human tissues. Mucosal Immunol 2022; 15:1012-1027. [PMID: 35821289 PMCID: PMC9391309 DOI: 10.1038/s41385-022-00542-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023]
Abstract
Regulatory T cells (Tregs) mediate immune homeostasis, yet also facilitate nuanced immune responses during infection, balancing pathogen control while limiting host inflammation. Recent studies have identified Treg populations in non-lymphoid tissues that are phenotypically distinct from Tregs in lymphoid tissues (LT), including performance of location-dependent roles. Mucosal tissues serve as critical barriers to microbes while performing unique physiologic functions, so we sought to identify distinct phenotypical and functional aspects of mucosal Tregs in the female reproductive tract. In healthy human and mouse vaginal mucosa, we found that Tregs are highly activated compared to blood or LT Tregs. To determine if this phenotype reflects acute activation or a general signature of vaginal tract (VT)-residency, we infected mice with HSV-2 to discover that VT Tregs express granzyme-B (GzmB) and acquire a VT Treg signature distinct from baseline. To determine the mechanisms that drive GzmB expression, we performed ex vivo assays to reveal that a combination of type-I interferons and interleukin-2 is sufficient for GzmB expression. Together, we highlight that VT Tregs are activated at steady state and become further activated in response to infection; thus, they may exert robust control of local immune responses, which could have implications for mucosal vaccine design.
Collapse
Affiliation(s)
- Brianna Traxinger
- Department of Global Health, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Sarah C Vick
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | | | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Jami R Erickson
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Julie Czartoski
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Candice Teague
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Martin Prlic
- Department of Global Health, University of Washington, Seattle, WA, USA.
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| | - Jennifer M Lund
- Department of Global Health, University of Washington, Seattle, WA, USA.
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA.
| |
Collapse
|