1
|
Rabbani P, Ramkhelawon B, Cronstein BN. Adenosine metabolism and receptors in aging of the skin, musculoskeletal, immune and cardiovascular systems. Ageing Res Rev 2025; 106:102695. [PMID: 39971100 PMCID: PMC11960428 DOI: 10.1016/j.arr.2025.102695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/26/2024] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Aging populations worldwide face an increasing burden of age-related chronic conditions, necessitating a deeper understanding of the underlying mechanisms. Purine metabolism has emerged as a crucial player in the pathophysiology of aging, affecting various tissues and organs. Dysregulation of purine metabolism, particularly alterations in extracellular adenosine levels and adenosine receptor signaling, contributes to age-related musculoskeletal problems, cardiovascular diseases, inflammation, and impaired immune responses. Changes in purine metabolism are associated with diminished tissue repair and regeneration, altered bone density, and impaired muscle regeneration. Mechanistically, age-related alterations in purine metabolism involve reductions in extracellular adenosine production, impaired autocrine signaling, and dysregulated expression of CD73 and CD39. Targeting adenosine receptors, such as A2A and A2B receptors, emerges as a promising therapeutic approach to mitigate age-related conditions, including sarcopenia, obesity, osteoarthritis, and impaired wound healing. Since we cannot reverse time, understanding the intricate molecular interplay between purine metabolism and aging-related pathologies holds significant potential for developing novel therapeutic strategies to improve the health and quality of life of aging populations. In this review, we compile the findings related to purine metabolism during aging in several tissues and organs and provide insights into how these signals can be manipulated to circumvent the deleterious effects of the passage of time on our body.
Collapse
Affiliation(s)
- Piul Rabbani
- Hansjorg Wyss Department of Plastic Surgery, New York University Langone Health, New York, NY, USA
| | - Bhama Ramkhelawon
- Division of Vascular Surgery, Department of Surgery, New York University Langone Health, New York, NY, USA; Department of Cell Biology, New York University Langone Health, New York, NY, USA
| | - Bruce N Cronstein
- Department of Medicine, Divisions of Rheumatology and Precision Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
2
|
Choi M, Choi S, Cho M, Kim C. Metabolic Signaling as a Driver of T Cell Aging. Immune Netw 2025; 25:e14. [PMID: 40078788 PMCID: PMC11896665 DOI: 10.4110/in.2025.25.e14] [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: 01/05/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Aging significantly diminishes T cell immunity, increasing susceptibility to infections and reducing vaccine efficacy in older individuals. Metabolism plays a key role in T cell function, shaping their energy requirements, activation, and differentiation. Recent studies highlight altered metabolic signaling as a pivotal factor in T cell aging, influencing the ability of T cells to maintain quiescence, respond to activation, and differentiate into functional subsets. Aberrant metabolic pathways disrupt the quiescence of aged T cells and skew their differentiation toward short-lived, pro-inflammatory effector T cells while hindering the generation of long-lived memory and T follicular helper cells. These changes contribute to a hyper-inflammatory state, exacerbate chronic low-grade inflammation, and compromise immune homeostasis. In this review, we explore how metabolic signaling is altered during T cell aging and the resulting functional impacts. We also discuss therapeutic approaches aimed at restoring proper T cell differentiation, improving vaccine responses, and rejuvenating immune function in older populations.
Collapse
Affiliation(s)
- Minju Choi
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| | - Sujin Choi
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| | - Minkyeong Cho
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| | - Chulwoo Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02708, Korea
| |
Collapse
|
3
|
Pruitt L, Abbott RK. Hypoxia-adenosinergic regulation of B cell responses. Front Immunol 2024; 15:1478506. [PMID: 39559353 PMCID: PMC11570280 DOI: 10.3389/fimmu.2024.1478506] [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: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Hypoxic microenvironments induce widespread metabolic changes that have been shown to be critical in regulating innate and adaptive immune responses. Hypoxia-induced changes include the generation of extracellular adenosine followed by subsequent signaling through adenosine receptors on immune cells. This evolutionarily conserved "hypoxia-adenosinergic" pathway of hypoxia → extracellular adenosine → adenosine receptor signaling has been shown to be critical in limiting and redirecting T cell responses including in tumor microenvironments and the gut mucosa. However, the question of whether hypoxic microenvironments are involved in the development of B cell responses has remained unexplored until recently. The discovery that germinal centers (GC), the anatomic site in which B cells undergo secondary diversification and affinity maturation, develop a hypoxic microenvironment has sparked new interest in how this evolutionarily conserved pathway affects antibody responses. In this review we will summarize what is known about hypoxia-adenosinergic microenvironments in lymphocyte development and ongoing immune responses. Specific focus will be placed on new developments regarding the role of the hypoxia-adenosinergic pathway in regulating GC development and humoral immunity.
Collapse
Affiliation(s)
| | - Robert K. Abbott
- Department of Pathology, University of Texas Medical Branch,
Galveston, TX, United States
| |
Collapse
|
4
|
da Rocha Torres Pavione N, de Moraes JVB, Ribeiro IC, de Castro RB, da Silva W, de Souza ACA, da Silva VHF, de Souza Vasconcellos R, da Costa Bressan G, Fietto JLR. Heterologous expression and biochemical characterization of the recombinant nucleoside triphosphate diphosphohydrolase 2 (LbNTPDase2) from Leishmania (Viannia) braziliensis. Purinergic Signal 2024; 20:509-520. [PMID: 37999896 PMCID: PMC11377403 DOI: 10.1007/s11302-023-09980-9] [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: 09/11/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Leishmania braziliensis is a pathogenic protozoan parasite that causes American Tegumentary Leishmaniasis (ATL), an important tropical neglected disease. ENTPDases are nucleotidases that hydrolyze intracellular and/or extracellular nucleotides. ENTPDases are known as regulators of purinergic signalling induced by extracellular nucleotides. Leishmania species have two isoforms of ENTPDase, and, particularly, ENTPDase2 seems to be involved in infectivity and virulence. In this study, we conducted the heterologous expression and biochemical characterization of the recombinant ENTPDase2 of L. braziliensis (rLbNTPDase2). Our results show that this enzyme is a canonical ENTPDase with apyrase activity, capable of hydrolysing triphosphate and diphosphate nucleotides, and it is dependent on divalent cations (calcium or magnesium). Substrate specificity was characterized as UDP>GDP>ADP>GTP>ATP=UTP. The enzyme showed optimal activity at a neutral to basic pH and was partially inhibited by suramin and DIDS. Furthermore, the low apparent Km for ADP suggests that the enzyme may play a role in adenosine-mediated signalling. The biochemical characterization of this enzyme can open new avenues for using LbNTPDase2 as a drug target.
Collapse
Affiliation(s)
- Nancy da Rocha Torres Pavione
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
- General Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - João Victor Badaró de Moraes
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
- General Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Isadora Cunha Ribeiro
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Raissa Barbosa de Castro
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Walmir da Silva
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | | | | | | - Gustavo da Costa Bressan
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | |
Collapse
|
5
|
Di Virgilio F, Vultaggio-Poma V, Tarantini M, Giuliani AL. Overview of the role of purinergic signaling and insights into its role in cancer therapy. Pharmacol Ther 2024; 262:108700. [PMID: 39111410 DOI: 10.1016/j.pharmthera.2024.108700] [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: 01/07/2024] [Revised: 07/05/2024] [Accepted: 07/31/2024] [Indexed: 08/30/2024]
Abstract
Innovation of cancer therapy has received a dramatic acceleration over the last fifteen years thanks to the introduction of the novel immune checkpoint inhibitors (ICI). On the other hand, the conspicuous scientific knowledge accumulated in purinergic signaling since the early seventies is finally being transferred to the clinic. Several Phase I/II clinical trials are currently underway to investigate the effect of drugs interfering with purinergic signaling as stand-alone or combination therapy in cancer. This is supporting the novel concept of "purinergic immune checkpoint" (PIC) in cancer therapy. In the present review we will address a) the basic pharmacology and cell biology of the purinergic system; b) principles of its pathophysiology in human diseases; c) implications for cell death, cell proliferation and cancer; d) novel molecular tools to investigate nucleotide homeostasis in the extracellular environment; e) recent developments in the pharmacology of P1, P2 receptors and related ecto-enzymes; f) P1 and P2 ligands as novel diagnostic tools; g) current issues in PIC-based anti-cancer therapy. This review will provide an appraisal of the current status of purinergic signaling in cancer and will help identify future avenues of development.
Collapse
Affiliation(s)
| | | | - Mario Tarantini
- Department of Medical Sciences, University of Ferrara, Italy
| | | |
Collapse
|
6
|
Fisher JS, Adán‐Barrientos I, Kumar NR, Lancaster JN. The aged microenvironment impairs BCL6 and CD40L induction in CD4 + T follicular helper cell differentiation. Aging Cell 2024; 23:e14140. [PMID: 38481058 PMCID: PMC11296098 DOI: 10.1111/acel.14140] [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: 10/03/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024] Open
Abstract
Weakened germinal center responses by the aged immune system result in diminished immunity against pathogens and reduced efficacy of vaccines. Prolonged contacts between activated B cells and CD4+ T cells are crucial to germinal center formation and T follicular helper cell (Tfh) differentiation, but it is unclear how aging impacts the quality of this interaction. Peptide immunization confirmed that aged mice have decreased expansion of antigen-specific germinal center B cells and reduced antibody titers. Furthermore, aging was associated with accumulated Tfh cells, even in naïve mice. Despite increased numbers, aged Tfh had reduced expression of master transcription factor BCL6 and increased expression of the ectonucleotidase CD39. In vitro activation revealed that proliferative capacity was maintained in aged CD4+ T cells, but not the costimulatory molecule CD40L. When activated in vitro by aged antigen-presenting cells, young CD4+ naïve T cells generated reduced numbers of activated cells with upregulated CD40L. To determine the contribution of cell-extrinsic influences on antigen-specific Tfh induction, young, antigen-specific B and CD4+ T cells were adoptively transferred into aged hosts prior to peptide immunization. Transferred cells had reduced expansion and differentiation into germinal center B cell and Tfh and reduced antigen-specific antibody titers when compared to young hosts. Young CD4+ T cells transferred aged hosts differentiated into Tfh cells with reduced PD-1 and BCL6 expression, and increased CD39 expression, though they maintained their mitochondrial capacity. These results highlight the role of the lymphoid microenvironment in modulating CD4+ T cell differentiation, which contributes to impaired establishment and maintenance of germinal centers.
Collapse
Affiliation(s)
| | - Irene Adán‐Barrientos
- Immunobiology LaboratoryCentro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Naveen R. Kumar
- Department of ImmunologyMayo ClinicScottsdaleArizonaUSA
- School of Life SciencesArizona State UniversityTempeArizonaUSA
| | - Jessica N. Lancaster
- Department of ImmunologyMayo ClinicScottsdaleArizonaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of Cancer BiologyMayo ClinicScottsdaleArizonaUSA
| |
Collapse
|
7
|
Zeng J, Liao Z, Yang H, Wang Q, Wu Z, Hua F, Zhou Z. T cell infiltration mediates neurodegeneration and cognitive decline in Alzheimer's disease. Neurobiol Dis 2024; 193:106461. [PMID: 38437992 DOI: 10.1016/j.nbd.2024.106461] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder with pathological features of β-amyloid (Aβ) and hyperphosphorylated tau protein accumulation in the brain, often accompanied by cognitive decline. So far, our understanding of the extent and role of adaptive immune responses in AD has been quite limited. T cells, as essential members of the adaptive immune system, exhibit quantitative and functional abnormalities in the brains of AD patients. Dysfunction of the blood-brain barrier (BBB) in AD is considered one of the factors leading to T cell infiltration. Moreover, the degree of neuronal loss in AD is correlated with the quantity of T cells. We first describe the differentiation and subset functions of peripheral T cells in AD patients and provide an overview of the key findings related to BBB dysfunction and how T cells infiltrate the brain parenchyma through the BBB. Furthermore, we emphasize the risk factors associated with AD, including Aβ, Tau protein, microglial cells, apolipoprotein E (ApoE), and neuroinflammation. We discuss their regulation of T cell activation and proliferation, as well as the connection between T cells, neurodegeneration, and cognitive decline. Understanding the innate immune response is crucial for providing comprehensive personalized therapeutic strategies for AD.
Collapse
Affiliation(s)
- Junjian Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Zhiqiang Liao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Hanqin Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Qiong Wang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Zhiyong Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China.
| | - Zhidong Zhou
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China.
| |
Collapse
|
8
|
Wellford SA, Schwartzberg PL. Help me help you: emerging concepts in T follicular helper cell differentiation, identity, and function. Curr Opin Immunol 2024; 87:102421. [PMID: 38733669 PMCID: PMC11482284 DOI: 10.1016/j.coi.2024.102421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
Effective high-affinity, long-term humoral immunity requires T cell help provided by a subset of differentiated CD4+ T cells known as T follicular helper (Tfh) cells. Classically, Tfh cells provide contact-dependent help for the generation of germinal centers (GCs) in secondary lymphoid organs (SLOs). Recent studies have expanded the conventional definition of Tfh cells, revealing new functions, new descriptions of Tfh subsets, new factors regulating Tfh differentiation, and new roles outside of SLO GCs. Together, these data suggest that one Tfh is not equivalent to another, helping redefine our understanding of Tfh cells and their biology.
Collapse
Affiliation(s)
- Sebastian A Wellford
- Cell Signalling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pamela L Schwartzberg
- Cell Signalling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
9
|
To JC, Gao S, Li XX, Zhao Y, Keng VW. Sorafenib Resistance Contributed by IL7 and MAL2 in Hepatocellular Carcinoma Can Be Overcome by Autophagy-Inducing Stapled Peptides. Cancers (Basel) 2023; 15:5280. [PMID: 37958451 PMCID: PMC10650575 DOI: 10.3390/cancers15215280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/26/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Drug resistance poses a great challenge in systemic therapy for hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms associated with resistance to anti-cancer drugs, such as Sorafenib, remain unclear. In this study, we use transposon insertional mutagenesis to generate Sorafenib-resistant HCC cell lines in order to identify potential drug resistant causative genes. Interleukin 7 (IL7) and mal, T cell differentiation protein 2 (MAL2) were identified as candidate genes that promote survival by activating JAK/STAT and PI3K/AKT signaling pathways. Sorafenib-resistant cells exhibited higher clonogenic survival and lower drug sensitivity due to IL7 and MAL2 upregulation. Higher anti-apoptotic effect, clonogenic survival and increased PI3K/AKT/STAT3 activities were observed in IL7 and MAL2 co-overexpressing cells compared with controls or cells overexpressing IL7 or MAL2 individually. Given the critical role of MAL2 in endocytosis, we propose that MAL2 might facilitate the endocytic trafficking of IL7 and its cognate receptors to the plasma membrane, which leads to upregulated JAK/STAT and PI3K/AKT signaling pathways and Sorafenib resistance. Additionally, our previous studies showed that an autophagy-inducing stapled peptide promoted the endolysosomal degradation of c-MET oncogene and overcame adaptive Sorafenib resistance in c-MET+ HCC cells. In this study, we demonstrate that these stapled peptides readily induced autophagy and inhibited the proliferation of both wild-type and Sorafenib-resistant HCC cells co-overexpressing both IL7 and MAL2. Furthermore, these peptides showed synergistic cytotoxicity with Sorafenib in drug-resistant HCC cells co-overexpressing both IL7 and MAL2. Our studies suggest that targeting autophagy may be a novel strategy to overcome IL7/MAL2-mediated Sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Jeffrey C. To
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China; (J.C.T.); (X.-X.L.)
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Shan Gao
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China; (J.C.T.); (X.-X.L.)
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xiao-Xiao Li
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China; (J.C.T.); (X.-X.L.)
| | - Yanxiang Zhao
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China; (J.C.T.); (X.-X.L.)
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Vincent W. Keng
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China; (J.C.T.); (X.-X.L.)
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| |
Collapse
|
10
|
Sturmlechner I, Jain A, Mu Y, Weyand CM, Goronzy JJ. T cell fate decisions during memory cell generation with aging. Semin Immunol 2023; 69:101800. [PMID: 37494738 PMCID: PMC10528238 DOI: 10.1016/j.smim.2023.101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
The defense against infectious diseases, either through natural immunity or after vaccinations, relies on the generation and maintenance of protective T cell memory. Naïve T cells are at the center of memory T cell generation during primary responses. Upon activation, they undergo a complex, highly regulated differentiation process towards different functional states. Naïve T cells maintained into older age have undergone epigenetic adaptations that influence their fate decisions during differentiation. We review age-sensitive, molecular pathways and gene regulatory networks that bias naïve T cell differentiation towards effector cell generation at the expense of memory and Tfh cells. As a result, T cell differentiation in older adults is associated with release of bioactive waste products into the microenvironment, higher stress sensitivity as well as skewing towards pro-inflammatory signatures and shorter life spans. These maladaptations not only contribute to poor vaccine responses in older adults but also fuel a more inflammatory state.
Collapse
Affiliation(s)
- Ines Sturmlechner
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Abhinav Jain
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yunmei Mu
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Cornelia M Weyand
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Medicine, Division of Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Jörg J Goronzy
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Department of Medicine, Division of Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| |
Collapse
|
11
|
Linterman MA. Age-dependent changes in T follicular helper cells shape the humoral immune response to vaccination. Semin Immunol 2023; 69:101801. [PMID: 37379670 DOI: 10.1016/j.smim.2023.101801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Vaccination is an excellent strategy to limit the morbidity and mortality associated with infectious disease. Vaccination creates protective, long-lived antibody-mediated immunity by inducing the germinal centre response, an intricate immune reaction that produces memory B cells and long-lived antibody-secreting plasma cells that provide protection against (re)infection. The magnitude and quality of the germinal centre response declines with age, contributing to poor vaccine-induced immunity in older individuals. T follicular helper cells are essential for the formation and function of the germinal centre response. This review will discuss how age-dependent changes in T follicular helper cells influence the germinal centre response, and the evidence that age-dependent changes need not be a barrier to successful vaccination in the later years of life.
Collapse
Affiliation(s)
- Michelle A Linterman
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom.
| |
Collapse
|
12
|
Stagg J, Golden E, Wennerberg E, Demaria S. The interplay between the DNA damage response and ectonucleotidases modulates tumor response to therapy. Sci Immunol 2023; 8:eabq3015. [PMID: 37418547 PMCID: PMC10394739 DOI: 10.1126/sciimmunol.abq3015] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
The extracellular nucleoside adenosine reduces tissue inflammation and is generated by irreversible dephosphorylation of adenosine monophosphate (AMP) mediated by the ectonucleotidase CD73. The pro-inflammatory nucleotides adenosine triphosphate, nicotinamide adenine dinucleotide, and cyclic guanosine -monophosphate-AMP (cGAMP), which are produced in the tumor microenvironment (TME) during therapy-induced immunogenic cell death and activation of innate immune signaling, can be converted into AMP by ectonucleotidases CD39, CD38, and CD203a/ENPP1. Thus, ectonucleotidases shape the TME by converting immune-activating signals into an immunosuppressive one. Ectonucleotidases also hinder the ability of therapies including radiation therapy, which enhance the release of pro-inflammatory nucleotides in the extracellular milieu, to induce immune-mediated tumor rejection. Here, we review the immunosuppressive effects of adenosine and the role of different ectonucleotidases in modulating antitumor immune responses. We discuss emerging opportunities to target adenosine generation and/or its ability to signal via adenosine receptors expressed by immune and cancer cells in the context of combination immunotherapy and radiotherapy.
Collapse
Affiliation(s)
- John Stagg
- Centre de Recherche du Centre Hospitalier de
l’Université de Montréal, 900 St-Denis street, Montreal,
Quebec, Canada, H2X 0A9
| | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medicine,
New York, NY 10065, USA
| | - Erik Wennerberg
- Division of Radiotherapy and Imaging, Institute of Cancer
Research, London SM2 5NG, UK
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine,
New York, NY 10065, USA
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, 10065, USA
| |
Collapse
|
13
|
Guo L, Li X, Gould T, Wang ZY, Cao W. T cell aging and Alzheimer's disease. Front Immunol 2023; 14:1154699. [PMID: 37081887 PMCID: PMC10110977 DOI: 10.3389/fimmu.2023.1154699] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
The brain has long been considered an immune-privileged organ due to the presence of the blood-brain barrier (BBB). However, recent discoveries have revealed the underestimated role of T cells in the brain through the meningeal lymphatic system. Age is the primary risk factor for Alzheimer's disease (AD), resulting in marked age-dependent changes in T cells. Manipulating peripheral T cell immune response has been shown to impact AD, but the relationship between T cell aging and AD remains poorly understood. Given the limited success of targeting amyloid beta (Aβ) and the growing evidence of T cells' involvement in non-lymphoid organ aging, a deeper understanding of the relationship between T cells and AD in the context of aging is crucial for advancing therapeutic progress. In this review, we comprehensively examine existing studies on T cells and AD and offer an integrated perspective on their interconnections in the context of aging. This understanding can inform the development of new interventions to prevent or treat AD.
Collapse
Affiliation(s)
- Lin Guo
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, China
| | - Xiaoting Li
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, China
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | | | - Zhan-You Wang
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, China
| | - Wenqiang Cao
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Zhang H, Jadhav RR, Cao W, Goronzy IN, Zhao TV, Jin J, Ohtsuki S, Hu Z, Morales J, Greenleaf WJ, Weyand CM, Goronzy JJ. Aging-associated HELIOS deficiency in naive CD4 + T cells alters chromatin remodeling and promotes effector cell responses. Nat Immunol 2023; 24:96-109. [PMID: 36510022 PMCID: PMC10118794 DOI: 10.1038/s41590-022-01369-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 10/24/2022] [Indexed: 12/14/2022]
Abstract
Immune aging combines cellular defects in adaptive immunity with the activation of pathways causing a low-inflammatory state. Here we examined the influence of age on the kinetic changes in the epigenomic and transcriptional landscape induced by T cell receptor (TCR) stimulation in naive CD4+ T cells. Despite attenuated TCR signaling in older adults, TCR activation accelerated remodeling of the epigenome and induced transcription factor networks favoring effector cell differentiation. We identified increased phosphorylation of STAT5, at least in part due to aberrant IL-2 receptor and lower HELIOS expression, as upstream regulators. Human HELIOS-deficient, naive CD4+ T cells, when transferred into human-synovium-mouse chimeras, infiltrated tissues more efficiently. Inhibition of IL-2 or STAT5 activity in T cell responses of older adults restored the epigenetic response pattern to the one seen in young adults. In summary, reduced HELIOS expression in non-regulatory naive CD4+ T cells in older adults directs T cell fate decisions toward inflammatory effector cells that infiltrate tissue.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Rohit R Jadhav
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Wenqiang Cao
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Health Sciences Institute, China Medical University, Shenyang, China
| | - Isabel N Goronzy
- Biochemistry and Molecular Biophysics, California Institute of Technology, Pasadena, CA, USA
| | - Tuantuan V Zhao
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Jun Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Shozo Ohtsuki
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Zhaolan Hu
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Jose Morales
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | | | - Cornelia M Weyand
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Jörg J Goronzy
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
15
|
Fang F, Cao W, Mu Y, Okuyama H, Li L, Qiu J, Weyand CM, Goronzy JJ. IL-4 prevents adenosine-mediated immunoregulation by inhibiting CD39 expression. JCI Insight 2022; 7:e157509. [PMID: 35730568 PMCID: PMC9309057 DOI: 10.1172/jci.insight.157509] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
The ectonucleotidase CD39 functions as a checkpoint in purinergic signaling on effector T cells. By depleting eATP and initiating the generation of adenosine, it impairs memory cell development and contributes to T cell exhaustion, thereby causing defective tumor immunity and deficient T cell responses in older adults who have increased CD39 expression. Tuning enzymatic activity of CD39 and targeting the transcriptional regulation of ENTPD1 can be used to modulate purinergic signaling. Here, we describe that STAT6 phosphorylation downstream of IL-4 signaling represses CD39 expression on activated T cells by inducing a transcription factor network including GATA3, GFI1, and YY1. GATA3 suppresses ENTPD1 transcription through prevention of RUNX3 recruitment to the ENTPD1 promoter. Conversely, pharmacological STAT6 inhibition decreases T cell effector functions via increased CD39 expression, resulting in the defective signaling of P2X receptors by ATP and stimulation of A2A receptors by adenosine. Our studies suggest that inhibiting the STAT6 pathway to increase CD39 expression has the potential to treat autoimmune disease while stimulation of the pathway could improve T cell immunity.
Collapse
Affiliation(s)
- Fengqin Fang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, California, USA
- Department of Laboratory Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqiang Cao
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, California, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Health Sciences Institute, China Medical University, Shenyang, China
| | - Yunmei Mu
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Hirohisa Okuyama
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Lingjie Li
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Reproductive Medicine, Shanghai, China
| | - Jingtao Qiu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California, USA
| | - Cornelia M. Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, California, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Department of Medicine/Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Jörg J. Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, California, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Department of Medicine/Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Zhao TV, Sato Y, Goronzy JJ, Weyand CM. T-Cell Aging-Associated Phenotypes in Autoimmune Disease. FRONTIERS IN AGING 2022; 3:867950. [PMID: 35821833 PMCID: PMC9261367 DOI: 10.3389/fragi.2022.867950] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/28/2022] [Indexed: 01/10/2023]
Abstract
The aging process causes profound restructuring of the host immune system, typically associated with declining host protection against cancer and infection. In the case of T cells, aging leads to the accumulation of a diverse set of T-cell aging-associated phenotypes (TASP), some of which have been implicated in driving tissue inflammation in autoimmune diseases. T cell aging as a risk determinant for autoimmunity is exemplified in two classical autoimmune conditions: rheumatoid arthritis (RA), a disease predominantly affecting postmenopausal women, and giant cell arteritis (GCA), an inflammatory vasculopathy exclusively occurring during the 6th-9th decade of life. Pathogenic T cells in RA emerge as a consequence of premature immune aging. They have shortening and fragility of telomeric DNA ends and instability of mitochondrial DNA. As a result, they produce a distinct profile of metabolites, disproportionally expand their endoplasmic reticulum (ER) membranes and release excess amounts of pro-inflammatory effector cytokines. Characteristically, they are tissue invasive, activate the inflammasome and die a pyroptotic death. Patients with GCA expand pathogenic CD4+ T cells due to aberrant expression of the co-stimulatory receptor NOTCH1 and the failure of the PD-1/PD-L1 immune checkpoint. In addition, GCA patients lose anti-inflammatory Treg cells, promoting tissue-destructive granulomatous vasculitis. In summary, emerging data identify T cell aging as a risk factor for autoimmune disease and directly link TASPs to the breakdown of T cell tolerance and T-cell-induced tissue inflammation.
Collapse
Affiliation(s)
- Tuantuan V. Zhao
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
| | - Yuki Sato
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
| | - Jorg J. Goronzy
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| | - Cornelia M. Weyand
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
17
|
Zhang H, Weyand CM, Goronzy JJ. Hallmarks of the aging T-cell system. FEBS J 2021; 288:7123-7142. [PMID: 33590946 PMCID: PMC8364928 DOI: 10.1111/febs.15770] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/24/2021] [Accepted: 02/15/2021] [Indexed: 12/21/2022]
Abstract
The adaptive immune system has the enormous challenge to protect the host through the generation and differentiation of pathogen-specific short-lived effector T cells while in parallel developing long-lived memory cells to control future encounters with the same pathogen. A complex regulatory network is needed to preserve a population of naïve cells over lifetime that exhibit sufficient diversity of antigen receptors to respond to new antigens, while also sustaining immune memory. In parallel, cells need to maintain their proliferative potential and the plasticity to differentiate into different functional lineages. Initial signs of waning immune competence emerge after 50 years of age, with increasing clinical relevance in the 7th-10th decade of life. Morbidity and mortality from infections increase, as drastically exemplified by the current COVID-19 pandemic. Many vaccines, such as for the influenza virus, are poorly effective to generate protective immunity in older individuals. Age-associated changes occur at the level of the T-cell population as well as the functionality of its cellular constituents. The system highly relies on the self-renewal of naïve and memory T cells, which is robust but eventually fails. Genetic and epigenetic modifications contribute to functional differences in responsiveness and differentiation potential. To some extent, these changes arise from defective maintenance; to some, they represent successful, but not universally beneficial adaptations to the aging host. Interventions that can compensate for the age-related defects and improve immune responses in older adults are increasingly within reach.
Collapse
Affiliation(s)
- Huimin Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Cornelia M. Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Jörg J. Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| |
Collapse
|
18
|
Fang F, Cao W, Zhu W, Lam N, Li L, Gaddam S, Wang Y, Kim C, Lambert S, Zhang H, Hu B, Farber DL, Weyand CM, Goronzy JJ. The cell-surface 5'-nucleotidase CD73 defines a functional T memory cell subset that declines with age. Cell Rep 2021; 37:109981. [PMID: 34758299 PMCID: PMC8612175 DOI: 10.1016/j.celrep.2021.109981] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/09/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Memory T cells exhibit considerable diversity that determines their ability to be protective. Here, we examine whether changes in T cell heterogeneity contribute to the age-associated failure of immune memory. By screening for age-dependent T cell-surface markers, we identify CD4 and CD8 memory T cell subsets that are unrelated to previously defined subsets of central and effector memory cells. Memory T cells expressing the ecto-5'-nucleotidase CD73 constitute a functionally distinct subset of memory T cells that declines with age. They resemble long-lived, polyfunctional memory cells but are also poised to display effector functions and to develop into cells resembling tissue-resident memory T cells (TRMs). Upstream regulators of differential chromatin accessibility and transcriptomes include transcription factors that facilitate CD73 expression and regulate TRM differentiation. CD73 is not just a surrogate marker of these regulatory networks but is directly involved in T cell survival.
Collapse
Affiliation(s)
- Fengqin Fang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Wenqiang Cao
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, US
| | - Weikang Zhu
- CEMS, NCMIS, HCMS, MDIS, Academy of Mathematics & Systems Science, Chinese Academy of Sciences, Beijing 100190, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lingjie Li
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China
| | - Sadhana Gaddam
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yong Wang
- CEMS, NCMIS, HCMS, MDIS, Academy of Mathematics & Systems Science, Chinese Academy of Sciences, Beijing 100190, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Chulwoo Kim
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Simon Lambert
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Huimin Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, US
| | - Bin Hu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, US
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, US.
| |
Collapse
|
19
|
CD39 Regulation and Functions in T Cells. Int J Mol Sci 2021; 22:ijms22158068. [PMID: 34360833 PMCID: PMC8348030 DOI: 10.3390/ijms22158068] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
CD39 is an enzyme which is responsible, together with CD73, for a cascade converting adenosine triphosphate into adenosine diphosphate and cyclic adenosine monophosphate, ultimately leading to the release of an immunosuppressive form of adenosine in the tumor microenvironment. Here, we first review the environmental and genetic factors shaping CD39 expression. Second, we report CD39 functions in the T cell compartment, highlighting its role in regulatory T cells, conventional CD4+ T cells and CD8+ T cells. Finally, we compile a list of studies, from preclinical models to clinical trials, which have made essential contributions to the discovery of novel combinatorial approaches in the treatment of cancer.
Collapse
|
20
|
O’Connor RA, Chauhan V, Mathieson L, Titmarsh H, Koppensteiner L, Young I, Tagliavini G, Dorward DA, Prost S, Dhaliwal K, Wallace WA, Akram AR. T cells drive negative feedback mechanisms in cancer associated fibroblasts, promoting expression of co-inhibitory ligands, CD73 and IL-27 in non-small cell lung cancer. Oncoimmunology 2021; 10:1940675. [PMID: 34290905 PMCID: PMC8274440 DOI: 10.1080/2162402x.2021.1940675] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/07/2021] [Indexed: 11/10/2022] Open
Abstract
The success of immune checkpoint therapy shows tumor-reactive T cells can eliminate cancer cells but are restrained by immunosuppression within the tumor micro-environment (TME). Cancer associated fibroblasts (CAFs) are the dominant stromal cell in the TME and co-localize with T cells in non-small cell lung cancer. We demonstrate the bidirectional nature of CAF/T cell interactions; T cells promote expression of co-inhibitory ligands, MHC molecules and CD73 on CAFs, increasing their production of IL-6 and eliciting production of IL-27. In turn CAFs upregulate co-inhibitory receptors on T cells including the ectonucleotidase CD39 promoting development of an exhausted but highly cytotoxic phenotype. Our results highlight the bidirectional interaction between T cells and CAFs in promoting components of the immunosuppressive CD39, CD73 adenosine pathway and demonstrate IL-27 production can be induced in CAF by activated T cells.
Collapse
Affiliation(s)
- Richard A O’Connor
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Vishwani Chauhan
- Edinburgh Medical School, The Chancellor’s Building, University of Edinburgh, Edinburgh, UK
| | - Layla Mathieson
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Helen Titmarsh
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lilian Koppensteiner
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Irene Young
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Guilia Tagliavini
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David A Dorward
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Sandrine Prost
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Pathology, The Chancellor’s Building, University of Edinburgh, Edinburgh, UK
| | - Kevin Dhaliwal
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - William A Wallace
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, UK
- Department of Pathology, The Chancellor’s Building, University of Edinburgh, Edinburgh, UK
| | - Ahsan R Akram
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
21
|
Jin J, Kim C, Xia Q, Gould TM, Cao W, Zhang H, Li X, Weiskopf D, Grifoni A, Sette A, Weyand CM, Goronzy JJ. Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals. Sci Immunol 2021; 6:eabg0791. [PMID: 34145066 PMCID: PMC8422387 DOI: 10.1126/sciimmunol.abg0791] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.
Collapse
Affiliation(s)
- Jun Jin
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Chulwoo Kim
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Qiong Xia
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Timothy M Gould
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Wenqiang Cao
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Huimin Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Xuanying Li
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Jorg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| |
Collapse
|
22
|
Kim C, Ye Z, Weyand CM, Goronzy JJ. miR-181a-regulated pathways in T-cell differentiation and aging. Immun Ageing 2021; 18:28. [PMID: 34130717 PMCID: PMC8203492 DOI: 10.1186/s12979-021-00240-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are regulatory noncoding RNAs important for many aspects of cellular processes including cell differentiation and proliferation. Functions of numerous miRNAs have been identified in T cells, with miR-181a regulating T cell activation thresholds during thymic T cell development and during activation of peripheral T cells. Intriguingly, miR-181a is implicated in defective antiviral and vaccine responses in older individuals, as its expression declines in naïve T cells with increasing age. Here, we review the pathways that are regulated by miR-181a and that explain the unique role of miR-181a in T cell development, T cell activation and antiviral T cell responses. These studies provide a framework for understanding how a decline in miR-181a expression in T cells could contribute to age-related defects in adaptive immunity. We furthermore review the mechanisms that cause the age-related decline in miR-181a expression and discuss the potential of restoring miR-181a expression or targeting miR-181a-regulated pathways to improve impaired T cell responses in older individuals.
Collapse
Affiliation(s)
- Chulwoo Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Zhongde Ye
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, USA.
| |
Collapse
|
23
|
Jin J, Zhang H, Weyand CM, Goronzy JJ. Lysosomes in T Cell Immunity and Aging. FRONTIERS IN AGING 2021; 2:809539. [PMID: 35822050 PMCID: PMC9261317 DOI: 10.3389/fragi.2021.809539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 05/01/2023]
Abstract
Lysosomes were initially recognized as degradation centers that regulate digestion and recycling of cellular waste. More recent studies document that the lysosome is an important signaling hub that regulates cell metabolism. Our knowledge of the role of lysosomes in immunity is mostly derived from innate immune cells, especially lysosomal degradation-specialized cells such as macrophages and dendritic cells. Their function in adaptive immunity is less understood. However, with the recent emphasis on metabolic regulation of T cell differentiation, lysosomes are entering center stage in T cell immunology. In this review, we will focus on the role of lysosomes in adaptive immunity and discuss recent findings on lysosomal regulation of T cell immune responses and lysosomal dysfunction in T cell aging.
Collapse
Affiliation(s)
- Jun Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Huimin Zhang
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Cornelia M. Weyand
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Jorg J. Goronzy
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine/Rheumatology, Mayo Clinic, Rochester, MN, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Jorg J. Goronzy,
| |
Collapse
|
24
|
The Transcription Factor TCF1 in T Cell Differentiation and Aging. Int J Mol Sci 2020; 21:ijms21186497. [PMID: 32899486 PMCID: PMC7554785 DOI: 10.3390/ijms21186497] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
The transcription factor T cell factor 1 (TCF1), a pioneer transcription factor as well as a downstream effector of WNT/β-catenin signaling, is indispensable for T cell development in the thymus. Recent studies have highlighted the additional critical role of TCF1 in peripheral T cell responses to acute and chronic infections as well as cancer. Here, we review the regulatory functions of TCF1 in the differentiation of T follicular helper cells, memory T cells and recently described stem-like exhausted T cells, where TCF1 promotes less differentiated stem-like cell states by controlling common gene-regulatory networks. These studies also provide insights into the mechanisms of defective T cell responses in older individuals. We discuss alterations in TCF1 expression and related regulatory networks with age and their consequences for T cell responses to infections and vaccination. The increasing understanding of the pathways regulating TCF1 expression and function in aged T cells holds the promise of enabling the design of therapeutic interventions aiming at improving T cell responses in older individuals.
Collapse
|
25
|
Gustafson CE, Kim C, Weyand CM, Goronzy JJ. Influence of immune aging on vaccine responses. J Allergy Clin Immunol 2020; 145:1309-1321. [PMID: 32386655 PMCID: PMC7198995 DOI: 10.1016/j.jaci.2020.03.017] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Impaired vaccine responses in older individuals are associated with alterations in both the quantity and quality of the T-cell compartment with age. As reviewed herein, the T-cell response to vaccination requires a fine balance between the generation of inflammatory effector T cells versus follicular helper T (TFH) cells that mediate high-affinity antibody production in tandem with the induction of long-lived memory cells for effective recall immunity. During aging, we find that this balance is tipped where T cells favor short-lived effector but not memory or TFH responses. Consistently, vaccine-induced antibodies commonly display a lower protective capacity. Mechanistically, multiple, potentially targetable, changes in T cells have been identified that contribute to these age-related defects, including posttranscription regulation, T-cell receptor signaling, and metabolic function. Although research into the induction of tissue-specific immunity by vaccines and with age is still limited, current mechanistic insights provide a framework for improved design of age-specific vaccination strategies that require further evaluation in a clinical setting.
Collapse
Affiliation(s)
- Claire E Gustafson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif
| | - Chulwoo Kim
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif.
| |
Collapse
|