1
|
Kraft AE, Bork NI, Subramanian H, Pavlaki N, Failla AV, Zobiak B, Conti M, Nikolaev VO. Phosphodiesterases 4B and 4D Differentially Regulate cAMP Signaling in Calcium Handling Microdomains of Mouse Hearts. Cells 2024; 13:476. [PMID: 38534320 DOI: 10.3390/cells13060476] [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: 11/23/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
The ubiquitous second messenger 3',5'-cyclic adenosine monophosphate (cAMP) regulates cardiac excitation-contraction coupling (ECC) by signaling in discrete subcellular microdomains. Phosphodiesterase subfamilies 4B and 4D are critically involved in the regulation of cAMP signaling in mammalian cardiomyocytes. Alterations of PDE4 activity in human hearts has been shown to result in arrhythmias and heart failure. Here, we sought to systematically investigate specific roles of PDE4B and PDE4D in the regulation of cAMP dynamics in three distinct subcellular microdomains, one of them located at the caveolin-rich plasma membrane which harbors the L-type calcium channels (LTCCs), as well as at two sarco/endoplasmic reticulum (SR) microdomains centered around SR Ca2+-ATPase (SERCA2a) and cardiac ryanodine receptor type 2 (RyR2). Transgenic mice expressing Förster Resonance Energy Transfer (FRET)-based cAMP-specific biosensors targeted to caveolin-rich plasma membrane, SERCA2a and RyR2 microdomains were crossed to PDE4B-KO and PDE4D-KO mice. Direct analysis of the specific effects of both PDE4 subfamilies on local cAMP dynamics was performed using FRET imaging. Our data demonstrate that all three microdomains are differentially regulated by these PDE4 subfamilies. Whereas both are involved in cAMP regulation at the caveolin-rich plasma membrane, there are clearly two distinct cAMP microdomains at the SR formed around RyR2 and SERCA2a, which are preferentially controlled by PDE4B and PDE4D, respectively. This correlates with local cAMP-dependent protein kinase (PKA) substrate phosphorylation and arrhythmia susceptibility. Immunoprecipitation assays confirmed that PDE4B is associated with RyR2 along with PDE4D. Stimulated Emission Depletion (STED) microscopy of immunostained cardiomyocytes suggested possible co-localization of PDE4B with both sarcolemmal and RyR2 microdomains. In conclusion, our functional approach could show that both PDE4B and PDE4D can differentially regulate cardiac cAMP microdomains associated with calcium homeostasis. PDE4B controls cAMP dynamics in both caveolin-rich plasma membrane and RyR2 vicinity. Interestingly, PDE4B is the major regulator of the RyR2 microdomain, as opposed to SERCA2a vicinity, which is predominantly under PDE4D control, suggesting a more complex regulatory pattern than previously thought, with multiple PDEs acting at the same location.
Collapse
Affiliation(s)
- Axel E Kraft
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Nadja I Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Hariharan Subramanian
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Nikoleta Pavlaki
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Antonio V Failla
- UKE Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bernd Zobiak
- UKE Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marco Conti
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
2
|
Goldmann O, Nwofor OV, Chen Q, Medina E. Mechanisms underlying immunosuppression by regulatory cells. Front Immunol 2024; 15:1328193. [PMID: 38380317 PMCID: PMC10876998 DOI: 10.3389/fimmu.2024.1328193] [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: 10/26/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024] Open
Abstract
Regulatory cells, such as regulatory T cells (Tregs), regulatory B cells (Bregs), and myeloid-derived suppressor cells (MDSCs), play a crucial role in preserving immune tolerance and controlling immune responses during infections to prevent excessive immune activation. However, pathogens have developed strategies to hijack these regulatory cells to decrease the overall effectiveness of the immune response and persist within the host. Consequently, therapeutic targeting of these immunosuppressive mechanisms during infection can reinvigorate the immune response and improve the infection outcome. The suppressive mechanisms of regulatory cells are not only numerous but also redundant, reflecting the complexity of the regulatory network in modulating the immune responses. The context of the immune response, such as the type of pathogen or tissue involved, further influences the regulatory mechanisms involved. Examples of these immunosuppressive mechanisms include the production of inhibitory cytokines such as interleukin 10 (IL-10) and transforming growth factor beta (TGF-β) that inhibit the production of pro-inflammatory cytokines and dampen the activation and proliferation of effector T cells. In addition, regulatory cells utilize inhibitory receptors like cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) to engage with their respective effector cells, thereby suppressing their function. An alternative approach involves the modulation of metabolic reprogramming in effector immune cells to limit their activation and proliferation. In this review, we provide an overview of the major mechanisms mediating the immunosuppressive effect of the different regulatory cell subsets in the context of infection.
Collapse
Affiliation(s)
| | | | | | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
3
|
Zhang H, Liu Y, Liu J, Chen J, Wang J, Hua H, Jiang Y. cAMP-PKA/EPAC signaling and cancer: the interplay in tumor microenvironment. J Hematol Oncol 2024; 17:5. [PMID: 38233872 PMCID: PMC10792844 DOI: 10.1186/s13045-024-01524-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex disease resulting from abnormal cell growth that is induced by a number of genetic and environmental factors. The tumor microenvironment (TME), which involves extracellular matrix, cancer-associated fibroblasts (CAF), tumor-infiltrating immune cells and angiogenesis, plays a critical role in tumor progression. Cyclic adenosine monophosphate (cAMP) is a second messenger that has pleiotropic effects on the TME. The downstream effectors of cAMP include cAMP-dependent protein kinase (PKA), exchange protein activated by cAMP (EPAC) and ion channels. While cAMP can activate PKA or EPAC and promote cancer cell growth, it can also inhibit cell proliferation and survival in context- and cancer type-dependent manner. Tumor-associated stromal cells, such as CAF and immune cells, can release cytokines and growth factors that either stimulate or inhibit cAMP production within the TME. Recent studies have shown that targeting cAMP signaling in the TME has therapeutic benefits in cancer. Small-molecule agents that inhibit adenylate cyclase and PKA have been shown to inhibit tumor growth. In addition, cAMP-elevating agents, such as forskolin, can not only induce cancer cell death, but also directly inhibit cell proliferation in some cancer types. In this review, we summarize current understanding of cAMP signaling in cancer biology and immunology and discuss the basis for its context-dependent dual role in oncogenesis. Understanding the precise mechanisms by which cAMP and the TME interact in cancer will be critical for the development of effective therapies. Future studies aimed at investigating the cAMP-cancer axis and its regulation in the TME may provide new insights into the underlying mechanisms of tumorigenesis and lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Hongying Zhang
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongliang Liu
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jieya Liu
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinzhu Chen
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yangfu Jiang
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Kawakami R, Sakaguchi S. Regulatory T Cells for Control of Autoimmunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:67-82. [PMID: 38467973 DOI: 10.1007/978-981-99-9781-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Regulatory T (Treg) cells, which specifically express the master transcription factor FoxP3, are indispensable for the maintenance of immunological self-tolerance and homeostasis. Their functional or numerical anomalies can be causative of autoimmune and other inflammatory diseases. Recent advances in the research of the cellular and molecular basis of how Treg cells develop, exert suppression, and maintain their function have enabled devising various ways for controlling physiological and pathological immune responses by targeting Treg cells. It is now envisaged that Treg cells as a "living drug" are able to achieve antigen-specific immune suppression of various immune responses and reestablish immunological self-tolerance in the clinic.
Collapse
Affiliation(s)
- Ryoji Kawakami
- Kyoto University, Kyoto, Japan
- Osaka University, Osaka, Japan
| | | |
Collapse
|
5
|
Teixeira HMP, Cruz ÁA, Jesus TS, de Santana MBR, Jesus MS, Tugores R, Araujo WS, Reis RCC, Pinheiro GP, Figueiredo CA, Costa RS. The rs2601796 variant in ADCY9 gene is associated with severe asthma and less bronchodilator response. Gene 2023; 886:147714. [PMID: 37579959 DOI: 10.1016/j.gene.2023.147714] [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: 06/17/2023] [Revised: 07/29/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Asthma is a respiratory disease caused by the interaction of genetic and environmental factors. The adenylyl cyclase type 9 (ADCY9) enzyme produces the cyclic-adenosinemonophosphate (cAMP), important mediator involved in bronchodilation and immunomodulatory response. The aim of this study was to investigate if rs2601796 and rs2532019 variants in the ADCY9 gene are associated with asthma and lung function. The study comprised 1,052 subjects. Logistic regressions were done using PLINK 1.9 adjusted by sex, age, BMI, smoke and principal components. Bronchodilator responsiveness was assessed using the percentage of difference in FEV1 before and after the bronchodilator use. The in silico analysis for gene expression was performed in the GTEx Portal. The variant rs2601796 (AA/AG genotype) was positively associated with asthma severity (OR: 1.60 IC95%: 1.08-2.39) and with obstruction in individuals with severe asthma (OR: 3.10, IC95%: 1.11-8.62). Individuals with severe asthma and the AA/AG genotype of rs2601796 had less responsiveness to bronchodilators and also a lower expression of ADCY9 in lung and whole blood. The variant rs2532019 (TT/GT genotype) also downregulated the ADCY9 gene expression, but no significant association with the studied phenotypes was found. Thus, the variant in ADCY9 was associated with worse asthma outcomes, including a lower response to bronchodilators, likely due to the impact on its gene expression rate. This variant may be useful in the future to assist in personalized management of patients with asthma.
Collapse
Affiliation(s)
- Helena M P Teixeira
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | - Álvaro A Cruz
- Fundação ProAR e, Universidade Federal da Bahia (UFBA), Bahia, Brazil
| | - Talita S Jesus
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | | | - Marinalva S Jesus
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | - Rafaela Tugores
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | | | | | | | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | - Ryan S Costa
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil.
| |
Collapse
|
6
|
Silva HDS, Teixeira HMP, Gomes LGDS, Cruz ÁA, Alcantara-Neves NM, Barreto M, Figueiredo CA, Costa RDS. PDE4D gene variants and haplotypes are associated with asthma and atopy in Brazilian children. Immunobiology 2023; 228:152724. [PMID: 37549468 DOI: 10.1016/j.imbio.2023.152724] [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: 01/30/2023] [Revised: 07/20/2023] [Accepted: 07/30/2023] [Indexed: 08/09/2023]
Abstract
PDE4D (Phosphodiesterase 4D) gene encodes a hydrolase of cyclic AMP. PDE4D genetic variants have been associated with asthma susceptibility. Therefore, this study aimed to investigate the association between PDE4D variants (and haplotypes) with asthma and atopy in a Brazilian population. The study comprised 1,246 unrelated participants from the SCAALA (Social Changes Asthma and Allergy in Latin America) program. Genotyping was performed using the Illumina 2.5 Human Omni bead chip. Multivariate logistic regression was used to investigate the association between PDE4D variants and asthma/atopy phenotypes in PLINK 1.09 software. Twenty-four SNVs in PDE4D were associated with atopy or asthma. The rs6898082 (A) variant increased asthma susceptibility (OR 2.76; CI 99% 1.26-6.03) and was also related to a greater PDE4D expression in the GTEx database. Also, the variant rs6870632 was further associated with asthma in meta-analysis with a replication cohort. In addition, the variants rs75699812 (C), rs8007656 (G), and rs958851 (T) were positively associated with atopy. Moreover, these variants formed an atopy risk haplotype (OR 1.82; CI 99% 1.15-2.88). Also, these variants were related to lower levels of IL-10. Functional in silico assessment showed that some PDE4D SNVs may have an impact on gene regulation and expression. Variants in the PDE4D are positively associated with asthma and allergy markers. It is possible that these variants lead to alteration in PDE4D expression and therefore impact immunity and pulmonary function.
Collapse
Affiliation(s)
| | | | | | | | | | - Maurício Barreto
- Centro de Integração de Dados e Conhecimento para Saúde (CIDACS), Fiocruz, Salvador, Bahia, Brazil; Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Ryan Dos Santos Costa
- Instituto de Ciências da Saúde, Universidade Federal da Bahia (UFBA), Salvador, Brazil.
| |
Collapse
|
7
|
You J, Reilly MD, Eljalby M, Bareja R, Yusupova M, Vyas NS, Bang J, Ding W, Desman G, Miller LS, Elemento O, Granstein RD, Zippin JH. Soluble adenylyl cyclase contributes to imiquimod-mediated inflammation and is a potential therapeutic target in psoriasis. Exp Dermatol 2023; 32:1051-1062. [PMID: 37039485 PMCID: PMC10523866 DOI: 10.1111/exd.14811] [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: 11/09/2022] [Revised: 03/21/2023] [Accepted: 04/02/2023] [Indexed: 04/12/2023]
Abstract
Cyclic AMP (cAMP) has a key role in psoriasis pathogenesis, as indicated by the therapeutic efficacy of phosphodiesterase inhibitors that prevent the degradation of cAMP. However, whether soluble adenylate cyclase (sAC) (encoded by the ADCY10 gene), which is an important source for cAMP, is involved in Th17 cell-mediated inflammation or could be an alternative therapeutic target in psoriasis is unknown. We have utilized the imiquimod model of murine psoriasiform dermatitis to address this question. Adcy10-/- mice had reduced erythema, scaling and swelling in the skin and reduced CD4+ IL17+ cell numbers in the draining lymph nodes, compared with wild-type mice after induction of psoriasiform dermatitis with imiquimod. Keratinocyte-specific knock out of Adcy10 had no effect on imiquimod-induced ear swelling suggesting keratinocyte sAC has no role in imiquimod-induced inflammation. During Th17 polarization in vitro, naive T cells from Adcy10-/- mice exhibited reduced IL17 secretion and IL-17+ T-cell proliferation suggesting that differentiation into Th17 cells is suppressed without sAC activity. Interestingly, loss of sAC did not impact the expression of Th17 lineage-defining transcription factors (such as Rorc and cMaf) but rather was required for CREB-dependent gene expression, which is known to support Th17 cell gene expression. Finally, topical application of small molecule sAC inhibitors (sACi) reduced imiquimod-induced psoriasiform dermatitis and Il17 gene expression in the skin. Collectively, these findings demonstrate that sAC is important for psoriasiform dermatitis in mouse skin. sACi may provide an alternative class of topical therapeutics for Th17-mediated skin diseases.
Collapse
Affiliation(s)
- Jaewon You
- Department of Dermatology, Weill Cornell Medicine, NY NY
| | | | | | - Rohan Bareja
- Englander Institute of Precision Medicine, Weill Cornell Medicine, NY NY
| | | | - Nikki S. Vyas
- Departments of Pathology and Dermatology, Icahn School of Medicine at Mount Sinai, NY NY
| | - Jakyung Bang
- Department of Dermatology, Weill Cornell Medicine, NY NY
| | - Wanhong Ding
- Department of Dermatology, Weill Cornell Medicine, NY NY
| | - Garrett Desman
- Departments of Pathology and Dermatology, Icahn School of Medicine at Mount Sinai, NY NY
- ProHEALTH Care Associates, OptumCare, New Hyde Park, NY
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD
- Immunology, Janssen Research and Development, Spring House, PA
| | - Olivier Elemento
- Englander Institute of Precision Medicine, Weill Cornell Medicine, NY NY
| | | | - Jonathan H. Zippin
- Department of Dermatology, Weill Cornell Medicine, NY NY
- Englander Institute of Precision Medicine, Weill Cornell Medicine, NY NY
- Department of Pharmacology, Weill Cornell Medicine, NY NY
| |
Collapse
|
8
|
Role of Protein Kinase A Activation in the Immune System with an Emphasis on Lipopolysaccharide-Responsive and Beige-like Anchor Protein in B Cells. Int J Mol Sci 2023; 24:ijms24043098. [PMID: 36834508 PMCID: PMC9962394 DOI: 10.3390/ijms24043098] [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: 11/12/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Cyclic AMP-dependent protein kinase A (PKA) is a ubiquitous enzymatic complex that is involved in a broad spectrum of intracellular receptor signaling. The activity of PKA depends on A-kinase anchoring proteins (AKAPs) that attach to PKAs close to their substrates to control signaling. Although the relevance of PKA-AKAP signaling in the immune system is evident in T cells, its relevance in B and other immune cells remains relatively unclear. In the last decade, lipopolysaccharide-responsive and beige-like anchor protein (LRBA) has emerged as an AKAP that is ubiquitously expressed in B and T cells, specifically after activation. A deficiency of LRBA leads to immune dysregulation and immunodeficiency. The cellular mechanisms regulated by LRBA have not yet been investigated. Therefore, this review summarizes the functions of PKA in immunity and provides the most recent information regarding LRBA deficiency to deepen our understanding of immune regulation and immunological diseases.
Collapse
|
9
|
Krieg PF, Sonner JK, Kurelic R, Engler JB, Scharenberg MF, Bauer S, Nikolaev VO, Friese MA. GPR52 regulates cAMP in T cells but is dispensable for encephalitogenic responses. Front Immunol 2023; 13:1113348. [PMID: 36761164 PMCID: PMC9902724 DOI: 10.3389/fimmu.2022.1113348] [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: 12/01/2022] [Accepted: 12/31/2022] [Indexed: 01/25/2023] Open
Abstract
G-protein coupled receptors (GPCR) regulate 3',5'-cyclic adenosine monophosphate (cAMP) levels in T cells. cAMP as ubiquitous second messenger is crucial for adequate physiology of T cells by mediating effector T cell (Teff) function as well as regulatory T cell (Treg)-mediated immunosuppression. Several GPCRs have been identified to be crucial for Teff and Treg function. However, the role of the orphan, constitutively active Gs-coupled GPCR GPR52 is unknown. Here we show that GPR52 regulates cAMP levels in T cells but does not affect T cell function. We found that stimulation of transfected HEK cells or primary T cells with a GPR52 agonist results in a rise of intracellular cAMP. However, neither Gpr52 deficiency nor pharmacological modulation of GPR52 by antagonists or agonists affected T cell activation, differentiation, and proliferation or Treg-mediated immunosuppression. Moreover, Gpr52 deletion did not modify the clinical disease course of experimental autoimmune encephalomyelitis (EAE). Our results demonstrate that a modulation of cAMP levels in T cells does not inevitably result in altered T cell function. While we could not identify an obvious role of GPR52 in in vitro T cell assays and in vivo CNS autoimmunity, it might regulate T cell function in a different context or affect the function of other GPR52-expressing cells.
Collapse
Affiliation(s)
- Paula F. Krieg
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K. Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roberta Kurelic
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marlena F. Scharenberg
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Manuel A. Friese,
| |
Collapse
|
10
|
Schlöder J, Shahneh F, Schneider FJ, Wieschendorf B. Boosting regulatory T cell function for the treatment of autoimmune diseases – That’s only half the battle! Front Immunol 2022; 13:973813. [PMID: 36032121 PMCID: PMC9400058 DOI: 10.3389/fimmu.2022.973813] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 01/04/2023] Open
Abstract
Regulatory T cells (Treg) represent a subset of specialized T cells that are essential for the regulation of immune responses and maintenance of peripheral tolerance. Once activated, Treg exert powerful immunosuppressive properties, for example by inhibiting T cell-mediated immune responses against self-antigens, thereby protecting our body from autoimmunity. Autoimmune diseases such as multiple sclerosis, rheumatoid arthritis or systemic lupus erythematosus, exhibit an immunological imbalance mainly characterized by a reduced frequency and impaired function of Treg. In addition, there has been increasing evidence that – besides Treg dysfunction – immunoregulatory mechanisms fail to control autoreactive T cells due to a reduced responsiveness of T effector cells (Teff) for the suppressive properties of Treg, a process termed Treg resistance. In order to efficiently treat autoimmune diseases and thus fully induce immunological tolerance, a combined therapy aimed at both enhancing Treg function and restoring Teff responsiveness could most likely be beneficial. This review provides an overview of immunomodulating drugs that are currently used to treat various autoimmune diseases in the clinic and have been shown to increase Treg frequency as well as Teff sensitivity to Treg-mediated suppression. Furthermore, we discuss strategies on how to boost Treg activity and function, and their potential use in the treatment of autoimmunity. Finally, we present a humanized mouse model for the preclinical testing of Treg-activating substances in vivo.
Collapse
Affiliation(s)
- Janine Schlöder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- *Correspondence: Janine Schlöder,
| | - Fatemeh Shahneh
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Franz-Joseph Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Björn Wieschendorf
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
11
|
Imenez Silva PH, Câmara NO, Wagner CA. Role of proton-activated G protein-coupled receptors in pathophysiology. Am J Physiol Cell Physiol 2022; 323:C400-C414. [PMID: 35759438 DOI: 10.1152/ajpcell.00114.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Local acidification is a common feature of many disease processes such as inflammation, infarction, or solid tumor growth. Acidic pH is not merely a sequelae of disease but contributes to recruitment and regulation of immune cells, modifies metabolism of parenchymal, immune and tumor cells, modulates fibrosis, vascular permeability, oxygen availability and consumption, invasiveness of tumor cells, and impacts on cell survival. Thus, multiple pH-sensing mechanisms must exist in cells involved in these processes. These pH-sensors play important roles in normal physiology and pathophysiology, and hence might be attractive targets for pharmacological interventions. Among the pH-sensing mechanisms, OGR1 (GPR68), GPR4 (GPR4), and TDAG8 (GPR65) have emerged as important molecules. These G protein-coupled receptors are widely expressed, are upregulated in inflammation and tumors, sense changes in extracellular pH in the range between pH 8 and 6, and are involved in modulating key processes in inflammation, tumor biology, and fibrosis. This review discusses key features of these receptors and highlights important disease states and pathways affected by their activity.
Collapse
Affiliation(s)
- Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research NCCR Kidney.CH, Switzerland
| | - Niels Olsen Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research NCCR Kidney.CH, Switzerland
| |
Collapse
|
12
|
Chinn AM, Salmerón C, Lee J, Sriram K, Raz E, Insel PA. PDE4B Is a Homeostatic Regulator of Cyclic AMP in Dendritic Cells. Front Pharmacol 2022; 13:833832. [PMID: 35387344 PMCID: PMC8977838 DOI: 10.3389/fphar.2022.833832] [Citation(s) in RCA: 3] [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: 12/12/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic decreases in the second messenger cyclic AMP (cAMP) occur in numerous settings, but how cells compensate for such decreases is unknown. We have used a unique system-murine dendritic cells (DCs) with a DC-selective depletion of the heterotrimeric GTP binding protein Gαs-to address this issue. These mice spontaneously develop Th2-allergic asthma and their DCs have persistently lower cAMP levels. We found that phosphodiesterase 4B (PDE4B) is the primary phosphodiesterase expressed in DCs and that its expression is preferentially decreased in Gαs-depleted DCs. PDE4B expression is dynamic, falling and rising in a protein kinase A-dependent manner with decreased and increased cAMP concentrations, respectively. Treatment of DCs that drive enhanced Th2 immunity with a PDE4B inhibitor ameliorated DC-induced helper T cell response. We conclude that PDE4B is a homeostatic regulator of cellular cAMP concentrations in DCs and may be a target for treating Th2-allergic asthma and other settings with low cellular cAMP concentrations.
Collapse
Affiliation(s)
- Amy M. Chinn
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Cristina Salmerón
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Jihyung Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Paul A. Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
13
|
Rajendeeran A, Tenbrock K. Regulatory T cell function in autoimmune disease. J Transl Autoimmun 2022; 4:100130. [PMID: 35005594 PMCID: PMC8716637 DOI: 10.1016/j.jtauto.2021.100130] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases are characterized by a failure of tolerance to own body components resulting in tissue damage. Regulatory T cells are gatekeepers of tolerance. This review focusses on the function and pathophysiology of regulatory T cells in the context of autoimmune diseases including rheumatoid and juvenile idiopathic arthritis as well as systemic lupus erythematosus with an overview over current and future therapeutic options to boost Treg function. Regulatory T cells are critical mediators of immune tolerance and critically depend on external IL-2. Tregs are expanded during inflammation, where the local milieu enhances resistance to suppression in T effector cells. Human Tregs are characterized by different markers, which hampers the comparability of studies in patients with autoimmunity.
Collapse
Affiliation(s)
- Anandi Rajendeeran
- RWTH Aachen University, Department of Pediatrics, Pediatric Rheumatology, Pauwelsstr 30, 52074, Aachen, Germany
| | - Klaus Tenbrock
- RWTH Aachen University, Department of Pediatrics, Pediatric Rheumatology, Pauwelsstr 30, 52074, Aachen, Germany
| |
Collapse
|
14
|
Sriram K, Insel MB, Insel PA. Inhaled β2 Adrenergic Agonists and Other cAMP-Elevating Agents: Therapeutics for Alveolar Injury and Acute Respiratory Disease Syndrome? Pharmacol Rev 2021; 73:488-526. [PMID: 34795026 DOI: 10.1124/pharmrev.121.000356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022] Open
Abstract
Inhaled long-acting β-adrenergic agonists (LABAs) and short-acting β-adrenergic agonists are approved for the treatment of obstructive lung disease via actions mediated by β2 adrenergic receptors (β2-ARs) that increase cellular cAMP synthesis. This review discusses the potential of β2-AR agonists, in particular LABAs, for the treatment of acute respiratory distress syndrome (ARDS). We emphasize ARDS induced by pneumonia and focus on the pathobiology of ARDS and actions of LABAs and cAMP on pulmonary and immune cell types. β2-AR agonists/cAMP have beneficial actions that include protection of epithelial and endothelial cells from injury, restoration of alveolar fluid clearance, and reduction of fibrotic remodeling. β2-AR agonists/cAMP also exert anti-inflammatory effects on the immune system by actions on several types of immune cells. Early administration is likely critical for optimizing efficacy of LABAs or other cAMP-elevating agents, such as agonists of other Gs-coupled G protein-coupled receptors or cyclic nucleotide phosphodiesterase inhibitors. Clinical studies that target lung injury early, prior to development of ARDS, are thus needed to further assess the use of inhaled LABAs, perhaps combined with inhaled corticosteroids and/or long-acting muscarinic cholinergic antagonists. Such agents may provide a multipronged, repurposing, and efficacious therapeutic approach while minimizing systemic toxicity. SIGNIFICANCE STATEMENT: Acute respiratory distress syndrome (ARDS) after pulmonary alveolar injury (e.g., certain viral infections) is associated with ∼40% mortality and in need of new therapeutic approaches. This review summarizes the pathobiology of ARDS, focusing on contributions of pulmonary and immune cell types and potentially beneficial actions of β2 adrenergic receptors and cAMP. Early administration of inhaled β2 adrenergic agonists and perhaps other cAMP-elevating agents after alveolar injury may be a prophylactic approach to prevent development of ARDS.
Collapse
Affiliation(s)
- Krishna Sriram
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| | - Michael B Insel
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| | - Paul A Insel
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| |
Collapse
|
15
|
Johann K, Bohn T, Shahneh F, Luther N, Birke A, Jaurich H, Helm M, Klein M, Raker VK, Bopp T, Barz M, Becker C. Therapeutic melanoma inhibition by local micelle-mediated cyclic nucleotide repression. Nat Commun 2021; 12:5981. [PMID: 34645812 PMCID: PMC8514514 DOI: 10.1038/s41467-021-26269-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 09/28/2021] [Indexed: 12/03/2022] Open
Abstract
The acidic tumor microenvironment in melanoma drives immune evasion by up-regulating cyclic adenosine monophosphate (cAMP) in tumor-infiltrating monocytes. Here we show that the release of non-toxic concentrations of an adenylate cyclase (AC) inhibitor from poly(sarcosine)-block-poly(L-glutamic acid γ-benzyl ester) (polypept(o)id) copolymer micelles restores antitumor immunity. In combination with selective, non-therapeutic regulatory T cell depletion, AC inhibitor micelles achieve a complete remission of established B16-F10-OVA tumors. Single-cell sequencing of melanoma-infiltrating immune cells shows that AC inhibitor micelles reduce the number of anti-inflammatory myeloid cells and checkpoint receptor expression on T cells. AC inhibitor micelles thus represent an immunotherapeutic measure to counteract melanoma immune escape. The acidic tumour microenvironment in melanoma drives immune evasion by cAMP in tumor-infiltrating monocytes. Here, the authors show that the release of an adenylate cyclase inhibitor from micelles restores antitumor immunity and, when combined with regulatory T cell depletion, leads to remission of established B16-F10-OVA tumors.
Collapse
Affiliation(s)
- Kerstin Johann
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Toszka Bohn
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Fatemeh Shahneh
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Natascha Luther
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Alexander Birke
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Henriette Jaurich
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany.,Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Mark Helm
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Verena K Raker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany.,Department of Dermatology, University Hospital Münster, Westfälische Wilhelms-University, Münster, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany.
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University, Mainz, Germany. .,Leiden Academic Center for Drug Research (LACDR), Leiden, Netherlands.
| | - Christian Becker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany. .,Department of Dermatology, University Hospital Münster, Westfälische Wilhelms-University, Münster, Germany.
| |
Collapse
|
16
|
Ring S, Inaba Y, Da M, Bopp T, Grabbe S, Enk A, Mahnke K. Regulatory T Cells Prevent Neutrophilic Infiltration of Skin during Contact Hypersensitivity Reactions by Strengthening the Endothelial Barrier. J Invest Dermatol 2021; 141:2006-2017. [PMID: 33675787 DOI: 10.1016/j.jid.2021.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
The healing phase of contact hypersensitivity reactions is critically dependent on regulatory T cells (Tregs), but even the early inflammatory phase, that is, 6-24 hours after induction of a contact hypersensitivity reaction, is susceptible to Treg-mediated suppression. To investigate the underlying mechanisms, we injected Tregs before the challenge and analyzed the skin-infiltrating cells as early as 6 hours later. Early on, we found mainly neutrophils in the challenged skin, but only a few T cells. This influx of neutrophils was blocked by the injection of Tregs, indicating that they were able to prevent the first wave of leukocytes, which are responsible for starting an immune reaction. As an underlying mechanism, we identified that Tregs can tighten endothelial junctions by inducing intracellular cAMP, leading to protein kinase A-RhoA‒dependent signaling. This eventually reorganizes endothelial junction proteins, such as Notch3, Nectin 2, Filamin B, and VE-cadherin, all of which contribute to the tightening of the endothelial barrier. In summary, Tregs prevent the leakage of proinflammatory cells from and into the tissue, which establishes a mechanism to downregulate immune reactions.
Collapse
Affiliation(s)
- Sabine Ring
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Yutaka Inaba
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Meihong Da
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Alexander Enk
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Karsten Mahnke
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany.
| |
Collapse
|
17
|
Kuczma MP, Szurek EA, Cebula A, Ngo VL, Pietrzak M, Kraj P, Denning TL, Ignatowicz L. Self and microbiota-derived epitopes induce CD4 + T cell anergy and conversion into CD4 +Foxp3 + regulatory cells. Mucosal Immunol 2021; 14:443-454. [PMID: 33139845 PMCID: PMC7946630 DOI: 10.1038/s41385-020-00349-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/31/2020] [Accepted: 09/23/2020] [Indexed: 02/04/2023]
Abstract
The physiological role of T cell anergy induction as a key mechanism supporting self-tolerance remains undefined, and natural antigens that induce anergy are largely unknown. In this report, we used TCR sequencing to show that the recruitment of CD4+CD44+Foxp3-CD73+FR4+ anergic (Tan) cells expands the CD4+Foxp3+ (Tregs) repertoire. Next, we report that blockade in peripherally-induced Tregs (pTregs) formation due to mutation in CNS1 region of Foxp3 or chronic exposure to a selecting self-peptide result in an accumulation of Tan cells. Finally, we show that microbial antigens from Akkermansia muciniphila commensal bacteria can induce anergy and drive conversion of naive CD4+CD44-Foxp3- T (Tn) cells to the Treg lineage. Overall, data presented here suggest that Tan induction helps the Treg repertoire to become optimally balanced to provide tolerance toward ubiquitous and microbiome-derived epitopes, improving host ability to avert systemic autoimmunity and intestinal inflammation.
Collapse
Affiliation(s)
- Michal P Kuczma
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Edyta A Szurek
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Anna Cebula
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Vu L Ngo
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Maciej Pietrzak
- Mathematical Biosciences Institute, Ohio State University, Columbus, OH, USA
| | - Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Timothy L Denning
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Leszek Ignatowicz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
18
|
Longhi MS, Mieli-Vergani G, Vergani D. Regulatory T cells in autoimmune hepatitis: an updated overview. J Autoimmun 2021; 119:102619. [PMID: 33652348 DOI: 10.1016/j.jaut.2021.102619] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Regulatory T-cells (Tregs) are key players in the maintenance of immune homeostasis by preventing immune responses to self-antigens. Defects in Treg frequency and/or function result in overwhelming CD4 and CD8 T cell immune responses participating in the autoimmune attack. Perpetuation of autoimmune damage is also favored by Treg predisposition to acquire effector cell features upon exposure to a proinflammatory challenge. Treg impairment plays a permissive role in the initiation and perpetuation of autoimmune liver diseases, namely autoimmune hepatitis, primary biliary cholangitis and primary sclerosing cholangitis. In this Review, we outline studies reporting the role of Treg impairment in the pathogenesis of these conditions and discuss methods to restore Treg number and function either by generation/expansion in the test tube or through in vivo expansion upon administration of low dose IL-2. Challenges and caveats of these potential therapeutic strategies are also reviewed and discussed.
Collapse
Affiliation(s)
- Maria Serena Longhi
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| | - Giorgina Mieli-Vergani
- Institute of Liver Studies, MowatLabs, Department of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Liver Sciences and Medicine, King's College London, London, United Kingdom.
| | - Diego Vergani
- Institute of Liver Studies, MowatLabs, Department of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Liver Sciences and Medicine, King's College London, London, United Kingdom.
| |
Collapse
|
19
|
Induction of Allograft Tolerance While Maintaining Immunity Against Microbial Pathogens: Does Coronin 1 Hold a Key? Transplantation 2020; 104:1350-1357. [PMID: 31895336 DOI: 10.1097/tp.0000000000003101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Selective suppression of graft rejection while maintaining anti-pathogen responses has been elusive. Thus far, the most successful strategies to induce suppression of graft rejection relies on inhibition of T-cell activation. However, the very same mechanisms that induce allograft-specific T-cell suppression are also important for immunity against microbial pathogens as well as oncogenically transformed cells, resulting in significant immunosuppression-associated comorbidities. Therefore, defining the pathways that differentially regulate anti-graft versus antimicrobial T-cell responses may allow the development of regimen to induce allograft-specific tolerance. Recent work has defined a molecular pathway driven by the immunoregulatory protein coronin 1 that regulates the phosphodiesterase/cyclic adenosine monophosphate pathway and modulates T cell responses. Interestingly, disruption of coronin 1 promotes allograft tolerance while immunity towards a range of pathogenic microbes is maintained. Here, we briefly review the work leading up to these findings as well as their possible implications for transplantation medicine.
Collapse
|
20
|
Chinn AM, Insel PA. Cyclic AMP in dendritic cells: A novel potential target for disease-modifying agents in asthma and other allergic disorders. Br J Pharmacol 2020; 177:3363-3377. [PMID: 32372523 DOI: 10.1111/bph.15095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/27/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
Allergic diseases are immune disorders that are a global health problem, affecting a large portion of the world's population. Allergic asthma is a heterogeneous disease that alters the biology of the airway. A substantial portion of patients with asthma do not respond to conventional therapies; thus, new and effective therapeutics are needed. Dendritic cells (DCs), antigen presenting cells that regulate helper T cell differentiation, are key drivers of allergic inflammation but are not the target of current therapies. Here we review the role of dendritic cells in allergic conditions and propose a disease-modifying strategy for treating allergic asthma: cAMP-mediated inhibition of dendritic cells to blunt allergic inflammation. This approach contrasts with current treatments that focus on treating clinical manifestations of airway inflammation. Disease-modifying agents that target cAMP and its signalling pathway in dendritic cells may provide a novel means to treat asthma and other allergic diseases.
Collapse
Affiliation(s)
- Amy M Chinn
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Paul A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
21
|
Dorneles GP, dos Passos AA, Romão PR, Peres A. New Insights about Regulatory T Cells Distribution and Function with Exercise: The Role of Immunometabolism. Curr Pharm Des 2020; 26:979-990. [DOI: 10.2174/1381612826666200305125210] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/08/2020] [Indexed: 02/08/2023]
Abstract
A lack of physical activity is linked to the development of many chronic diseases through a chronic
low-grade inflammation state. It is now well accepted that the immune system plays a central role in the development
of several chronic diseases, including insulin resistance, type 2 diabetes, atherosclerosis, heart failure and
certain types of cancer. Exercise elicits a strong anti-inflammatory response independently of weight loss and can
be a useful non-pharmacologic strategy to counteract the low-grade inflammation. The CD4+CD25+CD127-
FoxP3+ Regulatory T (Treg) cells are a unique subset of helper T-cells, which regulate immune response and
establish self-tolerance through the secretion of immunoregulatory cytokines, such as IL-10 and TGF-β, and the
suppression of the function and activity of many immune effector cells (including monocytes/macrophages, dendritic
cells, CD4+ and CD8+ T cells, and Natural Killers). The metabolic phenotype of Tregs are regulated by the
transcription factor Foxp3, providing flexibility in fuel choice, but a preference for higher fatty acid oxidation. In
this review, we focus on the mechanisms by which exercise - both acute and chronic - exerts its antiinflammatory
effects through Treg cells mobilization. Furthermore, we discuss the implications of immunometabolic
changes during exercise for the modulation of Treg phenotype and its immunosuppressive function. This
narrative review focuses on the current knowledge regarding the role of Treg cells in the context of acute and
chronic exercise using data from observational and experimental studies. Emerging evidence suggests that the
immunomodulatory effects of exercise are mediated by the ability of exercise to adjust and improve Tregs number
and function.
Collapse
Affiliation(s)
- Gilson P. Dorneles
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| | - Aline A.Z. dos Passos
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| | - Pedro R.T. Romão
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| | - Alessandra Peres
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| |
Collapse
|
22
|
The chilling of adenylyl cyclase 9 and its translational potential. Cell Signal 2020; 70:109589. [PMID: 32105777 DOI: 10.1016/j.cellsig.2020.109589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/21/2020] [Accepted: 02/23/2020] [Indexed: 12/26/2022]
Abstract
A recent break-through paper has revealed for the first time the high-resolution, three-dimensional structure of a mammalian trans-membrane adenylyl cyclase (tmAC) obtained by cryo-electronmicroscopy (cryo-EM). Reporting the structure of adenylyl cyclase 9 (AC9) in complex with activated Gsα, the cryo-EM study revealed that AC9 has three functionally interlinked, yet structurally distinct domains. The array of the twelve transmembrane helices is connected to the cytosolic catalytic core by two helical segments that are stabilized through the formation of a parallel coiled-coil. Surprisingly, in the presence of Gsα, the isoform-specific carboxyl-terminal tail of AC9 occludes the forskolin- as well as the active substrate-sites, resulting in marked autoinhibition of the enzyme. As AC9 has the lowest primary sequence homology with the eight further mammalian tmAC paralogues, it appears to be the best candidate for selective pharmacologic targeting. This is now closer to reality as the structural insight provided by the cryo-EM study indicates that all of the three structural domains are potential targets for bioactive agents. The present paper summarizes for molecular physiologists and pharmacologists what is known about the biological role of AC9, considers the potential modes of physiologic regulation, as well as pharmacologic targeting on the basis of the high-resolution cryo-EM structure. The translational potential of AC9 is considered upon highlighting the current state of genome-wide association screens, and the corresponding experimental evidence. Overall, whilst the high- resolution structure presents unique opportunities for the full understanding of the control of AC9, the data on the biological role of the enzyme and its translational potential are far from complete, and require extensive further study.
Collapse
|
23
|
Arroyo Hornero R, Hamad I, Côrte-Real B, Kleinewietfeld M. The Impact of Dietary Components on Regulatory T Cells and Disease. Front Immunol 2020; 11:253. [PMID: 32153577 PMCID: PMC7047770 DOI: 10.3389/fimmu.2020.00253] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
The rise in the prevalence of autoimmune diseases in developed societies has been associated with a change in lifestyle patterns. Among other factors, increased consumption of certain dietary components, such as table salt and fatty acids and excessive caloric intake has been associated with defective immunological tolerance. Dietary nutrients have shown to modulate the immune response by a direct effect on the function of immune cells or, indirectly, by acting on the microbiome of the gastrointestinal tract. FOXP3+ regulatory T cells (Tregs) suppress immune responses and are critical for maintaining peripheral tolerance and immune homeostasis, modulating chronic tissue inflammation and autoimmune disease. It is now well-recognized that Tregs show certain degree of plasticity and can gain effector functions to adapt their regulatory function to different physiological situations during an immune response. However, plasticity of Tregs might also result in conversion into effector T cells that may contribute to autoimmune pathogenesis. Yet, which environmental cues regulate Treg plasticity and function is currently poorly understood, but it is of significant importance for therapeutic purposes. Here we review the current understanding on the effect of certain dietary nutrients that characterize Western diets in Treg metabolism, stability, and function. Moreover, we will discuss the role of Tregs linking diet and autoimmunity and the potential of dietary-based interventions to modulate Treg function in disease.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Beatriz Côrte-Real
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| |
Collapse
|
24
|
Dekkema GJ, Bijma T, Jellema PG, Van Den Berg A, Kroesen BJ, Stegeman CA, Heeringa P, Abdulahad WH, Sanders JS. Increased miR-142-3p Expression Might Explain Reduced Regulatory T Cell Function in Granulomatosis With Polyangiitis. Front Immunol 2019; 10:2170. [PMID: 31572380 PMCID: PMC6751284 DOI: 10.3389/fimmu.2019.02170] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/28/2019] [Indexed: 12/16/2022] Open
Abstract
Objectives: Regulatory T cells (Tregs) are frequently functionally impaired in patients with granulomatosis with polyangiitis (GPA). However, the mechanism underlying their impaired function is unknown. Here, we hypothesized that Treg dysfunction in GPA is due to altered microRNA (miRNA) expression. Methods: RNA isolated from FACS-sorted memory (M) Tregs (CD4+CD45RO+CD25+CD127-) of 8 healthy controls (HCs) and 8 GPA patients without treatment was subjected to miRNA microarray analysis. Five differentially expressed miRNAs were validated in a larger cohort by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR). An miRNA target gene database search revealed targets that were tested with RT-qPCR in MTregs from patients and HCs. cAMP levels were measured using flow cytometry. Results: Microarray analysis revealed 19 differentially expressed miRNAs, of which miR-142-3p was confirmed to be significantly upregulated in MTregs from GPA patients compared to those from HCs (1.9-fold, p = 0.03). In vitro overexpression of miR-142-3p lowered the suppressive capacity of MTregs (2.1-fold, p = 0.03), and miR-142-3p expression correlated negatively with the suppressive capacity (rho = -0.446, p = 0.04). Overexpression of miR-142-3p significantly decreased cAMP levels (p = 0.02) and tended to decrease the mRNA levels of a predicted target gene, adenylate cyclase 9 (ADCY9; p = 0.06). In comparison to those from HCs, MTregs from GPA patients had lower ADCY9 mRNA levels (2-fold, p = 0.008) and produced significantly less cAMP after stimulation. Importantly, induction of cAMP production in miR-142-3p overexpressed MTregs by forskolin restored their suppressive function in vitro. Conclusion: Overexpression of miR-142-3p in MTregs from GPA patients might cause functional impairment by targeting ADCY9, which leads to the suppression of cAMP production.
Collapse
Affiliation(s)
- Gerjan J Dekkema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Theo Bijma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Pytrick G Jellema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anke Van Den Berg
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bart-Jan Kroesen
- Medical Immunology Laboratory, Department of Laboratory Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Coen A Stegeman
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wayel H Abdulahad
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan-Stephan Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
25
|
Adenine nucleotides as paracrine mediators and intracellular second messengers in immunity and inflammation. Biochem Soc Trans 2019; 47:329-337. [PMID: 30674608 DOI: 10.1042/bst20180419] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
Adenine nucleotides (AdNs) play important roles in immunity and inflammation. Extracellular AdNs, such as adenosine triphosphate (ATP) or nicotinamide adenine dinucleotide (NAD) and their metabolites, act as paracrine messengers by fine-tuning both pro- and anti-inflammatory processes. Moreover, intracellular AdNs derived from ATP or NAD play important roles in many cells of the immune system, including T lymphocytes, macrophages, neutrophils and others. These intracellular AdNs are signaling molecules that transduce incoming signals into meaningful cellular responses, e.g. activation of immune responses against pathogens.
Collapse
|
26
|
Jayachandran R, Gumienny A, Bolinger B, Ruehl S, Lang MJ, Fucile G, Mazumder S, Tchang V, Woischnig AK, Stiess M, Kunz G, Claudi B, Schmaler M, Siegmund K, Li J, Dertschnig S, Holländer G, Medina E, Karrer U, Moshous D, Bumann D, Khanna N, Rossi SW, Pieters J. Disruption of Coronin 1 Signaling in T Cells Promotes Allograft Tolerance while Maintaining Anti-Pathogen Immunity. Immunity 2019; 50:152-165.e8. [PMID: 30611611 DOI: 10.1016/j.immuni.2018.12.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/18/2018] [Accepted: 12/10/2018] [Indexed: 11/18/2022]
Abstract
The ability of the immune system to discriminate self from non-self is essential for eradicating microbial pathogens but is also responsible for allograft rejection. Whether it is possible to selectively suppress alloresponses while maintaining anti-pathogen immunity remains unknown. We found that mice deficient in coronin 1, a regulator of naive T cell homeostasis, fully retained allografts while maintaining T cell-specific responses against microbial pathogens. Mechanistically, coronin 1-deficiency increased cyclic adenosine monophosphate (cAMP) concentrations to suppress allo-specific T cell responses. Costimulation induced on microbe-infected antigen presenting cells was able to overcome cAMP-mediated immunosuppression to maintain anti-pathogen immunity. In vivo pharmacological modulation of this pathway or a prior transfer of coronin 1-deficient T cells actively suppressed allograft rejection. These results define a coronin 1-dependent regulatory axis in T cells important for allograft rejection and suggest that modulation of this pathway may be a promising approach to achieve long-term acceptance of mismatched allografts.
Collapse
Affiliation(s)
| | | | | | | | | | - Geoffrey Fucile
- Swiss Institute of Bioinformatics, sciCORE Computing Center, University of Basel, Basel, Switzerland
| | | | | | - Anne-Kathrin Woischnig
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | | | | | | | - Mathias Schmaler
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | | | | | - Simone Dertschnig
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - George Holländer
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland; Department of Paediatrics, University of Oxford, Oxford, UK
| | - Eva Medina
- Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Urs Karrer
- Division of Infectious Diseases and Department of Medicine, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Despina Moshous
- Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France and APHP Hôpital Universitaire Necker-Enfants Malades, Unité d'Immunologie-Hématologie et Rhumatologie Pédiatrique, Paris, France
| | - Dirk Bumann
- Biozentrum, University of Basel, Basel, Switzerland
| | - Nina Khanna
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland; Division of Infectious Diseases, University and University Hospital of Basel, Switzerland
| | - Simona W Rossi
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Jean Pieters
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
27
|
Membrane-Permeable Octanoyloxybenzyl-Masked cNMPs As Novel Tools for Non-Invasive Cell Assays. Molecules 2018; 23:molecules23112960. [PMID: 30428589 PMCID: PMC6278358 DOI: 10.3390/molecules23112960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 11/17/2022] Open
Abstract
Adenine nucleotide (AN) 2nd messengers, such as 3′,5′-cyclic adenosine monophosphate (cAMP), are central elements of intracellular signaling, but many details of their underlying processes remain elusive. Like all nucleotides, cyclic nucleotide monophosphates (cNMPs) are net-negatively charged at physiologic pH which limits their applicability in cell-based settings. Thus, many cellular assays rely on sophisticated techniques like microinjection or electroporation. This setup is not feasible for medium- to high-throughput formats, and the mechanic stress that cells are exposed to raises the probability of interfering artefacts or false-positives. Here, we present a short and flexible chemical route yielding membrane-permeable, bio-reversibly masked cNMPs for which we employed the octanoyloxybenzyl (OB) group. We further show hydrolysis studies on chemical stability and enzymatic activation, and present results of real-time assays, where we used cAMP and Ca2+ live cell imaging to demonstrate high permeability and prompt intracellular conversion of some selected masked cNMPs. Based on these results, our novel OB-masked cNMPs constitute valuable precursor-tools for non-invasive studies on intracellular signaling.
Collapse
|
28
|
The Costimulatory Pathways and T Regulatory Cells in Ischemia-Reperfusion Injury: A Strong Arm in the Inflammatory Response? Int J Mol Sci 2018; 19:ijms19051283. [PMID: 29693595 PMCID: PMC5983665 DOI: 10.3390/ijms19051283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 04/14/2018] [Accepted: 04/19/2018] [Indexed: 02/08/2023] Open
Abstract
Costimulatory molecules have been identified as crucial regulators in the inflammatory response in various immunologic disease models. These molecules are classified into four different families depending on their structure. Here, we will focus on various ischemia studies that use costimulatory molecules as a target to reduce the inherent inflammatory status. Furthermore, we will discuss the relevant role of T regulatory cells in these inflammatory mechanisms and the costimulatory pathways in which they are involved.
Collapse
|
29
|
Abstract
Nuclear factor of activated T cells (NFAT) was first described almost three decades ago as a Ca
2+/calcineurin-regulated transcription factor in T cells. Since then, a large body of research uncovered the regulation and physiological function of different NFAT homologues in the immune system and many other tissues. In this review, we will discuss novel roles of NFAT in T cells, focusing mainly on its function in humoral immune responses, immunological tolerance, and the regulation of immune metabolism.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
30
|
Dugger KJ, Chrisman T, Sayner SL, Chastain P, Watson K, Estes R. Beta-2 adrenergic receptors increase TREG cell suppression in an OVA-induced allergic asthma mouse model when mice are moderate aerobically exercised. BMC Immunol 2018; 19:9. [PMID: 29452585 PMCID: PMC5816563 DOI: 10.1186/s12865-018-0244-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
Background The potency of T regulatory (TREG) cells to inhibit T helper (Th)-driven immune cell responses has been linked to increased intracellular cyclic-AMP (cAMP) levels of TREG cells. In an ovalbumin (OVA)-driven allergic asthma mouse model, moderate aerobic exercise increases TREG cell function in a contact-dependent manner that leads to a significant reduction in chronic inflammation and restoration of lung function. However, the mechanism, whereby exercise increases TREG function, remains unknown and was the focus of these investigations. Exercise can communicate with TREG cells by their expression of β2-adrenergic receptors (β2-AR). Activation of these receptors results in an increase in intracellular levels of cyclic-AMP, potentially creating a potent inhibitor of Th cell responses. Results For the allergic asthma model, female wildtype BALB/c mice were challenged with OVA, and exercised (13.5 m/min for 45 min) 3×/week for 4 weeks. TREG cells were isolated from all mouse asthma/exercise groups, including β2-AR−/− mice, to test suppressive function and intracellular cAMP levels. In these studies, cAMP levels were increased in TREG cells isolated from exercised mice. When β2-AR expression was absent on TREG cells, cAMP levels were significantly decreased. Correlatively, their suppressive function was compromised. Next, TREG cells from all mouse groups were tested for suppressive function after treatment with either a pharmaceutical β2-adrenergic agonist or an effector-specific cAMP analogue. These experiments showed TREG cell function was increased when treated with either a β2-adrenergic agonist or effector-specific cAMP analogue. Finally, female wildtype BALB/c mice were antibody-depleted of CD25+CD4+ TREG cells (anti-CD25). Twenty-four hours after TREG depletion, either β2-AR−/− or wildtype TREG cells were adoptively transferred. Recipient mice underwent the asthma/exercise protocols. β2-AR−/− TREG cells isolated from these mice showed no increase in TREG function in response to moderate aerobic exercise. Conclusion These studies offer a novel role for β2-AR in regulating cAMP intracellular levels that can modify suppressive function in TREG cells.
Collapse
Affiliation(s)
- Kari J Dugger
- Department of Clinical and Diagnostic Sciences, Biomedical Sciences, School of Health Professions, University of Alabama at Birmingham, 1716 9th Ave S, SHPB 472, Birmingham, 35294, AL, USA.
| | - Taylor Chrisman
- Department of Biomedical Sciences, College of Allied Health; University of South Alabama, 5721 USA Dr. N, HAHN 4021, Mobile, 36688, AL, USA
| | - Sarah L Sayner
- Department of Physiology and Cell Biology, Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, 36688, AL, USA
| | - Parker Chastain
- Department of Biomedical Sciences, College of Allied Health; University of South Alabama, 5721 USA Dr. N, HAHN 4021, Mobile, 36688, AL, USA
| | - Kacie Watson
- Department of Biomedical Sciences, College of Allied Health; University of South Alabama, 5721 USA Dr. N, HAHN 4021, Mobile, 36688, AL, USA
| | - Robert Estes
- Department of Clinical and Diagnostic Sciences, Biomedical Sciences, School of Health Professions, University of Alabama at Birmingham, 1716 9th Ave S, SHPB 472, Birmingham, 35294, AL, USA
| |
Collapse
|
31
|
Teixeira HMP, Alcantara-Neves NM, Barreto M, Figueiredo CA, Costa RS. Adenylyl cyclase type 9 gene polymorphisms are associated with asthma and allergy in Brazilian children. Mol Immunol 2017; 82:137-145. [PMID: 28076799 DOI: 10.1016/j.molimm.2017.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/21/2016] [Accepted: 01/01/2017] [Indexed: 01/07/2023]
Abstract
Asthma is a chronic inflammatory disease of the respiratory tract. This heterogeneous disease is caused by the interaction of interindividual genetic variability and environmental factors. The gene adenylyl cyclase type 9 (ADCY9) encodes a protein called adenylyl cyclase (AC), responsible for producing the second messenger cyclic AMP (cAMP). cAMP is produced by T regulatory cells and is involved in the down-regulation of T effector cells. Failures in cAMP production may be related to an imbalance in the regulatory immune response, leading to immune-mediated diseases, such as allergic disorders. The aim of this study was to investigate how polymorphisms in the ADCY9 are associated with asthma and allergic markers. The study comprised 1309 subjects from the SCAALA (Social Changes Asthma and Allergy in Latin America) program. Genotyping was accomplished using the Illumina 2.5 Human Omni bead chip. Logistic regression was used to assess the association between allergy markers and ADCY9 variation in PLINK 1.07 software with adjustments for sex, age, helminth infection and ancestry markers. The ADCY9 candidate gene was associated with different phenotypes, such as asthma, specific IgE, skin prick test, and cytokine production. Among 133 markers analyzed, 29 SNPs where associated with asthma and allergic markers in silico analysis revealed the functional impact of the 6 SNPs on ADCY9 expression. It can be concluded that polymorphisms in the ADCY9 gene are significantly associated with asthma and/or allergy markers. We believe that such polymorphisms may lead to increased expression of adenylyl cyclase with a consequent increase in immunoregulatory activity. Therefore, these SNPs may offer an impact on the occurrence of these conditions in admixture population from countries such as Brazil.
Collapse
Affiliation(s)
- Helena M P Teixeira
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | | | - Maurício Barreto
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Bahia, Brazil
| | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | - Ryan S Costa
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Bahia, Brazil.
| |
Collapse
|
32
|
Riccomi A, Gesa V, Sacchi A, De Magistris MT, Vendetti S. Modulation of Phenotype and Function of Human CD4 +CD25 + T Regulatory Lymphocytes Mediated by cAMP-Elevating Agents. Front Immunol 2016; 7:358. [PMID: 27703455 PMCID: PMC5028705 DOI: 10.3389/fimmu.2016.00358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/01/2016] [Indexed: 01/21/2023] Open
Abstract
We have shown that cholera toxin (CT) and other cyclic AMP (cAMP)-elevating agents induce upregulation of the inhibitory molecule CTLA-4 in human resting CD4+ T lymphocytes, which following the treatment acquired suppressive functions. In this study, we evaluated the effect of cAMP-elevating agents on human CD4+CD25+ T cells, which include the T regulatory cells (Tregs) that play a pivotal role in the maintenance of immunological tolerance. We found that cAMP-elevating agents induce upregulation of CTLA-4 in CD4+CD25− and further enhance its expression in CD4+CD25+ T cells. We observed an increase of two isoforms of mRNA coding for the membrane and the soluble CTLA-4 molecules, suggesting that the regulation of CTLA-4 expression by cAMP is at the transcriptional level. In addition, we found that the increase of cAMP in CD4+CD25+ T cells converts the CD4+CD25+Foxp3− T cells in CD4+CD25+Foxp3+ T cells, whereas the increase of cAMP in CD4+CD25− T cells did not upregulate Foxp3 in the absence of activation stimuli. To investigate the function of these cells, we performed an in vitro suppression assay by culturing CD4+CD25+ T cells untreated or pre-treated with CT with anti-CD3 mAbs-stimulated autologous peripheral blood mononuclear cell. We found that CT enhances the inhibitory function of CD4+CD25+ T cells, CD4+, and CD8+ T cell proliferation and IFNγ production are strongly inhibited by CD4+CD25+ T cells pre-treated with cAMP-elevating agents. Furthermore, we found that CD4+CD25+ T lymphocytes pre-treated with cAMP-elevating agents induce the upregulation of CD80 and CD86 co-stimulatory molecules on immature dendritic cells (DCs) in the absence of antigenic stimulation, however without leading to full DC maturation. These data show that the increase of intracellular cAMP modulates the phenotype and function of human CD4+CD25+ T cells.
Collapse
Affiliation(s)
- Antonella Riccomi
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità , Rome , Italy
| | - Valentina Gesa
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità , Rome , Italy
| | - Alessandra Sacchi
- Department of Epidemiology, Preclinical Research, and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani" , Rome , Italy
| | - Maria Teresa De Magistris
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità , Rome , Italy
| | - Silvia Vendetti
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
33
|
Kelly K, Mejia A, Suhasini AN, Lin AP, Kuhn J, Karnad AB, Weitman S, Aguiar RCT. Safety and Pharmacodynamics of the PDE4 Inhibitor Roflumilast in Advanced B-cell Malignancies. Clin Cancer Res 2016; 23:1186-1192. [PMID: 27542768 DOI: 10.1158/1078-0432.ccr-16-1207] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/21/2016] [Accepted: 08/07/2016] [Indexed: 12/31/2022]
Abstract
Purpose: In this study, we aimed to validate our extensive preclinical data on phosphodiesterase 4 (PDE4) as actionable target in B-cell malignancies. Our specific objectives were to determine the safety, pharmacokinetics, and pharmacodynamics (PI3K/AKT activity), as well as to capture any potential antitumor activity of the PDE4 inhibitor roflumilast in combination with prednisone in patients with advanced B-cell malignancies.Experimental Design: Single-center, exploratory phase Ib open-label, nonrandomized study. Roflumilast (500 mcg PO) was given daily for 21 days with prednisone on days 8 to 14. Additional 21-day cycles were started if patients tolerated cycle 1 and had at least stable disease.Results: Ten patients, median age 65 years with an average of three prior therapies, were enrolled. The median number of cycles administered was 4 (range, 1-13). Treatment was well tolerated; the most common ≥grade 2 treatment-related adverse events were fatigue, anorexia (≥25%), and transient ≥ grade 2 neutropenia (30%). Treatment with roflumilast as a single agent significantly suppressed PI3K activity in the 77% of patients evaluated; on average, patients with PI3K/AKT suppression stayed in trial for 156 days (49-315) versus 91 days (28-139 days) for those without this biomarker response. Six of the nine evaluable patients (66%) had partial response or stable disease. The median number of days in trial was 105 days (range, 28-315).Conclusions: Repurposing the PDE4 inhibitor roflumilast for treatment of B-cell malignancies is safe, suppresses the oncogenic PI3K/AKT kinases, and may be clinically active. Clin Cancer Res; 23(5); 1186-92. ©2016 AACR.
Collapse
Affiliation(s)
- Kevin Kelly
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,Institute for Drug Development, Cancer Research and Therapy Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Alex Mejia
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,Institute for Drug Development, Cancer Research and Therapy Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Avvaru N Suhasini
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - An-Ping Lin
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - John Kuhn
- College of Pharmacy, University of Texas Health Science Center at San Antonio and UT Austin, Texas
| | - Anand B Karnad
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,Institute for Drug Development, Cancer Research and Therapy Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Steven Weitman
- Institute for Drug Development, Cancer Research and Therapy Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ricardo C T Aguiar
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas. .,Institute for Drug Development, Cancer Research and Therapy Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas.,Greehey Children's Cancer Research Institute, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas.,South Texas Veterans Health Care System, Audie Murphy VA Hospital, San Antonio, Texas
| |
Collapse
|
34
|
Krzyzanowski MC, Woldemariam S, Wood JF, Chaubey AH, Brueggemann C, Bowitch A, Bethke M, L’Etoile ND, Ferkey DM. Aversive Behavior in the Nematode C. elegans Is Modulated by cGMP and a Neuronal Gap Junction Network. PLoS Genet 2016; 12:e1006153. [PMID: 27459302 PMCID: PMC4961389 DOI: 10.1371/journal.pgen.1006153] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/08/2016] [Indexed: 01/03/2023] Open
Abstract
All animals rely on their ability to sense and respond to their environment to survive. However, the suitability of a behavioral response is context-dependent, and must reflect both an animal's life history and its present internal state. Based on the integration of these variables, an animal's needs can be prioritized to optimize survival strategies. Nociceptive sensory systems detect harmful stimuli and allow for the initiation of protective behavioral responses. The polymodal ASH sensory neurons are the primary nociceptors in C. elegans. We show here that the guanylyl cyclase ODR-1 functions non-cell-autonomously to downregulate ASH-mediated aversive behaviors and that ectopic cGMP generation in ASH is sufficient to dampen ASH sensitivity. We define a gap junction neural network that regulates nociception and propose that decentralized regulation of ASH signaling can allow for rapid correlation between an animal's internal state and its behavioral output, lending modulatory flexibility to this hard-wired nociceptive neural circuit.
Collapse
Affiliation(s)
- Michelle C. Krzyzanowski
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Sarah Woldemariam
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Jordan F. Wood
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Aditi H. Chaubey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Chantal Brueggemann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Alexander Bowitch
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Mary Bethke
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Noelle D. L’Etoile
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Denise M. Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| |
Collapse
|
35
|
Wehbi VL, Taskén K. Molecular Mechanisms for cAMP-Mediated Immunoregulation in T cells - Role of Anchored Protein Kinase A Signaling Units. Front Immunol 2016; 7:222. [PMID: 27375620 PMCID: PMC4896925 DOI: 10.3389/fimmu.2016.00222] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
The cyclic AMP/protein kinase A (cAMP/PKA) pathway is one of the most common and versatile signal pathways in eukaryotic cells. A-kinase anchoring proteins (AKAPs) target PKA to specific substrates and distinct subcellular compartments providing spatial and temporal specificity for mediation of biological effects channeled through the cAMP/PKA pathway. In the immune system, cAMP is a potent negative regulator of T cell receptor-mediated activation of effector T cells (Teff) acting through a proximal PKA/Csk/Lck pathway anchored via a scaffold consisting of the AKAP Ezrin holding PKA, the linker protein EBP50, and the anchoring protein phosphoprotein associated with glycosphingolipid-enriched microdomains holding Csk. As PKA activates Csk and Csk inhibits Lck, this pathway in response to cAMP shuts down proximal T cell activation. This immunomodulating pathway in Teff mediates clinically important responses to regulatory T cell (Treg) suppression and inflammatory mediators, such as prostaglandins (PGs), adrenergic stimuli, adenosine, and a number of other ligands. A major inducer of T cell cAMP levels is PG E2 (PGE2) acting through EP2 and EP4 prostanoid receptors. PGE2 plays a crucial role in the normal physiological control of immune homeostasis as well as in inflammation and cancer immune evasion. Peripherally induced Tregs express cyclooxygenase-2, secrete PGE2, and elicit the immunosuppressive cAMP pathway in Teff as one tumor immune evasion mechanism. Moreover, a cAMP increase can also be induced by indirect mechanisms, such as intercellular transfer between T cells. Indeed, Treg, known to have elevated levels of intracellular cAMP, may mediate their suppressive function by transferring cAMP to Teff through gap junctions, which we speculate could also be regulated by PKA/AKAP complexes. In this review, we present an updated overview on the influence of cAMP-mediated immunoregulatory mechanisms acting through localized cAMP signaling and the therapeutical increasing prospects of AKAPs disruptors in T-cell immune function.
Collapse
Affiliation(s)
- Vanessa L. Wehbi
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Kjetil Taskén
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Centre for Cancer Immunotherapy, Oslo University Hospital, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
36
|
Rueda CM, Jackson CM, Chougnet CA. Regulatory T-Cell-Mediated Suppression of Conventional T-Cells and Dendritic Cells by Different cAMP Intracellular Pathways. Front Immunol 2016; 7:216. [PMID: 27313580 PMCID: PMC4889573 DOI: 10.3389/fimmu.2016.00216] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
Regulatory T-cells (Tregs) mediate their suppressive action by acting directly on conventional T-cells (Tcons) or dendritic cells (DCs). One mechanism of Treg suppression is the increase of cyclic adenosine 3′,5′-monophosphate (cAMP) levels in target cells. Tregs utilize cAMP to control Tcon responses, such as proliferation and cytokine production. Tregs also exert their suppression on DCs, diminishing DC immunogenicity by downmodulating the expression of costimulatory molecules and actin polymerization at the immunological synapse. The Treg-mediated usage of cAMP occurs through two major mechanisms. The first involves the Treg-mediated influx of cAMP in target cells through gap junctions. The second is the conversion of adenosine triphosphate into adenosine by the ectonucleases CD39 and CD73 present on the surface of Tregs. Adenosine then binds to receptors on the surface of target cells, leading to increased intracellular cAMP levels in these targets. Downstream, cAMP can activate the canonical protein kinase A (PKA) pathway and the exchange protein activated by cyclic AMP (EPAC) non-canonical pathway. In this review, we discuss the most recent findings related to cAMP activation of PKA and EPAC, which are implicated in Treg homeostasis as well as the functional alterations induced by cAMP in cellular targets of Treg suppression.
Collapse
Affiliation(s)
- Cesar M Rueda
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine , Cincinnati, OH , USA
| | - Courtney M Jackson
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine , Cincinnati, OH , USA
| | - Claire A Chougnet
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine , Cincinnati, OH , USA
| |
Collapse
|
37
|
López-Abente J, Correa-Rocha R, Pion M. Functional Mechanisms of Treg in the Context of HIV Infection and the Janus Face of Immune Suppression. Front Immunol 2016; 7:192. [PMID: 27242797 PMCID: PMC4871867 DOI: 10.3389/fimmu.2016.00192] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/02/2016] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) play an important role in infections, by modulating host immune responses and avoiding the overreactive immunity that in the case of human immunodeficiency virus (HIV) infection leads to a marked erosion and deregulation of the entire immune system. Therefore, the suppressive function of Treg in HIV-infected patients is critical because of their implication on preventing the immune hyperactivation, even though it could also have a detrimental effect by suppressing HIV-specific immune responses. In recent years, several studies have shown that HIV-1 can directly infect Treg, disturbing their phenotype and suppressive capacity via different mechanisms. These effects include Foxp3 and CD25 downregulation, and the impairment of suppressive capacity. This review describes the functional mechanisms of Treg to modulate immune activation during HIV infection, and how such control is no longer fine-tune orchestrated once Treg itself get infected. We will review the current knowledge about the HIV effects on the Treg cytokine expression, on pathways implying the participation of different ectoenzymes (i.e., CD39/CD73 axis), transcription factors (ICER), and lastly on cyclic adenosine monophosphate (cAMP), one of the keystones in Treg-suppressive function. To define which are the HIV effects upon these regulatory mechanisms is crucial not only for the comprehension of immune deregulation in HIV-infected patients but also for the correct understanding of the role of Tregs in HIV infection.
Collapse
Affiliation(s)
- Jacobo López-Abente
- Laboratory of Immunoregulation, "Gregorio Marañón" Health Research Institute (IISGM) , Madrid , Spain
| | - Rafael Correa-Rocha
- Laboratory of Immunoregulation, "Gregorio Marañón" Health Research Institute (IISGM) , Madrid , Spain
| | - Marjorie Pion
- Laboratory of Immunoregulation, "Gregorio Marañón" Health Research Institute (IISGM) , Madrid , Spain
| |
Collapse
|
38
|
Clark AA, Nurmukhambetova S, Li X, Munger SD, Lees JR. Odorants specifically modulate chemotaxis and tissue retention of CD4+ T cells via cyclic adenosine monophosphate induction. J Leukoc Biol 2016; 100:699-709. [PMID: 27154353 DOI: 10.1189/jlb.1a0914-425rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 04/09/2016] [Indexed: 01/23/2023] Open
Abstract
Retention of T cells within affected tissue is a critical component of adaptive immune inflammation. However, the mechanisms involved in T cell retention remain largely undefined. Previous studies revealed the capacity of cAMP signaling to regulate immune cell migration, as well as dynamic regulation of receptors that could induce cAMP production in immune cells. The potential for cAMP to act as a retention signal has been mostly unexplored, partially as a result of this second messenger's well-characterized inhibition of effector function in immune cells. Here, we report that cAMP regulates the tissue retention of mouse T cells at concentrations well below those that inhibited proliferation or decreased acquisition of an effector phenotype. Stimulation of CD4+ T cells with odorants known to be cognate ligands for T cell-expressed olfactory receptors induced cAMP and inhibited chemokine-driven chemotaxis without decreasing T cell proliferation or effector functions. Similar effects were observed following treatment with relatively low concentrations of the cAMP analog Sp-5,6-dichloro-1-β-d-ribofuranosylbenzimidazole-3',5'-monophosphorothioate. Furthermore, pretreatment with odorants or cAMP at concentrations that did not inhibit effector function induced T cell tissue retention in mice by inhibiting chemokine-dependent T cell egress from the footpad to the draining lymph node. Together, these results suggest that odorant receptor-mediated increases in intracellular cAMP can modulate T cell tissue trafficking and may offer new therapeutic targets for controlling T cell tissue accumulation.
Collapse
Affiliation(s)
- Adam A Clark
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Saule Nurmukhambetova
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Xin Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Steven D Munger
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, USA; and
| | - Jason R Lees
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
39
|
Oligopeptides of Chorionic Gonadotropin β-Subunit in Induction of T Cell Differentiation into Treg and Th17. Bull Exp Biol Med 2015; 160:72-5. [DOI: 10.1007/s10517-015-3101-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Indexed: 01/05/2023]
|
40
|
Hermann-Kleiter N, Klepsch V, Wallner S, Siegmund K, Klepsch S, Tuzlak S, Villunger A, Kaminski S, Pfeifhofer-Obermair C, Gruber T, Wolf D, Baier G. The Nuclear Orphan Receptor NR2F6 Is a Central Checkpoint for Cancer Immune Surveillance. Cell Rep 2015; 12:2072-85. [PMID: 26387951 PMCID: PMC4594157 DOI: 10.1016/j.celrep.2015.08.035] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/02/2015] [Accepted: 08/11/2015] [Indexed: 12/18/2022] Open
Abstract
Nuclear receptor subfamily 2, group F, member 6 (NR2F6) is an orphan member of the nuclear receptor superfamily. Here, we show that genetic ablation of Nr2f6 significantly improves survival in the murine transgenic TRAMP prostate cancer model. Furthermore, Nr2f6(-/-) mice spontaneously reject implanted tumors and develop host-protective immunological memory against tumor rechallenge. This is paralleled by increased frequencies of both CD4(+) and CD8(+) T cells and higher expression levels of interleukin 2 and interferon γ at the tumor site. Mechanistically, CD4(+) and CD8(+) T cell-intrinsic NR2F6 acts as a direct repressor of the NFAT/AP-1 complex on both the interleukin 2 and the interferon γ cytokine promoters, attenuating their transcriptional thresholds. Adoptive transfer of Nr2f6-deficient T cells into tumor-bearing immunocompetent mice is sufficient to delay tumor outgrowth. Altogether, this defines NR2F6 as an intracellular immune checkpoint in effector T cells, governing the amplitude of anti-cancer immunity.
Collapse
Affiliation(s)
- Natascha Hermann-Kleiter
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Victoria Klepsch
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephanie Wallner
- Laboratory of Tumor Immunology, Tyrolean Cancer Institute & Internal Medicine V, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Kerstin Siegmund
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Sebastian Klepsch
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Selma Tuzlak
- Division of Developmental Immunology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Sandra Kaminski
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Christa Pfeifhofer-Obermair
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Gruber
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dominik Wolf
- Laboratory of Tumor Immunology, Tyrolean Cancer Institute & Internal Medicine V, Medical University of Innsbruck, 6020 Innsbruck, Austria; Medical Clinic III, Oncology, Hematology & Rheumatology, University Clinic Bonn, 53127 Bonn, Germany
| | - Gottfried Baier
- Translational Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
41
|
Research Progress on Regulatory T Cells in Acute Kidney Injury. J Immunol Res 2015; 2015:174164. [PMID: 26273681 PMCID: PMC4529954 DOI: 10.1155/2015/174164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/02/2015] [Indexed: 02/06/2023] Open
Abstract
Immune inflammation is crucial in mediating acute kidney injury (AKI). Immune cells of both the innate and adaptive immune systems substantially contribute to overall renal damage in AKI. Regulatory T cells (Tregs) are key regulator of immunological function and have been demonstrated to ameliorate injury in several murine experimental models of renal inflammation. Recent studies have illuminated the renal-protective function of Tregs in AKI. Tregs appear to exert beneficial effects in both the acute injury phase and the recovery phase of AKI. Additionally, Tregs-based immunotherapy may represent a promising approach to ameliorate AKI and promote recovery from AKI. This review will highlight the recent insights into the role of Tregs and their therapeutic potential in AKI.
Collapse
|
42
|
Wang C, Gui Q, Zhang K. Functional polymorphisms in CD86 gene are associated with susceptibility to pneumonia-induced sepsis. APMIS 2015; 123:433-8. [PMID: 25912130 DOI: 10.1111/apm.12364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/17/2014] [Indexed: 11/30/2022]
Abstract
Sepsis is an illness in which the body has a severe response to bacteria or other germs. A bacterial infection in the body such as lungs may set off the response that leads to the disease. CD86 (B7-2) is expressed on various immune cells and plays critical roles in immune responses. Genetic polymorphisms in CD86 gene may affect the development of several diseases. Here, we evaluated the association between two CD86 polymorphisms (rs1915087C/T and rs2332096T/G) and susceptibility to pneumonia-induced sepsis. CD86 rs1915087C/T and rs2332096T/G were identified in 186 pneumonia-induced septic patients and 196 healthy controls in the Chinese population. Results revealed that subjects with rs1915087CT and TT genotypes had significantly lower risk of pneumonia-induced sepsis than those with CC genotype [odds ratio (OR) = 0.58, 95% confidence interval (CI), 0.37-0.91, p = 0.017, and OR = 0.40, 95%CI, 0.21-0.76, p = 0.005]. However, prevalence of rs2332096GG genotype and G allele were significantly increased in patients than in healthy controls (OR = 2.75, 95%CI, 1.46-5.16, p = 0.001, and OR = 1.65, 95%CI, 1.21-2.24, p = 0.001]. We further investigated functions of these two polymorphisms by assessing gene expression in peripheral blood mononuclear cells and in monocytes. Data showed subjects carrying rs2332096GG genotype had significantly decreased level of CD86 in monocytes than those carrying rs2332096TT genotype. These results indicate that CD86 polymorphisms are associated with susceptibility to pneumonia-induced sepsis and may affect gene expression in monocytes.
Collapse
Affiliation(s)
- Chenfei Wang
- Department of Emergency, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | |
Collapse
|
43
|
Ke K, Safder AM, Sul OJ, Suh JH, Joe Y, Chung HT, Choi HS. Cilostazol attenuates ovariectomy-induced bone loss by inhibiting osteoclastogenesis. PLoS One 2015; 10:e0124869. [PMID: 25992691 PMCID: PMC4436362 DOI: 10.1371/journal.pone.0124869] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/21/2015] [Indexed: 01/13/2023] Open
Abstract
Background Cilostazol has been reported to alleviate the metabolic syndrome induced by increased intracellular adenosine 3’,5’-cyclic monophosphate (cAMP) levels, which is also associated with osteoclast (OC) differentiation. We hypothesized that bone loss might be attenuated via an action on OC by cilostazol. Methodology and Principal Findings To test this idea, we investigated the effect of cilostazol on ovariectomy (OVX)-induced bone loss in mice and on OC differentiation in vitro, using μCT and tartrate-resistant acid phosphatase staining, respectively. Cilostazol prevented from OVX-induced bone loss and decreased oxidative stress in vivo. It also decreased the number and activity of OC in vitro. The effect of cilostazol on reactive oxygen species (ROS) occurred via protein kinase A (PKA) and cAMP-regulated guanine nucleotide exchange factor 1, two major effectors of cAMP. Knockdown of NADPH oxidase using siRNA of p47phox attenuated the inhibitory effect of cilostazol on OC formation, suggesting that decreased OC formation by cilostazol was partly due to impaired ROS generation. Cilostazol enhanced phosphorylation of nuclear factor of activated T cells, cytoplasmic 1 (NFAT2) at PKA phosphorylation sites, preventing its nuclear translocation to result in reduced receptor activator of nuclear factor-κB ligand-induced NFAT2 expression and decreased binding of nuclear factor-κB-DNA, finally leading to reduced levels of two transcription factors required for OC differentiation. Conclusions/Significance Our data highlight the therapeutic potential of cilostazol for attenuating bone loss and oxidative stress caused by loss of ovarian function.
Collapse
Affiliation(s)
- Ke Ke
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Ali Muhammad Safder
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Ok-Joo Sul
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan 682-714, Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Hun-Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| |
Collapse
|
44
|
Kuczma M, Wang CY, Ignatowicz L, Gourdie R, Kraj P. Altered connexin 43 expression underlies age-dependent decrease of regulatory T cell suppressor function in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2015; 194:5261-71. [PMID: 25911751 DOI: 10.4049/jimmunol.1400887] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 03/24/2015] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes is one of the most extensively studied autoimmune diseases, but the cellular and molecular mechanisms leading to T cell-mediated destruction of insulin-producing β cells are still not well understood. In this study, we show that regulatory T cells (T(regs)) in NOD mice undergo age-dependent loss of suppressor functions exacerbated by the decreased ability of activated effector T cells to upregulate Foxp3 and generate T(regs) in the peripheral organs. This age-dependent loss is associated with reduced intercellular communication mediated by gap junctions, which is caused by impaired upregulation and decreased expression of connexin 43. Regulatory functions can be corrected, even in T cells isolated from aged, diabetic mice, by a synergistic activity of retinoic acid, TGF-β, and IL-2, which enhance connexin 43 and Foxp3 expression in T(regs) and restore the ability of conventional CD4(+) T cells to upregulate Foxp3 and generate peripherally derived T(regs). Moreover, we demonstrate that suppression mediated by T(regs) from diabetic mice is enhanced by a novel reagent, which facilitates gap junction aggregation. In summary, our report identifies gap junction-mediated intercellular communication as an important component of the T(reg) suppression mechanism compromised in NOD mice and suggests how T(reg) mediated immune regulation can be improved.
Collapse
Affiliation(s)
- Michal Kuczma
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912
| | - Cong-Yi Wang
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912; The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; and
| | - Leszek Ignatowicz
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912
| | - Robert Gourdie
- Virginia Tech Carilion Research Institute, Roanoke, VA 24015
| | - Piotr Kraj
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912;
| |
Collapse
|
45
|
Hoeppli RE, Wu D, Cook L, Levings MK. The environment of regulatory T cell biology: cytokines, metabolites, and the microbiome. Front Immunol 2015; 6:61. [PMID: 25741338 PMCID: PMC4332351 DOI: 10.3389/fimmu.2015.00061] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/30/2015] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Tregs) are suppressive T cells that have an essential role in maintaining the balance between immune activation and tolerance. Their development, either in the thymus, periphery, or experimentally in vitro, and stability and function all depend on the right mix of environmental stimuli. This review focuses on the effects of cytokines, metabolites, and the microbiome on both human and mouse Treg biology. The role of cytokines secreted by innate and adaptive immune cells in directing Treg development and shaping their function is well established. New and emerging data suggest that metabolites, such as retinoic acid, and microbial products, such as short-chain fatty acids, also have a critical role in guiding the functional specialization of Tregs. Overall, the complex interaction between distinct environmental stimuli results in unique, and in some cases tissue-specific, tolerogenic environments. Understanding the conditions that favor Treg induction, accumulation, and function is critical to defining the pathophysiology of many immune-mediated diseases and to developing new therapeutic interventions.
Collapse
Affiliation(s)
- Romy E Hoeppli
- Department of Surgery, Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| | - Dan Wu
- Department of Surgery, Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| | - Laura Cook
- Department of Surgery, Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| | - Megan K Levings
- Department of Surgery, Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
46
|
Zhao F, Xie Q, Xu M, Wang S, Zhou J, Liu F. RNA aptamer based electrochemical biosensor for sensitive and selective detection of cAMP. Biosens Bioelectron 2014; 66:238-43. [PMID: 25437358 DOI: 10.1016/j.bios.2014.11.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/11/2014] [Accepted: 11/14/2014] [Indexed: 01/26/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) is an important small biological molecule associated with the healthy state of living organism. In order to realize highly sensitive and specific detection of cAMP, here an RNA aptamer and electrochemical impedance spectroscopy (EIS) based biosensor enhanced by gold nanoparticles electrodeposited on the surface of gold electrode is designed. The designed aptasensor has a wide effective measuring range from 50pM to 250pM with a detection limit of 50pM in PBS buffer, and an effective measuring range from 50nM to 1μM with a detection limit of 50nM in serum. The designed biosensor is also able to detect cAMP with high sensitivity, specificity, and stability. Since the biosensor can be easily fabricated with low cost and repeatedly used for at least two times, it owns great potential in wide application fields such as clinical test and food inspection, etc.
Collapse
Affiliation(s)
- Fulin Zhao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Qingyun Xie
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Mingfei Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Shouyu Wang
- Department of Information Physics and Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
| | - Jiyong Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Fei Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| |
Collapse
|
47
|
Lelle M, Hameed A, Ackermann LM, Kaloyanova S, Wagner M, Berisha F, Nikolaev VO, Peneva K. Functional non-nucleoside adenylyl cyclase inhibitors. Chem Biol Drug Des 2014; 85:633-7. [PMID: 25319071 DOI: 10.1111/cbdd.12452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 11/28/2022]
Abstract
In this study, we describe the synthesis of novel functional non-nucleoside adenylyl cyclase inhibitors, which can be easily modified with thiol containing biomolecules such as tumour targeting structures. The linkage between inhibitor and biomolecule contains cleavable bonds to enable efficient intracellular delivery in the reductive milieu of the cytosol as well as in the acidic environment within endosomes and lysosomes. The suitability of this synthetic approach was shown by the successful bioconjugation of a poor cell-permeable inhibitor with a cell-penetrating peptide. Additionally, we have demonstrated the excellent inhibitory effect of the compounds presented here in a live-cell Förster resonance energy transfer-based assay in human embryonic kidney cells.
Collapse
Affiliation(s)
- Marco Lelle
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Lindenberg M, Solmaz G, Puttur F, Sparwasser T. Mouse cytomegalovirus infection overrules T regulatory cell suppression on natural killer cells. Virol J 2014; 11:145. [PMID: 25108672 PMCID: PMC4254395 DOI: 10.1186/1743-422x-11-145] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/24/2014] [Indexed: 12/25/2022] Open
Abstract
Background Cytomegalovirus establishes lifelong persistency in the host and leads to life threatening situations in immunocompromised patients. FoxP3+ T regulatory cells (Tregs) critically control and suppress innate and adaptive immune responses. However, their specific role during MCMV infection, especially pertaining to their interaction with NK cells, remains incompletely defined. Methods To understand the contribution of Tregs on NK cell function during acute MCMV infection, we infected Treg depleted and undepleted DEREG mice with WT MCMV and examined Treg and NK cell frequency, number, activation and effector function in vivo. Results Our results reveal an increased frequency of activated Tregs within the CD4+ T cell population shortly after MCMV infection. Specific depletion of Tregs in DEREG mice under homeostatic conditions leads to an increase in NK cell number as well as to a higher activation status of these cells as compared with non-depleted controls. Interestingly, upon infection this effect on NK cells is completely neutralized in terms of cell frequency, CD69 expression and functionality with respect to IFN-γ production. Furthermore, composition of the NK cell population with regard to Ly49H expression remains unchanged. In contrast, absence of Tregs still boosts the general T cell response upon infection to a level comparable to the enhanced activation seen in uninfected mice. CD4+ T cells especially benefit from Treg depletion exhibiting a two-fold increase of CD69+ cells 40 h and IFN-γ+ cells 7 days p.i. while, MCMV infection per se induces robust CD8+ T cell activation which is also further augmented in Treg-depleted mice. Nevertheless, the viral burden in the liver and spleen remain unaltered upon Treg ablation during the course of infection. Conclusions Thus, MCMV infection abolishes Treg suppressing effects on NK cells whereas T cells benefit from their absence during acute infection. This study provides novel information in understanding the collaborative interaction between NK cells and Tregs during a viral infection and provides further knowledge that could be adopted in therapeutic setups to improve current treatment of organ transplant patients where modulation of Tregs is envisioned as a strategy to overcome transplant rejection. Electronic supplementary material The online version of this article (doi:10.1186/1743-422X-11-145) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Franz Puttur
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Feodor-Lynen-Strasse 7, 30625 Hannover, Germany.
| | | |
Collapse
|
49
|
Ohta A, Sitkovsky M. Extracellular adenosine-mediated modulation of regulatory T cells. Front Immunol 2014; 5:304. [PMID: 25071765 PMCID: PMC4091046 DOI: 10.3389/fimmu.2014.00304] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 06/15/2014] [Indexed: 12/20/2022] Open
Abstract
Extracellular adenosine-dependent suppression and redirection of pro-inflammatory activities are mediated by the signaling through adenosine receptors on the surface of most immune cells. The immunosuppression by endogenously-produced adenosine is pathophysiologically significant since inactivation of A2A/A2B adenosine receptor (A2AR/A2BR) and adenosine-producing ecto-enzymes CD39/CD73 results in the higher intensity of immune response and exaggeration of inflammatory damage. Regulatory T cells (Treg) can generate extracellular adenosine, which is implicated in the immunoregulatory activity of Tregs. Interestingly, adenosine has been shown to increase the numbers of Tregs and further promotes their immunoregulatory activity. A2AR-deficiency in Tregs reduces their immunosuppressive efficacy in vivo. Thus, adenosine is not only directly and instantly inhibiting to the immune response through interaction with A2AR/A2BR on the effector cells, but also adenosine signaling can recruit other immunoregulatory mechanisms, including Tregs. Such interaction between adenosine and Tregs suggests the presence of a positive feedback mechanism, which further promotes negative regulation of immune system through the establishment of immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Akio Ohta
- New England Inflammation and Tissue Protection Institute, Northeastern University , Boston, MA , USA
| | - Michail Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University , Boston, MA , USA
| |
Collapse
|
50
|
Pankratz S, Bittner S, Herrmann AM, Schuhmann MK, Ruck T, Meuth SG, Wiendl H. Human CD4+ HLA-G+ regulatory T cells are potent suppressors of graft-versus-host disease in vivo. FASEB J 2014; 28:3435-45. [PMID: 24744146 DOI: 10.1096/fj.14-251074] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CD4(+) T cells expressing the immunotolerizing molecule HLA-G have been described as a unique human thymus-derived regulatory T (tTreg) cell subset involved in immunoregulation and parenchymal homeostasis during infectious and autoimmune inflammation. We compared properties and molecular characteristics of human CD4(+)HLA-G(+) with those of CD4(+)CD25(+)FoxP3-expressing tTreg cells using in vitro studies of T-cell receptor (TCR) signaling, single-cell electrophysiology, and functional in vivo studies. Both tTreg populations are characterized by alterations in proximal-signaling pathways on TCR stimulation and a hyperpolarization of the plasma membrane when compared to conventional CD4(+) T cells. However, both clearly differ in phenotype and pattern of secreted cytokines, which results in distinct mechanisms of suppression: While CD4(+)HLA-G(+) cells secrete high levels of inhibitory molecules (IL-10, soluble HLA-G, IL-35), CD4(+)CD25(+)FoxP3(+) cells express these molecules at significantly lower levels and seem to exert their function mainly in a contact-dependent manner via cyclic adenosine-monophosphate. Finally we demonstrate that human CD4(+)HLA-G(+) tTreg cells significantly ameliorated graft-versus-host disease in a humanized mouse model as a first proof of their in vivo relevance. Our data further characterize and establish CD4(+)HLA-G(+) cells as a potent human tTreg population that can modulate polyclonal adaptive immune responses in vivo and thus being a promising candidate for potential clinical applications in the future.
Collapse
Affiliation(s)
| | | | | | | | | | - Sven G Meuth
- Department of Neurology and Institute of Physiology I, Department of Neuropathophysiology, University of Münster, Münster, Germany; and
| | | |
Collapse
|