1
|
Fesneau O, Thevin V, Pinet V, Goldsmith C, Vieille B, M'Homa Soudja S, Lattanzio R, Hahne M, Dardalhon V, Hernandez-Vargas H, Benech N, Marie JC. An intestinal T H17 cell-derived subset can initiate cancer. Nat Immunol 2024; 25:1637-1649. [PMID: 39060651 PMCID: PMC11362008 DOI: 10.1038/s41590-024-01909-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Approximately 25% of cancers are preceded by chronic inflammation that occurs at the site of tumor development. However, whether this multifactorial oncogenic process, which commonly occurs in the intestines, can be initiated by a specific immune cell population is unclear. Here, we show that an intestinal T cell subset, derived from interleukin-17 (IL-17)-producing helper T (TH17) cells, induces the spontaneous transformation of the intestinal epithelium. This subset produces inflammatory cytokines, and its tumorigenic potential is not dependent on IL-17 production but on the transcription factors KLF6 and T-BET and interferon-γ. The development of this cell type is inhibited by transforming growth factor-β1 (TGFβ1) produced by intestinal epithelial cells. TGFβ signaling acts on the pretumorigenic TH17 cell subset, preventing its progression to the tumorigenic stage by inhibiting KLF6-dependent T-BET expression. This study therefore identifies an intestinal T cell subset initiating cancer.
Collapse
Affiliation(s)
- Olivier Fesneau
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
| | - Valentin Thevin
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
| | - Valérie Pinet
- Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Chloe Goldsmith
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
| | - Baptiste Vieille
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
| | - Saïdi M'Homa Soudja
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
| | - Rossano Lattanzio
- Department of Innovative Technologies in Medicine & Dentistry, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Michael Hahne
- Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Hector Hernandez-Vargas
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
| | - Nicolas Benech
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France
- Hospices Civils de Lyon, Service d'Hépato-Gastroentérologie, Croix Rousse Hospital, Lyon, France
| | - Julien C Marie
- Cancer Research Center of Lyon (CRCL) INSERM U 1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Lyon 1 University, Lyon, France.
- Equipe Labellisée Ligue Nationale Contre le Cancer, Lyon, France.
| |
Collapse
|
2
|
Wang L, Mishra S, Fan KKH, Quon S, Li G, Yu B, Liao W, Liu Y, Zhang X, Qiu Y, Li Y, Goldrath AW, Ma C, Zhang N. T-bet deficiency and Hic1 induction override TGF-β-dependency in the formation of CD103 + intestine-resident memory CD8 + T cells. Cell Rep 2024; 43:114258. [PMID: 38781073 PMCID: PMC11240284 DOI: 10.1016/j.celrep.2024.114258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/01/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Transforming growth factor β (TGF-β) represents a well-established signal required for tissue-resident memory T cell (TRM) formation at intestinal surfaces, regulating the expression of a large collection of genes coordinately promoting intestinal TRM differentiation. The functional contribution from each TGF-β-controlled transcription factor is not entirely known. Here, we find that TGF-β-induced T-bet downregulation and Hic1 induction represent two critical events during intestinal TRM differentiation. Importantly, T-bet deficiency significantly rescues intestinal TRM formation in the absence of the TGF-β receptor. Hic1 induction further strengthens TRM maturation in the absence of TGF-β and T-bet. Our results reveal that provision of certain TGF-β-induced molecular events can partially replace TGF-β signaling to promote the establishment of intestinal TRMs, which allows the functional dissection of TGF-β-induced transcriptional targets and molecular mechanisms for TRM differentiation.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shruti Mishra
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kenneth Ka-Ho Fan
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Sara Quon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Guo Li
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Bingfei Yu
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Wei Liao
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yue Li
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ananda W Goldrath
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; South Texas Veterans Health Care System, San Antonio, TX 78229, USA.
| |
Collapse
|
3
|
Goldsmith C, Thevin V, Fesneau O, Matias MI, Perrault J, Abid AH, Taylor N, Dardalhon V, Marie JC, Hernandez-Vargas H. Single-Molecule DNA Methylation Reveals Unique Epigenetic Identity Profiles of T Helper Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1029-1039. [PMID: 38284984 PMCID: PMC11002815 DOI: 10.4049/jimmunol.2300091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 01/04/2024] [Indexed: 01/30/2024]
Abstract
Both identity and plasticity of CD4 T helper (Th) cells are regulated in part by epigenetic mechanisms. However, a method that reliably and readily profiles DNA base modifications is still needed to finely study Th cell differentiation. Cytosine methylation in CpG context (5mCpG) and cytosine hydroxymethylation (5hmCpG) are DNA modifications that identify stable cell phenotypes, but their potential to characterize intermediate cell transitions has not yet been evaluated. To assess transition states in Th cells, we developed a method to profile Th cell identity using Cas9-targeted single-molecule nanopore sequencing. Targeting as few as 10 selected genomic loci, we were able to distinguish major in vitro polarized murine T cell subtypes, as well as intermediate phenotypes, by their native DNA 5mCpG patterns. Moreover, by using off-target sequences, we were able to infer transcription factor activities relevant to each cell subtype. Detection of 5mCpG and 5hmCpG was validated on intestinal Th17 cells escaping transforming growth factor β control, using single-molecule adaptive sampling. A total of 21 differentially methylated regions mapping to the 10-gene panel were identified in pathogenic Th17 cells relative to their nonpathogenic counterpart. Hence, our data highlight the potential to exploit native DNA methylation profiling to study physiological and pathological transition states of Th cells.
Collapse
Affiliation(s)
- Chloe Goldsmith
- Tumor Escape Resistance and Immunity Department, Cancer Research Center of Lyon, The French League Against Cancer Certified Team, INSERM U1052, CNRS UMR 5286, Léon Bérard Centre and University of Lyon, Lyon, France
| | - Valentin Thevin
- Tumor Escape Resistance and Immunity Department, Cancer Research Center of Lyon, The French League Against Cancer Certified Team, INSERM U1052, CNRS UMR 5286, Léon Bérard Centre and University of Lyon, Lyon, France
| | - Olivier Fesneau
- Tumor Escape Resistance and Immunity Department, Cancer Research Center of Lyon, The French League Against Cancer Certified Team, INSERM U1052, CNRS UMR 5286, Léon Bérard Centre and University of Lyon, Lyon, France
| | - Maria I Matias
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Julie Perrault
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Ali Hani Abid
- Tumor Escape Resistance and Immunity Department, Cancer Research Center of Lyon, The French League Against Cancer Certified Team, INSERM U1052, CNRS UMR 5286, Léon Bérard Centre and University of Lyon, Lyon, France
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Julien C Marie
- Tumor Escape Resistance and Immunity Department, Cancer Research Center of Lyon, The French League Against Cancer Certified Team, INSERM U1052, CNRS UMR 5286, Léon Bérard Centre and University of Lyon, Lyon, France
| | - Hector Hernandez-Vargas
- Tumor Escape Resistance and Immunity Department, Cancer Research Center of Lyon, The French League Against Cancer Certified Team, INSERM U1052, CNRS UMR 5286, Léon Bérard Centre and University of Lyon, Lyon, France
- Genomics Consulting, Bron, France
| |
Collapse
|
4
|
Li G, Srinivasan S, Wang L, Ma C, Guo K, Xiao W, Liao W, Mishra S, Zhang X, Qiu Y, Lu Q, Liu Y, Zhang N. TGF-β-dependent lymphoid tissue residency of stem-like T cells limits response to tumor vaccine. Nat Commun 2022; 13:6043. [PMID: 36229613 PMCID: PMC9562983 DOI: 10.1038/s41467-022-33768-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/01/2022] [Indexed: 12/24/2022] Open
Abstract
TGF-β signaling is necessary for CD8+ T cell differentiation into tissue resident memory T cells (TRM). Although higher frequency of CD8+ TRM cells in the tumor microenvironment is associated with better prognosis, TGF-β-blockade typically improves rather than worsens outcomes. Here we show that in a mouse melanoma model, in the tumor-draining lymph nodes (TDLN) rather than in the tumors themselves, stem-like CD8+ T cells differentiate into TRMs in a TGF-β and tumor antigen dependent manner. Following vaccination against a melanoma-specific epitope, most tumour-specific CD8+ T cells are maintained in a stem-like state, but a proportion of cells lost TRM status and differentiate into CX3CR1+ effector CD8+ T cells in the TDLN, which are subsequently migrating into the tumours. Disruption of TGF-β signaling changes the dynamics of these developmental processes, with the net result of improving effector CD8+ T cell migration into the tumours. In summary, TDLN stem-like T cells transiently switch from a TGF-β-dependent TRM differentiation program to an anti-tumor migratory effector development upon vaccination, which transition can be facilitated by targeted TGF-β blockade.
Collapse
Affiliation(s)
- Guo Li
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Saranya Srinivasan
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Liwen Wang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Kai Guo
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Wenhao Xiao
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Wei Liao
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Dermatology, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan, 410007, China
| | - Shruti Mishra
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
5
|
Mishra S, Liao W, Liu Y, Yang M, Ma C, Wu H, Zhao M, Zhang X, Qiu Y, Lu Q, Zhang N. TGF-β and Eomes control the homeostasis of CD8+ regulatory T cells. J Exp Med 2021; 218:152129. [PMID: 32991667 PMCID: PMC7527976 DOI: 10.1084/jem.20200030] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/03/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022] Open
Abstract
In addition to Foxp3+ CD4+ regulatory T cells (CD4+ T reg cells), Foxp3- CD8+ regulatory T cells (CD8+ T reg cells) are critical to maintain immune tolerance. However, the molecular programs that specifically control CD8+ but not CD4+ T reg cells are largely unknown. Here, we demonstrate that simultaneous disruption of both TGF-β receptor and transcription factor Eomesodermin (Eomes) in T cells results in lethal autoimmunity due to a specific defect in CD8+ but not CD4+ T reg cells. Further, TGF-β signal maintains the regulatory identity, while Eomes controls the follicular location of CD8+ T reg cells. Both TGF-β signal and Eomes coordinate to promote the homeostasis of CD8+ T reg cells. Together, we have identified a unique molecular program designed for CD8+ T reg cells.
Collapse
Affiliation(s)
- Shruti Mishra
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Wei Liao
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX.,Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yong Liu
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX.,Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China
| | - Ming Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China
| | - Yuanzheng Qiu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| |
Collapse
|
6
|
Harnessing Natural Killer Immunity in Metastatic SCLC. J Thorac Oncol 2020; 15:1507-1521. [DOI: 10.1016/j.jtho.2020.05.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/31/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022]
|
7
|
Wu J, Weisshaar N, Hotz-Wagenblatt A, Madi A, Ma S, Mieg A, Hering M, Mohr K, Schlimbach T, Borgers H, Cui G. Skeletal muscle antagonizes antiviral CD8 + T cell exhaustion. SCIENCE ADVANCES 2020; 6:eaba3458. [PMID: 32582853 PMCID: PMC7292629 DOI: 10.1126/sciadv.aba3458] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/28/2020] [Indexed: 05/05/2023]
Abstract
CD8+ T cells become functionally impaired or "exhausted" in chronic infections, accompanied by unwanted body weight reduction and muscle mass loss. Whether muscle regulates T cell exhaustion remains incompletely understood. We report that mouse skeletal muscle increased interleukin (IL)-15 production during LCMV clone 13 chronic infection. Muscle-specific ablation of Il15 enhanced the CD8+ T cell exhaustion phenotype. Muscle-derived IL-15 was required to maintain a population of CD8+CD103+ muscle-infiltrating lymphocytes (MILs). MILs resided in a less inflamed microenvironment, expressed more T cell factor 1 (Tcf1), and had higher proliferative potential than splenic T cells. MILs differentiated into functional effector T cells after reentering lymphoid tissues. Increasing muscle mass via muscle-specific inhibition of TGFβ signaling enhanced IL-15 production and antiviral CD8+ T cell responses. We conclude that skeletal muscle antagonizes T cell exhaustion by protecting T cell proliferative potential from inflammation and replenishing the effector T cell progeny pool in lymphoid organs.
Collapse
Affiliation(s)
- Jingxia Wu
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nina Weisshaar
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Agnes Hotz-Wagenblatt
- Core Facility Omics IT and Data Management, DKFZ, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alaa Madi
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Sicong Ma
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alessa Mieg
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Marvin Hering
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Kerstin Mohr
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Tilo Schlimbach
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Helena Borgers
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Guoliang Cui
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Corresponding author.
| |
Collapse
|
8
|
Abstract
Adenosine is an ancient extracellular signaling molecule that regulates various biological functions via activating four G-protein-coupled receptors, A1, A2A, A2B, and A3 adenosine receptors. As such, several studies have highlighted a role for adenosine signaling in affecting the T cell development in the thymus. Recent studies indicate that adenosine is produced in the context of apoptotic thymocyte clearance. This review critically discusses the involvement of adenosine and its receptors in the complex interplay that exists between the developing thymocytes and the thymic macrophages which engulf the apoptotic cells. This crosstalk contributes to the effective and immunologically silent removal of apoptotic thymocytes, as well as affects the TCR-driven T-cell selection processes.
Collapse
Affiliation(s)
- Krisztina Köröskényi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Basic Medical Sciences of Dental Faculty, University of Debrecen, Debrecen, Hungary
| | - Gergely Joós
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Basic Medical Sciences of Dental Faculty, University of Debrecen, Debrecen, Hungary
| | - Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Basic Medical Sciences of Dental Faculty, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
9
|
Warsinske HC, Pienaar E, Linderman JJ, Mattila JT, Kirschner DE. Deletion of TGF-β1 Increases Bacterial Clearance by Cytotoxic T Cells in a Tuberculosis Granuloma Model. Front Immunol 2017; 8:1843. [PMID: 29326718 PMCID: PMC5742530 DOI: 10.3389/fimmu.2017.01843] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/06/2017] [Indexed: 01/10/2023] Open
Abstract
Mycobacterium tuberculosis is the pathogenic bacterium that causes tuberculosis (TB), one of the most lethal infectious diseases in the world. The only vaccine against TB is minimally protective, and multi-drug resistant TB necessitates new therapeutics to treat infection. Developing new therapies requires a better understanding of the complex host immune response to infection, including dissecting the processes leading to formation of granulomas, the dense cellular lesions associated with TB. In this work, we pair experimental and computational modeling studies to explore cytokine regulation in the context of TB. We use our next-generation hybrid multi-scale model of granuloma formation (GranSim) to capture molecular, cellular, and tissue scale dynamics of granuloma formation. We identify TGF-β1 as a major inhibitor of cytotoxic T-cell effector function in granulomas. Deletion of TGF-β1 from the system results in improved bacterial clearance and lesion sterilization. We also identify a novel dichotomous regulation of cytotoxic T cells and macrophages by TGF-β1 and IL-10, respectively. These findings suggest that increasing cytotoxic T-cell effector functions may increase bacterial clearance in granulomas and highlight potential new therapeutic targets for treating TB.
Collapse
Affiliation(s)
- Hayley C Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elsje Pienaar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, United States
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Qing Y, Gerson SL. Mismatch repair deficient hematopoietic stem cells are preleukemic stem cells. PLoS One 2017; 12:e0182175. [PMID: 28767666 PMCID: PMC5540588 DOI: 10.1371/journal.pone.0182175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/13/2017] [Indexed: 12/27/2022] Open
Abstract
Whereas transformation events in hematopoietic malignancies may occur at different developmental stages, the initial mutation originates in hematopoietic stem cells (HSCs), creating a preleukemic stem cell (PLSC). Subsequent mutations at either stem cell or progenitor cell levels transform the PLSC into lymphoma/leukemia initiating cells (LIC). Thymic lymphomas have been thought to develop from developing thymocytes. T cell progenitors are generated from HSCs in the bone marrow (BM), but maturation and proliferation of T cells as well as T-lymphomagenesis depends on both regulatory mechanisms and microenvironment within the thymus. We studied PLSC linked to thymic lymphomas. In this study, we use MSH2-/- mice as a model to investigate the existence of PLSC and the evolution of PLSC to LIC. Following BM transplantation, we found that MSH2-/- BM cells from young mice are able to fully reconstitute multiple hematopoietic lineages of lethally irradiated wild-type recipients. However, all recipients developed thymic lymphomas within three and four months post transplantation. Transplantation of different fractions of BM cells or thymocytes from young health MSH2-/- mice showed that an HSC enriched fraction always reconstituted hematopoiesis followed by lymphoma development. In addition, lymphomas did not occur in thymectomized recipients of MSH2-/- BM. These results suggest that HSCs with DNA repair defects such as MSH2-/- are PLSCs because they retain hematopoietic function, but also carry an obligate lymphomagenic potential within their T-cell progeny that is dependent on the thymic microenvironment.
Collapse
Affiliation(s)
- Yulan Qing
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Stanton L. Gerson
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
11
|
Buttgereit A, Lelios I, Yu X, Vrohlings M, Krakoski NR, Gautier EL, Nishinakamura R, Becher B, Greter M. Sall1 is a transcriptional regulator defining microglia identity and function. Nat Immunol 2016; 17:1397-1406. [DOI: 10.1038/ni.3585] [Citation(s) in RCA: 333] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/16/2016] [Indexed: 02/07/2023]
|
12
|
Warsinske HC, Wheaton AK, Kim KK, Linderman JJ, Moore BB, Kirschner DE. Computational Modeling Predicts Simultaneous Targeting of Fibroblasts and Epithelial Cells Is Necessary for Treatment of Pulmonary Fibrosis. Front Pharmacol 2016; 7:183. [PMID: 27445819 PMCID: PMC4917547 DOI: 10.3389/fphar.2016.00183] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/10/2016] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is pathologic remodeling of lung tissue that can result in difficulty breathing, reduced quality of life, and a poor prognosis for patients. Fibrosis occurs as a result of insult to lung tissue, though mechanisms of this response are not well-characterized. The disease is driven in part by dysregulation of fibroblast proliferation and differentiation into myofibroblast cells, as well as pro-fibrotic mediator-driven epithelial cell apoptosis. The most well-characterized pro-fibrotic mediator associated with pulmonary fibrosis is TGF-β1. Excessive synthesis of, and sensitivity to, pro-fibrotic mediators as well as insufficient production of and sensitivity to anti-fibrotic mediators has been credited with enabling fibroblast accumulation. Available treatments neither halt nor reverse lung damage. In this study we have two aims: to identify molecular and cellular scale mechanisms driving fibroblast proliferation and differentiation as well as epithelial cell survival in the context of fibrosis, and to predict therapeutic targets and strategies. We combine in vitro studies with a multi-scale hybrid agent-based computational model that describes fibroblasts and epithelial cells in co-culture. Within this model TGF-β1 represents a pro-fibrotic mediator and we include detailed dynamics of TGF-β1 receptor ligand signaling in fibroblasts. PGE2 represents an anti-fibrotic mediator. Using uncertainty and sensitivity analysis we identify TGF-β1 synthesis, TGF-β1 activation, and PGE2 synthesis among the key mechanisms contributing to fibrotic outcomes. We further demonstrate that intervention strategies combining potential therapeutics targeting both fibroblast regulation and epithelial cell survival can promote healthy tissue repair better than individual strategies. Combinations of existing drugs and compounds may provide significant improvements to the current standard of care for pulmonary fibrosis. Thus, a two-hit therapeutic intervention strategy may prove necessary to halt and reverse disease dynamics.
Collapse
Affiliation(s)
- Hayley C. Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Amanda K. Wheaton
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Kevin K. Kim
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | | | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| |
Collapse
|
13
|
Viel S, Marçais A, Guimaraes FSF, Loftus R, Rabilloud J, Grau M, Degouve S, Djebali S, Sanlaville A, Charrier E, Bienvenu J, Marie JC, Caux C, Marvel J, Town L, Huntington ND, Bartholin L, Finlay D, Smyth MJ, Walzer T. TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci Signal 2016; 9:ra19. [PMID: 26884601 DOI: 10.1126/scisignal.aad1884] [Citation(s) in RCA: 438] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-β (TGF-β) is a major immunosuppressive cytokine that maintains immune homeostasis and prevents autoimmunity through its antiproliferative and anti-inflammatory properties in various immune cell types. We provide genetic, pharmacologic, and biochemical evidence that a critical target of TGF-β signaling in mouse and human natural killer (NK) cells is the serine and threonine kinase mTOR (mammalian target of rapamycin). Treatment of mouse or human NK cells with TGF-β in vitro blocked interleukin-15 (IL-15)-induced activation of mTOR. TGF-β and the mTOR inhibitor rapamycin both reduced the metabolic activity and proliferation of NK cells and reduced the abundances of various NK cell receptors and the cytotoxic activity of NK cells. In vivo, constitutive TGF-β signaling or depletion of mTOR arrested NK cell development, whereas deletion of the TGF-β receptor subunit TGF-βRII enhanced mTOR activity and the cytotoxic activity of the NK cells in response to IL-15. Suppression of TGF-β signaling in NK cells did not affect either NK cell development or homeostasis; however, it enhanced the ability of NK cells to limit metastases in two different tumor models in mice. Together, these results suggest that the kinase mTOR is a crucial signaling integrator of pro- and anti-inflammatory cytokines in NK cells. Moreover, we propose that boosting the metabolic activity of antitumor lymphocytes could be an effective strategy to promote immune-mediated tumor suppression.
Collapse
Affiliation(s)
- Sébastien Viel
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France. Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite 69310, France
| | - Antoine Marçais
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France
| | - Fernando Souza-Fonseca Guimaraes
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia. School of Medicine, The University of Queensland, Herston, Queensland 4006, Australia
| | - Roisin Loftus
- School of Biochemistry and Immunology and School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Jessica Rabilloud
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France
| | - Morgan Grau
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France
| | - Sophie Degouve
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France
| | - Sophia Djebali
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France
| | - Amélien Sanlaville
- Immunology Virology and Inflammation Department, INSERM U1052, CNRS 5286 Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France
| | - Emily Charrier
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France. Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite 69310, France
| | - Jacques Bienvenu
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France. Laboratoire d'Immunologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre-Bénite 69310, France
| | - Julien C Marie
- Immunology Virology and Inflammation Department, INSERM U1052, CNRS 5286 Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France. TGF-beta and immunoregulation group, DKFZ, Heidelberg 69121, Germany
| | - Christophe Caux
- Immunology Virology and Inflammation Department, INSERM U1052, CNRS 5286 Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France
| | - Jacqueline Marvel
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France
| | - Liam Town
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Laurent Bartholin
- Immunology Virology and Inflammation Department, INSERM U1052, CNRS 5286 Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France
| | - David Finlay
- School of Biochemistry and Immunology and School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia. School of Medicine, The University of Queensland, Herston, Queensland 4006, Australia.
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie (CIRI), 69007 Lyon, France. INSERM U1111, 69007 Lyon, France. Ecole Normale Supérieure de Lyon, 69007 Lyon, France. Université Lyon 1, 69007 Lyon, France. CNRS, UMR5308, 69007 Lyon, France.
| |
Collapse
|
14
|
Warsinske HC, Ashley SL, Linderman JJ, Moore BB, Kirschner DE. Identifying Mechanisms of Homeostatic Signaling in Fibroblast Differentiation. Bull Math Biol 2015; 77:1556-82. [PMID: 26384829 DOI: 10.1007/s11538-015-0096-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/21/2015] [Indexed: 10/23/2022]
Abstract
Fibroblasts play an important role in the wound-healing process by generating extracellular matrix (ECM) and undergoing differentiation into myofibroblasts, but these cells can also be involved in pathologic remodeling of tissue. Nascent ECM provides a substrate for re-epithelialization to occur, restoring damaged tissue to a functional state. Dysregulation of this process can result in fibrosis--stiffening and scarring of the tissue. Current treatments cannot halt or reverse this process. The molecular mechanisms underlying fibrotic dysregulation are poorly understood, providing an untapped pool of potential therapeutic targets. Transforming growth factor-β (TGF-β) and adhesion signaling are involved in inducing fibroblast differentiation into α-smooth muscle actin (αSMA) expressing myofibroblasts, while prostaglandin E₂ (PGE₂) has been shown to antagonize TGF-β signaling; however, the temporal and mechanistic details of this relationship have not yet been fully characterized. We measured αSMA, a marker of fibroblast to myofibroblast differentiation, as a function of: TGF-β1 receptor-ligand complex internalization, PGE₂ binding, and adhesion signaling and developed a mathematical model capturing the molecular mechanisms of fibroblast differentiation. Using our model, we predict the following: Periodic dosing with PGE₂ temporarily renders fibroblasts incapable of differentiation and refractory to additional TGF-β1 stimulation; conversely, periodic dosing with TGF-β1 in the presence of PGE₂ induces a reduced signal response that can be further inhibited by the addition of more PGE₂. Controlled fibroblast differentiation is necessary for effective wound healing; however, excessive accumulation of αSMA-expressing myofibroblasts can result in fibrosis. Homeostasis of αSMA in our model requires a balance of positive and negative regulatory signals. Sensitivity analysis predicts that PGE₂ availability, TGF-β1 availability, and the rate of TGF-β1 receptor recycling each highly influence the rates of αSMA production. With this model, we are able to demonstrate that regulation of both TGF-β1 and PGE₂ signaling levels is essential for preventing fibroblast dysregulation.
Collapse
Affiliation(s)
- Hayley C Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shanna L Ashley
- Immunology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | | | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Bone marrow Schwann cells induce hematopoietic stem cell hibernation. Int J Hematol 2014; 99:695-8. [PMID: 24817152 DOI: 10.1007/s12185-014-1588-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/25/2014] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem cells (HSCs) are clonogenic cells capable of both self-renewal and multilineage differentiation. In adult mouse bone marrow (BM), most HSCs remain in the non-dividing G0-phase of cell cycle, in close contact with supporting cells known as the HSC "niche". In the present study, we focused on signaling mechanisms that regulate stem cell dormancy in the BM niche. We show that TGF-β type II receptor deficiency causes reduced phosphorylation of Smad2/3 and impairs long-term repopulating activity in HSCs, suggesting a significant role for TGF-β/Smad signaling in hematopoiesis. Furthermore, we aimed at defining the candidate BM niche responsible for homeostasis of hematopoiesis, and revealed that non-myelinating Schwann cells sustain HSC hibernation by converting TGF-β from its latent to its active form.
Collapse
|
16
|
Śledzińska A, Hemmers S, Mair F, Gorka O, Ruland J, Fairbairn L, Nissler A, Müller W, Waisman A, Becher B, Buch T. TGF-β signalling is required for CD4⁺ T cell homeostasis but dispensable for regulatory T cell function. PLoS Biol 2013; 11:e1001674. [PMID: 24115907 PMCID: PMC3792861 DOI: 10.1371/journal.pbio.1001674] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 08/23/2013] [Indexed: 12/16/2022] Open
Abstract
Signalling by the cytokine TGF-β regulates mature CD4+ T cell populations but is not involved in the survival and function of regulatory T cells. TGF-β is widely held to be critical for the maintenance and function of regulatory T (Treg) cells and thus peripheral tolerance. This is highlighted by constitutive ablation of TGF-β receptor (TR) during thymic development in mice, which leads to a lethal autoimmune syndrome. Here we describe that TGF-β–driven peripheral tolerance is not regulated by TGF-β signalling on mature CD4+ T cells. Inducible TR2 ablation specifically on CD4+ T cells did not result in a lethal autoinflammation. Transfer of these TR2-deficient CD4+ T cells to lymphopenic recipients resulted in colitis, but not overt autoimmunity. In contrast, thymic ablation of TR2 in combination with lymphopenia led to lethal multi-organ inflammation. Interestingly, deletion of TR2 on mature CD4+ T cells does not result in the collapse of the Treg cell population as observed in constitutive models. Instead, a pronounced enlargement of both regulatory and effector memory T cell pools was observed. This expansion is cell-intrinsic and seems to be caused by increased T cell receptor sensitivity independently of common gamma chain-dependent cytokine signals. The expression of Foxp3 and other regulatory T cells markers was not dependent on TGF-β signalling and the TR2–deficient Treg cells retained their suppressive function both in vitro and in vivo. In summary, absence of TGF-β signalling on mature CD4+ T cells is not responsible for breakdown of peripheral tolerance, but rather controls homeostasis of mature T cells in adult mice. TGF-β is a cytokine thought to be critical for the maintenance and function of tolerance in the immune system. In many studies the disruption of TGF-β signalling in CD4+ T cells (a type of white blood cell that coordinates immune responses) has resulted in autoimmune syndromes. We show here that the induced removal of this cytokine's receptor from these specialised blood cells results in an astonishingly mild outcome. Contrary to expectations, the number of regulatory T cells is actually increased, and we find that these cells are not dependent on TGF-β signalling. We also show that removal of the receptor from mature CD4+ T cells does not lead to lethal autoinflammation; only when we removed the receptor during development of the cells did we see the characteristic lethal multi-organ inflammation reported previously in constitutive models of TGF-β receptor ablation. In summary, our findings indicate that although TGF-β regulates maintenance of mature CD4+ T cells, its signals are dispensable for immune tolerance within this cell population.
Collapse
Affiliation(s)
- Anna Śledzińska
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Saskia Hemmers
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Florian Mair
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Oliver Gorka
- Clinical Chemistry, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Jürgen Ruland
- Clinical Chemistry, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Lynsey Fairbairn
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Germany
| | - Anja Nissler
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Werner Müller
- Department of Experimental Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Ari Waisman
- Institute for Genetics, University of Cologne, Cologne, Germany
- Institute for Molecular Medicine, University Medical Center of the Johannes-Gutenberg University of Mainz, Mainz, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- * E-mail: (TB); (BB)
| | - Thorsten Buch
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Institute for Genetics, University of Cologne, Cologne, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Germany
- * E-mail: (TB); (BB)
| |
Collapse
|
17
|
Szondy Z, Garabuczi É, Tóth K, Kiss B, Köröskényi K. Thymocyte death by neglect: Contribution of engulfing macrophages. Eur J Immunol 2012; 42:1662-7. [DOI: 10.1002/eji.201142338] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
18
|
Gong D, Shi W, Yi SJ, Chen H, Groffen J, Heisterkamp N. TGFβ signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol 2012; 13:31. [PMID: 22703233 PMCID: PMC3406960 DOI: 10.1186/1471-2172-13-31] [Citation(s) in RCA: 295] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Accepted: 06/15/2012] [Indexed: 01/13/2023] Open
Abstract
Background Upon stimulation with different cytokines, macrophages can undergo classical or alternative activation to become M1 or M2 macrophages. Alternatively activated (or M2) macrophages are defined by their expression of specific gene products and play an important role in containing inflammation, removing apoptotic cells and repairing tissue damage. Whereas it is well-established that IL-4 can drive alternative activation, if lack of TGFβ signaling at physiological levels affects M2 polarization has not been addressed. Results Vav1-Cre x TβRIIfx/fx mice, lacking TβRII function in hematopoietic cells, exhibited uncontrolled pulmonary inflammation and developed a lethal autoimmune syndrome at young age. This was accompanied by significantly increased numbers of splenic neutrophils and T cells as well as elevated hepatic macrophage infiltration and bone marrow monocyte counts. TβRII-/- CD4+ and CD8+ T-cells in the lymph nodes and spleen expressed increased cell surface CD44, and CD69 was also higher on CD4+ lymph node T-cells. Loss of TβRII in bone marrow-derived macrophages (BMDMs) did not affect the ability of these cells to perform efferocytosis. However, these cells were defective in basal and IL-4-induced arg1 mRNA and Arginase-1 protein production. Moreover, the transcription of genes that are typically upregulated in M2-polarized macrophages, such as ym1, mcr2 and mgl2, was also decreased in peritoneal macrophages and IL-4-stimulated TβRII-/- BMDMs. We found that cell surface and mRNA expression of Galectin-3, which also regulates M2 macrophage polarization, was lower in TβRII-/- BMDMs. Very interestingly, the impaired ability of these null mutant BMDMs to differentiate into IL-4 polarized macrophages was Stat6- and Smad3-independent, but correlated with reduced levels of phospho-Akt and β-catenin. Conclusions Our results establish a novel biological role for TGFβ signaling in controlling expression of genes characteristic for alternatively activated macrophages. We speculate that lack of TβRII signaling reduces the anti-inflammatory M2 phenotype of macrophages because of reduced expression of these products. This would cause defects in the ability of the M2 macrophages to negatively regulate other immune cells such as T-cells in the lung, possibly explaining the systemic inflammation observed in Vav1-Cre x TβRIIfx/fx mice.
Collapse
Affiliation(s)
- Dapeng Gong
- Division of Hematology/Oncology, Children's Hospital Los Angeles, 4650 Sunset Boulevard, Los Angeles, CA 90027, USA
| | | | | | | | | | | |
Collapse
|
19
|
TGF-β signaling to T cells inhibits autoimmunity during lymphopenia-driven proliferation. Nat Immunol 2012; 13:667-73. [PMID: 22634866 PMCID: PMC3380154 DOI: 10.1038/ni.2319] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 04/24/2012] [Indexed: 12/13/2022]
Abstract
T cell specific deletion of the Transforming growth factor-β (TGF-β) receptor mediated by CD4-cre leads to early onset lethal autoimmune disease that cannot be controlled by regulatory T cells. However, when we delete the receptor using distal Lck (dLck) promoter driven cre, adult mice in which the majority of peripheral CD4+ and CD8+ T cells lacked the TGF-β receptor, showed no signs of autoimmunity. Due to their heightened response to weak T cell receptor stimuli, when transferred into lymphopenic recipients, naive TGF-β unresponsive T cells exhibited dramatically enhanced proliferation, effector differentiation, and induced lymphoproliferative disease. We propose that TGF-β signaling controls self-reactivity of peripheral T cells but in the absence of TGF-β signals, an added trigger, such as lymphopenia, is required to drive overt autoimmune disease.
Collapse
|
20
|
Loss of T cell microRNA provides systemic protection against autoimmune pathology in mice. J Autoimmun 2012; 38:39-48. [DOI: 10.1016/j.jaut.2011.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 12/08/2011] [Accepted: 12/08/2011] [Indexed: 11/19/2022]
|
21
|
Yamazaki S, Ema H, Karlsson G, Yamaguchi T, Miyoshi H, Shioda S, Taketo MM, Karlsson S, Iwama A, Nakauchi H. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 2012; 147:1146-58. [PMID: 22118468 DOI: 10.1016/j.cell.2011.09.053] [Citation(s) in RCA: 561] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 04/12/2011] [Accepted: 09/06/2011] [Indexed: 01/02/2023]
Abstract
Hematopoietic stem cells (HSCs) reside and self-renew in the bone marrow (BM) niche. Overall, the signaling that regulates stem cell dormancy in the HSC niche remains controversial. Here, we demonstrate that TGF-β type II receptor-deficient HSCs show low-level Smad activation and impaired long-term repopulating activity, underlining the critical role of TGF-β/Smad signaling in HSC maintenance. TGF-β is produced as a latent form by a variety of cells, so we searched for those that express activator molecules for latent TGF-β. Nonmyelinating Schwann cells in BM proved responsible for activation. These glial cells ensheathed autonomic nerves, expressed HSC niche factor genes, and were in contact with a substantial proportion of HSCs. Autonomic nerve denervation reduced the number of these active TGF-β-producing cells and led to rapid loss of HSCs from BM. We propose that glial cells are components of a BM niche and maintain HSC hibernation by regulating activation of latent TGF-β.
Collapse
Affiliation(s)
- Satoshi Yamazaki
- Japan Science and Technology Agency, ERATO, Chiyoda-ku, Tokyo 102-0075, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Fogle JE, Mexas AM, Tompkins WA, Tompkins MB. CD4(+)CD25(+) T regulatory cells inhibit CD8(+) IFN-gamma production during acute and chronic FIV infection utilizing a membrane TGF-beta-dependent mechanism. AIDS Res Hum Retroviruses 2010; 26:201-16. [PMID: 20156102 DOI: 10.1089/aid.2009.0162] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
CD8(+) lymphocytes are critical to the control and elimination of viral pathogens. Impaired CD8(+) responses are well recognized in lentiviral infections; however, the mechanisms underlying CD8(+) impairment remain elusive. Using the feline immunodeficiency virus (FIV) model for human AIDS, we reported previously that CD4(+)CD25(+) Treg cells in both the acute and long-term, asymptomatic phase of infection are constitutively activated and suppress CD4(+)CD25(-) T cell responses. In the current study, we have demonstrated that CD4(+)CD25(+) Treg cells suppress CD8(+) responses to immune stimulation during both the acute and chronic, asymptomatic phase of FIV infection and that the mechanism of suppression may be mediated by membrane-associated TGF-beta (mTGF-beta) on CD4(+)CD25(+) lymphocytes. Depletion of CD4(+)CD25(+) lymphocytes from lymph node suspensions significantly enhanced production of IFN-gamma during the acute phase of infection and coculture of CD8(+) lymphocytes with CD4(+)CD25(+) lymphocytes resulted in suppression of CD8(+) IFN-gamma during both the acute and chronic stages of infection. FACS analysis indicated that there was TGF-betaRII upregulation on CD8(+) cells from FIV(+) cats during the acute and chronic stage of infection. In addition, there was upregulation of mTGF-beta on the CD4(+)CD25(+) subset in chronically infected cats. In support of activation of the TGF-beta signaling pathway, Western blotting showed Smad 2 phosphorylation in CD8(+) targets following CD4(+)CD25(+)/CD8(+) coculture. These results demonstrate the suppressive effect CD4(+)CD25(+) Treg cells have on the CD8(+) immune response during the acute and chronic stages of FIV infection and suggest that the mechanism of suppression may be mediated by mTGF-beta.
Collapse
Affiliation(s)
- Jonathan E. Fogle
- Immunology Program, Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina 27606
| | - Angela M. Mexas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Wayne A. Tompkins
- Immunology Program, Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina 27606
| | - Mary B. Tompkins
- Immunology Program, Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina 27606
| |
Collapse
|
23
|
Azhar M, Yin M, Bommireddy R, Duffy JJ, Yang J, Pawlowski SA, Boivin GP, Engle SJ, Sanford LP, Grisham C, Singh RR, Babcock GF, Doetschman T. Generation of mice with a conditional allele for transforming growth factor beta 1 gene. Genesis 2009; 47:423-31. [PMID: 19415629 DOI: 10.1002/dvg.20516] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Transforming growth factor beta1 (TGFbeta1) is a multifunctional growth factor involved in wound healing, tissue fibrosis, and in the pathogenesis of many syndromic diseases (e.g., Marfan syndrome, Camurati-Engelmann disease) and muscular, neurological, ophthalmic, cardiovascular and immunological disorders, and cancer. Since the generation of Tgfb1 knockout mice, there has been extraordinary progress in understanding its physiological and pathophysiological function. Here, we report the generation of a conditional knockout allele for Tgfb1 in which its exon 6 is flanked with LoxP sites. As proof of principle, we crossed these mice to LckCre transgenic mice and specifically disrupted Tgfb1 in T cells. The results indicate that T-cell-produced TGFbeta1 is required for normal in vivo regulation of peripheral T-cell activation, maintenance of T-cell homeostasis, and suppression of autoimmunity.
Collapse
Affiliation(s)
- Mohamad Azhar
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Denton CP, Khan K, Hoyles RK, Shiwen X, Leoni P, Chen Y, Eastwood M, Abraham DJ. Inducible Lineage-Specific Deletion of TβRII in Fibroblasts Defines a Pivotal Regulatory Role during Adult Skin Wound Healing. J Invest Dermatol 2009; 129:194-204. [DOI: 10.1038/jid.2008.171] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
25
|
Abstract
It is now apparent that naïve peripheral T cells are a dynamic population where active processes prevent inappropriate activation while supporting survival. The process of thymic education makes naïve peripheral T cells dependent on interactions with self-MHC for survival. However, as these signals can potentially result in inappropriate activation, various non-redundant, intrinsic negative regulatory molecules including Tob, Nfatc2, and Smad3 actively enforce T cell quiescence. Interactions among these pathways are only now coming to light and may include positive or negative crosstalk. In the case of positive crosstalk, self-MHC initiated signals and intrinsic negative regulatory factors may cooperate to dampen T cell activation and sustain peripheral tolerance in a binary fashion (on-off). In the case of negative crosstalk, self-MHC signals may promote survival through partial activation while intrinsic negative regulatory factors act as rheostats to restrain cell cycle entry and prevent T cells from crossing a threshold that would break tolerance.
Collapse
Affiliation(s)
- Jaime F Modiano
- Integrated Department of Immunology, University of Colorado-Denver, Denver, CO, USA.
| | | | | |
Collapse
|
26
|
Abstract
A functional adaptive immune system depends on a diverse and self-tolerant population of T lymphocytes that are generated in the thymus and maintained in the peripheral lymphoid organs. Recent studies have defined the cytokine transforming growth factor-beta (TGF-beta) as a critical regulator of thymic T cell development as well as a crucial player in peripheral T cell homeostasis, tolerance to self antigens, and T cell differentiation during the immune response. The unique mechanism of TGF-beta activation and the plasticity of TGF-beta signaling create a stage for TGF-beta to integrate signals from multiple cell types and environmental cues to regulate T cells.
Collapse
|
27
|
Histochemical and molecular overview of the thymus as site for T-cells development. ACTA ACUST UNITED AC 2008; 43:73-120. [PMID: 18555891 DOI: 10.1016/j.proghi.2008.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 03/11/2008] [Indexed: 12/19/2022]
Abstract
The thymus represents the primary site for T cell lymphopoiesis, providing a coordinated set for critical factors to induce and support lineage commitment, differentiation and survival of thymus-seeding cells. One irrefutable fact is that the presence of non-lymphoid cells through the thymic parenchyma serves to provide coordinated migration and differentiation of T lymphocytes. Moreover, the link between foetal development and normal anatomy has been stressed in this review. Regarding thymic embryology, its epithelium is derived from the embryonic endodermal layer, with possible contributions from the ectoderm. A series of differentiating steps is essential, each of which must be completed in order to provide the optimum environment for thymic development and function. The second part of this article is focused on thymic T-cell development and differentiation, which is a stepwise process, mediated by a variety of stromal cells in different regions of the organ. It depends strongly on the thymic microenvironment, a cellular network formed by epithelial cells, macrophages, dendritic cells and fibroblasts, that provide the combination of cellular interactions, cytokines and chemokines to induce thymocyte precursors for the generation of functional T cells. The mediators of this process are not well defined but it has been demonstrated that some interactions are under neuroendocrine control. Moreover, some studies pointed out that reciprocal signals from developing T cells also are essential for establishment and maintenance of the thymic microenvironment. Finally, we have also highlighted the heterogeneity of the lymphoid, non-lymphoid components and the multi-phasic steps of thymic differentiation. In conclusion, this review contributes to an understanding of the complex mechanisms in which the foetal and postnatal thymus is involved. This could be a prerequisite for developing new therapies specifically aimed to overcome immunological defects, linked or not-linked to aging.
Collapse
|
28
|
Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood 2007; 111:3931-40. [PMID: 18160670 DOI: 10.1182/blood-2007-08-107748] [Citation(s) in RCA: 341] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
An acquired somatic mutation in the JAK2 gene (JAK2-V617F) is present in the majority of patients with myeloproliferative disorders (MPDs). Several phenotypic manifestations (polycythemia vera [PV], essential thrombocythemia [ET], and primary myelofibrosis) can be associated with the same mutation. We generated JAK2-V617F transgenic mice using a human JAK2 gene with the sequences encoding the kinase domain placed in the inverse orientation and flanked by antiparallel loxP sites. Crossing mice of one transgenic line (FF1) with transgenic mice expressing Cre-recombinase under the control of the hematopoiesis specific Vav promoter led to expression of JAK2-V617F that was lower than the endogenous wild-type Jak2. These mice developed a phenotype resembling ET with strongly elevated platelet counts and moderate neutrophilia. Induction of the JAK2-V617F transgene with the interferon-inducible MxCre resulted in expression of JAK2-V617F approximately equal to wild-type Jak2 and a PV-like phenotype with increased hemoglobin, thrombocytosis, and neutrophilia. Higher levels of JAK2-V617F in mouse bone marrow by retroviral transduction caused a PV-like phenotype without thrombocytosis. These data are consistent with the hypothesis that the ratio of mutant to wild-type JAK2 is critical for the phenotypic manifestation. A similar correlation was also found in patients with MPD.
Collapse
|
29
|
Abstract
Like all hematopoietic cells, T lymphocytes are derived from bone-marrow-resident stem cells. However, whereas most blood lineages are generated within the marrow, the majority of T cell development occurs in a specialized organ, the thymus. This distinction underscores the unique capacity of the thymic microenvironment to support T lineage restriction and differentiation. Although the identity of many of the contributing thymus-derived signals is well established and rooted in highly conserved pathways involving Notch, morphogenetic, and protein tyrosine kinase signals, the manner in which the ensuing cascades are integrated to orchestrate the underlying processes of T cell development remains under investigation. This review focuses on the current definition of the early stages of T cell lymphopoiesis, with an emphasis on the nature of thymus-derived signals delivered to T cell progenitors that support the commitment and differentiation of T cells toward the alphabeta and gammadelta T cell lineages.
Collapse
Affiliation(s)
- Maria Ciofani
- Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
30
|
Pan D, Schomber T, Kalberer CP, Terracciano LM, Hafen K, Krenger W, Hao-Shen H, Deng C, Skoda RC. Normal erythropoiesis but severe polyposis and bleeding anemia in Smad4-deficient mice. Blood 2007; 110:3049-55. [PMID: 17638848 DOI: 10.1182/blood-2007-02-074393] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The tumor suppressor Smad4 mediates signaling by the transforming growth factor beta (TGF-beta) superfamily of ligands. Previous studies showed that several TGF-beta family members exert important functions in hematopoiesis. Here, we studied the role of Smad4 in adult murine hematopoiesis using the inducible Mx-Cre/loxP system. Mice with homozygous Smad4 deletion (Smad4(Delta/Delta)) developed severe anemia 6 to 8 weeks after induction (mean hemoglobin level 70 g/L). The anemia was not transplantable, as wild-type mice reconstituted with Smad4(Delta/Delta) bone marrow cells had normal peripheral blood counts. These mice did not develop an inflammatory disease typical for mice deficient in TGF-beta receptors I and II, suggesting that the suppression of inflammation by TGF-beta is Smad4 independent. The same results were obtained when Smad4 alleles were deleted selectively in hematopoietic cells using the VavCre transgenic mice. In contrast, lethally irradiated Smad4(Delta/Delta) mice that received wild-type bone marrow cells developed anemia similar to Smad4(Delta/Delta) mice that did not receive a transplant. Liver iron stores were decreased and blood was present in stool, indicating that the anemia was due to blood loss. Multiple polyps in stomach and colon represent a likely source of the bleeding. We conclude that Smad4 is not required for adult erythropoiesis and that anemia is solely the consequence of blood loss.
Collapse
Affiliation(s)
- Dejing Pan
- Department of Research, Experimental Hematology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Melichar H, Kang J. Integrated morphogen signal inputs in gammadelta versus alphabeta T-cell differentiation. Immunol Rev 2007; 215:32-45. [PMID: 17291277 DOI: 10.1111/j.1600-065x.2006.00469.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Morphogens, a class of secreted proteins that regulate gene expression in a concentration-dependent manner, are responsible for directing nearly all lineage fate choices during embryogenesis. In the thymus, morphogen signal pathways consisting of WNT, Hedgehog, and the transforming growth factor-beta superfamily are active and have been implicated in various developmental processes including proliferation, survival, and differentiation of maturing thymocytes. Intriguingly, it has been inferred that some of these morphogen signal pathways differentially affect gammadelta and alphabeta T-cell development or maintenance, but their role in T-cell lineage commitment has not been directly probed. We have recently identified a modulator of morphogen signaling that significantly influences binary gammadelta versus alphabeta T-cell lineage diversification. In this review, we summarize functions of morphogens in the thymus and provide a highly speculative model of integrated morphogen signals, potentially directing the gammadelta versus alphabeta T-cell fate determination process.
Collapse
Affiliation(s)
- Heather Melichar
- Department of Pathology University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | |
Collapse
|
32
|
Licona-Limón P, Soldevila G. The role of TGF-beta superfamily during T cell development: new insights. Immunol Lett 2007; 109:1-12. [PMID: 17287030 DOI: 10.1016/j.imlet.2006.12.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Revised: 12/21/2006] [Accepted: 12/23/2006] [Indexed: 10/23/2022]
Abstract
Members of the transforming growth factor beta (TGF-beta) superfamily are soluble factors that regulate a variety of functional responses including proliferation, differentiation, apoptosis and cell cycle, among others, depending not only on the cell type and its differentiation state, but also on the milieu of cytokines present. All three members of this superfamily: TGF-betas, bone morphogenetic proteins (BMPs) and Activins, have been shown to be expressed in the thymus suggesting their potential role as regulators of the T lymphocyte differentiation process. Although initial reports described the role of TGF-beta in controlling specific checkpoints during thymocyte development, recent data has provided new evidence on the role of BMPs and Activins in this process. This review provides new insights on the function of members of the TGF-beta superfamily at different stages of thymocyte development.
Collapse
Affiliation(s)
- P Licona-Limón
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, México DF-04510, Mexico
| | | |
Collapse
|
33
|
Li MO, Sanjabi S, Flavell RA. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 2006; 25:455-71. [PMID: 16973386 DOI: 10.1016/j.immuni.2006.07.011] [Citation(s) in RCA: 632] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 06/22/2006] [Accepted: 07/12/2006] [Indexed: 10/24/2022]
Abstract
The role of transforming growth factor-beta (TGF-beta) in inhibiting T cell functions has been studied with dominant-negative TGF-beta receptor transgenic models; however, the full impact of TGF-beta signaling on T cells and the mechanisms by which TGF-beta signals remain poorly understood. Here we show that mice with T cell-specific deletion of TGF-beta receptor II developed lethal inflammation associated with T cell activation and differentiation. In addition, TGF-beta signaling positively regulated T cell development and homeostasis. Development of CD8+ T cells and NKT cells, maintenance of peripheral Foxp3-expressing regulatory T cells, and survival of CD4+ T cells all depended on TGF-beta signaling. Both T helper 1 (Th1) differentiation and survival of activated CD4+ T cells required T-bet, the TGF-beta-regulated transcription factor, which controlled CD122 expression and IL-15 signaling in Th1 cells. This study reveals pleiotropic functions of TGF-beta signaling in T cells that may ensure a diverse and self-tolerant T cell repertoire in vivo.
Collapse
Affiliation(s)
- Ming O Li
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
34
|
Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-beta receptor. Immunity 2006; 25:441-54. [PMID: 16973387 DOI: 10.1016/j.immuni.2006.07.012] [Citation(s) in RCA: 368] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2006] [Revised: 06/21/2006] [Accepted: 07/12/2006] [Indexed: 12/31/2022]
Abstract
Transforming growth factor-beta (TGF-beta) has been implicated in the control of differentiation and proliferation of multiple cell types. However, a role for TGF-beta in the control of immune homeostasis is not fully understood because of its pleiotropic action. Here we report that complete ablation of the TGF-beta signaling in T cells engendered aggressive early-onset, multiorgan, autoimmune-associated lesions with 100% mortality. Peripheral CD4+ and CD8+ T cells with TGF-beta-receptor II (TGF-betaRII) deficiency activated cytolytic and T helper 1 (Th1) differentiation program in a cell-intrinsic T cell receptor (TCR)-specific fashion. Furthermore, TGF-betaRII deficiency blocked the development of canonical CD1d-restricted NKT cells. Instead, it facilitated generation of a highly pathogenic T cell subset exhibiting multiple hallmarks of NK cells and sharply elevated amounts of FasL, perforin, granzymes, and interferon-gamma. Thus, TGF-beta signaling in peripheral T cells is crucial in restraining TCR activation-dependent Th1, cytotoxic, and NK cell-like differentiation program which, when left unchecked, leads to rapidly progressing fatal autoimmunity.
Collapse
Affiliation(s)
- Julien C Marie
- Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
35
|
Zhang L, Yi H, Xia XP, Zhao Y. Transforming growth factor-beta: an important role in CD4+CD25+ regulatory T cells and immune tolerance. Autoimmunity 2006; 39:269-76. [PMID: 16891215 DOI: 10.1080/08916930600753903] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The transforming growth factor-beta (TGF-beta) protein family is highly evolutionarily conserved and they have been implicated in many biological processes. Also, TGF-beta can exert pivotal functions in the immune system. It is widely accepted that regulatory T cells (Treg cells) play an important role in the maintenance of the immune homeostasis, but the underlying molecular mechanisms through which they can gain and/or perform suppressive functions in an active way remains to be defined. Though the engagement of TGF-beta in the Treg cells has been discounted for a period of time, an emerging body of data has established a close link between Treg cells and TGF-beta, as TGF-beta has been demonstrated to induce the expression of Foxp3, which acts as a master regulator for the development and function of Treg cells. We will, herein, focus on the crucial role of TGF-beta signaling in Treg cell biology and summarize the current studies regarding TGF-beta in the generation and function of CD4+CD25+Treg cells both in vivo and in vitro.
Collapse
Affiliation(s)
- Lianjun Zhang
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | | | | | | |
Collapse
|