1
|
Chen YT, Chang YM, Chen YL, Su YH, Liao CC, Chiang TH, Chen WY, Su YC. N-ethyl-N-nitrosourea (ENU)-induced C-terminal truncation of Runx3 results in autoimmune colitis associated with Th17/Treg imbalance. Immunol Lett 2024; 268:106869. [PMID: 38788802 DOI: 10.1016/j.imlet.2024.106869] [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: 06/05/2023] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic and progressive inflammatory intestinal disease that affects people around the world. The primary cause of IBD is an imbalance in the host immune response to intestinal flora. Several human genes, including IL10, STAT3, IRGM, ATG16L1, NOD2 and RUNX3, are associated with inappropriate immune responses in IBD. It has been reported that homozygous Runx3-knockout (ko) mice spontaneously develop colitis. However, the high mortality rate in these mice within the first two weeks makes it challenging to study the role of Runx3 in colitis. To address this issue, a spontaneous colitis (SC) mouse model carrying a C-terminal truncated form of Runx3 with Tyr319stop point mutation has been generated. After weaning, SC mice developed spontaneous diarrhea and exhibited prominent enlargement of the colon, accompanied by severe inflammatory cell infiltration. Results of immunofluorescence staining showed massive CD4+ T cell infiltration in the inflammatory colon of SC mice. Colonic IL-17A mRNA expression and serum IL-17A level were increased in SC mice. CD4+ T cells from SC mice produced stronger IL-17A than those from wildtype mice in Th17-skewing conditions in vitro. In addition, the percentages of Foxp3+ Treg cells as well as the RORγt+Foxp3+ Treg subset, known for its role in suppressing Th17 response in the gut, were notably lower in colon lamina propria of SC mice than those in WT mice. Furthermore, transfer of total CD4+ T cells from SC mice, but not from wildtype mice, into Rag1-ko host mice resulted in severe autoimmune colitis. In conclusion, the C-terminal truncated Runx3 caused autoimmune colitis associated with Th17/Treg imbalance. The SC mouse model is a feasible approach to investigate the effect of immune response on spontaneous colitis.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
| | - Yi-Mei Chang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Yu-Ling Chen
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Yu-Hsuan Su
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Chia-Chi Liao
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Tien-Huang Chiang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Wen-Yu Chen
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Yu-Chia Su
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC.
| |
Collapse
|
2
|
Lutter L, Ter Linde JJM, Brand EC, Hoytema van Konijnenburg DP, Roosenboom B, Horjus Talabur-Horje C, Oldenburg B, van Wijk F. Compartment-driven imprinting of intestinal CD4 T cells in inflammatory bowel disease and homeostasis. Clin Exp Immunol 2023; 214:235-248. [PMID: 37565620 PMCID: PMC10719222 DOI: 10.1093/cei/uxad095] [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: 03/19/2023] [Revised: 07/16/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023] Open
Abstract
The mucosal immune system is implicated in the etiology and progression of inflammatory bowel diseases. The lamina propria and epithelium of the gut mucosa constitute two separate compartments, containing distinct T-cell populations. Human CD4 T-cell programming and regulation of lamina propria and epithelium CD4 T cells, especially during inflammation, remain incompletely understood. We performed flow cytometry, bulk, and single-cell RNA-sequencing to profile ileal lamina propria and intraepithelial CD4 T cells (CD4CD8αα, regulatory T cells (Tregs), CD69- and CD69high Trm T cells) in controls and Crohn's disease (CD) patients (paired non-inflamed and inflamed). Inflammation results in alterations of the CD4 T-cell population with a pronounced increase in Tregs and migrating/infiltrating cells. On a transcriptional level, inflammation within the epithelium induced T-cell activation, increased IFNγ responses, and an effector Treg profile. Conversely, few transcriptional changes within the lamina propria were observed. Key regulators including the chromatin remodelers ARID4B and SATB1 were found to drive compartment-specific transcriptional programming of CD4 T(reg) cells. In summary, inflammation in CD patients primarily induces changes within the epithelium and not the lamina propria. Additionally, there is compartment-specific CD4 T-cell imprinting, driven by shared regulators, between the lamina propria and the epithelium. The main consequence of intraepithelial adaptation, irrespective of inflammation, seems to be an overall dampening of broad (pro-inflammatory) responses and tight regulation of lifespan. These data suggest differential regulation of the lamina propria and epithelium, with a specific regulatory role in the inflamed epithelium.
Collapse
Affiliation(s)
- Lisanne Lutter
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - José J M Ter Linde
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Eelco C Brand
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - David P Hoytema van Konijnenburg
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Britt Roosenboom
- Department of Gastroenterology and Hepatology, Rijnstate Hospital, Arnhem, The Netherlands
| | | | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Femke van Wijk
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
3
|
Selven H, Busund LTR, Andersen S, Pedersen MI, Lombardi APG, Kilvaer TK. High Expression of IRS-1, RUNX3 and SMAD4 Are Positive Prognostic Factors in Stage I-III Colon Cancer. Cancers (Basel) 2023; 15:cancers15051448. [PMID: 36900240 PMCID: PMC10000923 DOI: 10.3390/cancers15051448] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Colon cancer is a common malignancy and a major contributor to human morbidity and mortality. In this study, we explore the expression and prognostic impact of IRS-1, IRS-2, RUNx3, and SMAD4 in colon cancer. Furthermore, we elucidate their correlations with miRs 126, 17-5p, and 20a-5p, which are identified as potential regulators of these proteins. Tumor tissue from 452 patients operated for stage I-III colon cancer was retrospectively collected and assembled into tissue microarrays. Biomarkers' expressions were examined by immunohistochemistry and analyzed using digital pathology. In univariate analyses, high expression levels of IRS1 in stromal cytoplasm, RUNX3 in tumor (nucleus and cytoplasm) and stroma (nucleus and cytoplasm), and SMAD4 in tumor (nucleus and cytoplasm) and stromal cytoplasm were related to increased disease-specific survival (DSS). In multivariate analyses, high expression of IRS1 in stromal cytoplasm, RUNX3 in tumor nucleus and stromal cytoplasm, and high expression of SMAD4 in tumor and stromal cytoplasm remained independent predictors of improved DSS. Surprisingly, with the exception of weak correlations (0.2 < r < 0.25) between miR-126 and SMAD4, the investigated markers were mostly uncorrelated with the miRs. However, weak to moderate/strong correlations (0.3 < r < 0.6) were observed between CD3 and CD8 positive lymphocyte density and stromal RUNX3 expression. High expression levels of IRS1, RUNX3, and SMAD4 are positive prognostic factors in stage I-III colon cancer. Furthermore, stromal expression of RUNX3 is associated with increased lymphocyte density, suggesting that RUNX3 is an important mediator during recruitment and activation of immune cells in colon cancer.
Collapse
Affiliation(s)
- Hallgeir Selven
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, 9038 Tromsø, Norway
| | - Lill-Tove Rasmussen Busund
- Department of Pathology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, 9038 Tromsø, Norway
| | - Sigve Andersen
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, 9038 Tromsø, Norway
| | - Mona Irene Pedersen
- Department of Clinical Medicine, UiT The Arctic University of Norway, 9038 Tromsø, Norway
| | | | - Thomas Karsten Kilvaer
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, 9038 Tromsø, Norway
- Correspondence: ; Tel.: +47-905-24-635
| |
Collapse
|
4
|
Liang Y, Ding H. Expression levels of RUNX3 and FGFR2 in peripheral blood of severe acute pancreatitis and their clinical significance. Ann Surg Treat Res 2023; 104:90-100. [PMID: 36816737 PMCID: PMC9929434 DOI: 10.4174/astr.2023.104.2.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 02/10/2023] Open
Abstract
Purpose Severe acute pancreatitis (SAP) is a life-threatening inflammatory syndrome of the pancreas. This study aimed to analyze the clinical significance of runt-associated transcription factor 3 (RUNX3) and fibroblast growth factor receptor 2 (FGFR2) expression alterations in SAP. Methods This study included 18 SAP patients in Wuzhong People's Hospital from November 2019 to December 2021 and 18 healthy controls. RUNX3 and FGFR2 expression levels were determined by RT-quantitative PCR. Correlations between RUNX3/FGFR2 and sex, age, etiology, CRP, procalcitonin, AST, LDH, BUN, Acute Physiology and Chronic Health Evaluation II (APACHE II), Ranson score, Bedside Index for Severity in Acute Pancreatitis (BISAP) score, sequential organ failure assessment (SOFA), and modified computed tomography severity index (MCTSI) score were analyzed. Diagnostic values of RUNX3 and FGFR2 in SAP were analyzed using the receiver-operating characteristic curve. The binding of RUNX3 to FGFR2 was analyzed by chromatin immunoprecipitation. Results RUNX3 and FGFR2 were downregulated in peripheral blood of SAP patients. RUNX3 and FGFR2 were negatively correlated with CRP, procalcitonin, AST, LDH, BUN, APACHE II score, Ranson score, BISAP score, SOFA score, and MCTSI score. Sensitivity and specificity of RUNX3 level of <0.9650 for SAP diagnosis were 88.89% and 72.22%, respectively. Sensitivity and specificity of FGFR2 level of <0.8950 for SAP diagnosis were 66.67% and 83.33%, respectively. RUNX3 was enriched in the FGFR2 promoter and was positively correlated with FGFR2. Conclusion RUNX3 and FGFR2 were downregulated in peripheral blood of SAP patients and served as candidate biomarkers for SAP diagnosis. RUNX3 bound to the FGFR2 promoter to promote FGFR2 transcription.
Collapse
Affiliation(s)
- Yongyong Liang
- Department of Critical Medicine, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Huaming Ding
- Department of Critical Medicine, Wuzhong People’s Hospital, Suzhou, China
| |
Collapse
|
5
|
Vaghari-Tabari M, Targhazeh N, Moein S, Qujeq D, Alemi F, Majidina M, Younesi S, Asemi Z, Yousefi B. From inflammatory bowel disease to colorectal cancer: what's the role of miRNAs? Cancer Cell Int 2022; 22:146. [PMID: 35410210 PMCID: PMC8996392 DOI: 10.1186/s12935-022-02557-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/21/2022] [Indexed: 12/27/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) is a chronic inflammatory disease with relapse and remission periods. Ulcerative colitis and Crohn's disease are two major forms of the disease. IBD imposes a lot of sufferings on the patient and has many consequences; however, the most important is the increased risk of colorectal cancer, especially in patients with Ulcerative colitis. This risk is increased with increasing the duration of disease, thus preventing the progression of IBD to cancer is very important. Therefore, it is necessary to know the details of events contributed to the progression of IBD to cancer. In recent years, the importance of miRNAs as small molecules with 20-22 nucleotides has been recognized in pathophysiology of many diseases, in which IBD and colorectal cancer have not been excluded. As a result, the effectiveness of these small molecules as therapeutic target is hopefully confirmed. This paper has reviewed the related studies and findings about the role of miRNAs in the course of events that promote the progression of IBD to colorectal carcinoma, as well as a review about the effectiveness of some of these miRNAs as therapeutic targets.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Targhazeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Forough Alemi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidina
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Simin Younesi
- Schoole of Health and Biomedical Sciences, RMIT University, Melborne, VIC, Australia
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Xydia M, Rahbari R, Ruggiero E, Macaulay I, Tarabichi M, Lohmayer R, Wilkening S, Michels T, Brown D, Vanuytven S, Mastitskaya S, Laidlaw S, Grabe N, Pritsch M, Fronza R, Hexel K, Schmitt S, Müller-Steinhardt M, Halama N, Domschke C, Schmidt M, von Kalle C, Schütz F, Voet T, Beckhove P. Common clonal origin of conventional T cells and induced regulatory T cells in breast cancer patients. Nat Commun 2021; 12:1119. [PMID: 33602930 PMCID: PMC7893042 DOI: 10.1038/s41467-021-21297-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Regulatory CD4+ T cells (Treg) prevent tumor clearance by conventional T cells (Tconv) comprising a major obstacle of cancer immune-surveillance. Hitherto, the mechanisms of Treg repertoire formation in human cancers remain largely unclear. Here, we analyze Treg clonal origin in breast cancer patients using T-Cell Receptor and single-cell transcriptome sequencing. While Treg in peripheral blood and breast tumors are clonally distinct, Tconv clones, including tumor-antigen reactive effectors (Teff), are detected in both compartments. Tumor-infiltrating CD4+ cells accumulate into distinct transcriptome clusters, including early activated Tconv, uncommitted Teff, Th1 Teff, suppressive Treg and pro-tumorigenic Treg. Trajectory analysis suggests early activated Tconv differentiation either into Th1 Teff or into suppressive and pro-tumorigenic Treg. Importantly, Tconv, activated Tconv and Treg share highly-expanded clones contributing up to 65% of intratumoral Treg. Here we show that Treg in human breast cancer may considerably stem from antigen-experienced Tconv converting into secondary induced Treg through intratumoral activation.
Collapse
Affiliation(s)
- Maria Xydia
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany.
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany.
| | - Raheleh Rahbari
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
| | - Eliana Ruggiero
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Iain Macaulay
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- Technical Development, Earlham Institute, Norwich, UK
| | - Maxime Tarabichi
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- The Francis Crick Institute, London, UK
| | - Robert Lohmayer
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany
- Institute for Theoretical Physics, University of Regensburg, Regensburg, Germany
| | - Stefan Wilkening
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Tillmann Michels
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany
| | - Daniel Brown
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Sebastiaan Vanuytven
- The Francis Crick Institute, London, UK
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Svetlana Mastitskaya
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Sean Laidlaw
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
| | - Niels Grabe
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Hamamatsu Tissue Imaging and Analysis Centre, BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Maria Pritsch
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany
| | - Raffaele Fronza
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Klaus Hexel
- Flow Cytometry Core Facility, German Cancer Research Centre, Heidelberg, Germany
| | - Steffen Schmitt
- Flow Cytometry Core Facility, German Cancer Research Centre, Heidelberg, Germany
| | - Michael Müller-Steinhardt
- German Red Cross (DRK Blood Donation Service in Baden-Württemberg-Hessen) and Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Niels Halama
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Hamamatsu Tissue Imaging and Analysis Centre, BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Christoph Domschke
- Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Manfred Schmidt
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Christof von Kalle
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
- Clinical Study Centre, Charité/BIH, Berlin, Germany
| | - Florian Schütz
- Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Thierry Voet
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Philipp Beckhove
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany.
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany.
| |
Collapse
|
7
|
Olguín JE, Medina-Andrade I, Rodríguez T, Rodríguez-Sosa M, Terrazas LI. Relevance of Regulatory T Cells during Colorectal Cancer Development. Cancers (Basel) 2020; 12:E1888. [PMID: 32674255 PMCID: PMC7409056 DOI: 10.3390/cancers12071888] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
In recent years, there has been a significant increase in the study of own and foreign human factors favoring the development of different types of cancer, including genetic and environmental ones. However, the fact that the immune response plays a fundamental role in the development of immunity and susceptibility to colorectal cancer (CRC) is much stronger. Among the many cell populations of the immune system that participate in restricting or favoring CRC development, regulatory T cells (Treg) play a major role in orchestrating immunomodulation during CRC. In this review, we established concrete evidence supporting the fact that Treg cells have an important role in the promotion of tumor development during CRC, mediating an increasing suppressive capacity which controls the effector immune response, and generating protection for tumors. Furthermore, Treg cells go through a process called "phenotypic plasticity", where they co-express transcription factors that promote an inflammatory profile. We reunited evidence that describes the interaction between the different effector populations of the immune response and its modulation by Treg cells adapted to the tumor microenvironment, including the mechanisms used by Treg cells to suppress the protective immune response, as well as the different subpopulations of Treg cells participating in tumor progression, generating susceptibility during CRC development. Finally, we discussed whether Treg cells might or might not be a therapeutic target for an effective reduction in the morbidity and mortality caused by CRC.
Collapse
Affiliation(s)
- Jonadab E. Olguín
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
- Unidad de Biomedicina, FES Iztacala, UNAM, Av. De los Barrios # 1, Tlalnepantla 54090, Mexico
| | - Itzel Medina-Andrade
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
- Unidad de Biomedicina, FES Iztacala, UNAM, Av. De los Barrios # 1, Tlalnepantla 54090, Mexico
| | - Tonathiu Rodríguez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
| | - Miriam Rodríguez-Sosa
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
| | - Luis I. Terrazas
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
- Unidad de Biomedicina, FES Iztacala, UNAM, Av. De los Barrios # 1, Tlalnepantla 54090, Mexico
| |
Collapse
|
8
|
Hantisteanu S, Dicken Y, Negreanu V, Goldenberg D, Brenner O, Leshkowitz D, Lotem J, Levanon D, Groner Y. Runx3 prevents spontaneous colitis by directing the differentiation of anti-inflammatory mononuclear phagocytes. PLoS One 2020; 15:e0233044. [PMID: 32453801 PMCID: PMC7250423 DOI: 10.1371/journal.pone.0233044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
Mice deficient in the transcription factor Runx3 develop a multitude of immune system defects, including early onset colitis. This paper demonstrates that Runx3 is expressed in colonic mononuclear phagocytes (MNP), including resident macrophages (RM) and dendritic cell subsets (cDC2). Runx3 deletion in MNP causes early onset colitis due to their impaired maturation. Mechanistically, the resulting MNP subset imbalance leads to up-regulation of pro-inflammatory genes as occurs in IL10R-deficient RM. In addition, RM and cDC2 display a marked decrease in expression of anti-inflammatory/TGF β-regulated genes and β-catenin signaling associated genes, respectively. MNP transcriptome and ChIP-seq data analysis suggest that a significant fraction of genes affected by Runx3 loss are direct Runx3 targets. Collectively, Runx3 imposes intestinal immune tolerance by regulating maturation of colonic anti-inflammatory MNP, befitting the identification of RUNX3 as a genome-wide associated risk gene for various immune-related diseases in humans, including gastrointestinal tract diseases such as Crohn’s disease and celiac.
Collapse
Affiliation(s)
- Shay Hantisteanu
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Dicken
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Varda Negreanu
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Dalia Goldenberg
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ori Brenner
- Veterinary Resources, The Weizmann Institute of Science, Rehovot, Israel
| | - Dena Leshkowitz
- Bioinformatics Unit, The Weizmann Institute of Science, Rehovot, Israel
| | - Joseph Lotem
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ditsa Levanon
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Groner
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
9
|
Mirlekar B. Co-expression of master transcription factors determines CD4 + T cell plasticity and functions in auto-inflammatory diseases. Immunol Lett 2020; 222:58-66. [PMID: 32220615 DOI: 10.1016/j.imlet.2020.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/05/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023]
Abstract
Master CD4+ T cell lineage determined transcription factors are found to be dysregulated in pathogenesis of autoimmune and inflammatory diseases. CD4+ T cells categorized into different lineages based on their functions, cell surface markers and master transcription factors those required for expression of lineage specific cytokines. T-bet, GATA3, RORγt and Foxp3 are major transcription regulators of Th1, Th2, Th17 and Treg cells respectively. Significant progress has been made in understanding expression of lineage specific master regulators that drives CD4+ T cell differentiation. It is known that each CD4+ T cell lineage express precise determined transcription factor and due to cross regulation between these factors the CD4+ T cells able to maintain thier specific phenotype. However, recent studies shows that the lineage specifying transcription factors frequently co-expressed. There is an emerging area of research revealing that the co-expression of lineage-specifying transcription factors alters the potential function and flexibility of subsets of CD4+ T cell, this in turn favors the autoimmune pathology. Here, we discuss similarities and differences between mutually co-expressed transcription factors in CD4+ T cell subsets and then recapitulates on cell type specific and dynamic balance between the lineage restricted transcription factors in determining plasticity of CD4+ T cell subsets. Furthermore, we discuss abnormal regulation of such transcription factors that establishes a pathogenic CD4+ T cell phenotype in autoimmune diseases and how this understanding will provide further insight into potential therapeutic development.
Collapse
Affiliation(s)
- Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC, 27514, USA.
| |
Collapse
|
10
|
Mukherjee T, Udupa VAV, Prakhar P, Chandra K, Chakravortty D, Balaji KN. Epidermal Growth Factor Receptor-Responsive Indoleamine 2,3-Dioxygenase Confers Immune Homeostasis During Shigella flexneri Infection. J Infect Dis 2020; 219:1841-1851. [PMID: 30615126 DOI: 10.1093/infdis/jiz009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/05/2019] [Indexed: 12/19/2022] Open
Abstract
The resolution of Shigella flexneri infection-associated hyperinflammation is crucial for host survival. Using in vitro and in vivo models of shigellosis, we found that S. flexneri induces the expression of indoleamine 2,3-dioxygenase 1 (IDO1) through the nucleotide oligomerization domain 2 (NOD2) and epidermal growth factor receptor (EGFR) signaling pathway. Congruently, abrogation of NOD2 or EGFR compromises the ability of S. flexneri to induce IDO1 expression. We observed that the loss of IDO1 function in vivo exacerbates shigellosis by skewing the inflammatory cytokine response, disrupting colon epithelial barrier integrity and consequently limiting the host life-span. Interestingly, administration of recombinant EGF rescued mice from IDO1 inhibition-driven aggravated shigellosis by restoring the cytokine balance and subsequently restricting bacterial growth. This is the first study that underscores the direct implication of the NOD2-EGFR axis in IDO1 production and its crucial homeostatic contributions during shigellosis. Together, these findings reveal EGF as a potential therapeutic intervention for infectious diseases.
Collapse
Affiliation(s)
- Tanushree Mukherjee
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Vibha A V Udupa
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Praveen Prakhar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Kasturi Chandra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
11
|
Yu X, Chen YA, Conejo-Garcia JR, Chung CH, Wang X. Estimation of immune cell content in tumor using single-cell RNA-seq reference data. BMC Cancer 2019; 19:715. [PMID: 31324168 PMCID: PMC6642583 DOI: 10.1186/s12885-019-5927-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/12/2019] [Indexed: 12/12/2022] Open
Abstract
Background The rapid development of single-cell RNA sequencing (scRNA-seq) provides unprecedented opportunities to study the tumor ecosystem that involves a heterogeneous mixture of cell types. However, the majority of previous and current studies related to translational and molecular oncology have only focused on the bulk tumor and there is a wealth of gene expression data accumulated with matched clinical outcomes. Results In this paper, we introduce a scheme for characterizing cell compositions from bulk tumor gene expression by integrating signatures learned from scRNA-seq data. We derived the reference expression matrix to each cell type based on cell subpopulations identified in head and neck cancer dataset. Our results suggest that scRNA-Seq-derived reference matrix outperforms the existing gene panel and reference matrix with respect to distinguishing immune cell subtypes. Conclusions Findings and resources created from this study enable future and secondary analysis of tumor RNA mixtures in head and neck cancer for a more accurate cellular deconvolution, and can facilitate the profiling of the immune infiltration in other solid tumors due to the expression homogeneity observed in immune cells. Electronic supplementary material The online version of this article (10.1186/s12885-019-5927-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Y Ann Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
12
|
Kyburz A, Fallegger A, Zhang X, Altobelli A, Artola-Boran M, Borbet T, Urban S, Paul P, Münz C, Floess S, Huehn J, Cover TL, Blaser MJ, Taube C, Müller A. Transmaternal Helicobacter pylori exposure reduces allergic airway inflammation in offspring through regulatory T cells. J Allergy Clin Immunol 2019; 143:1496-1512.e11. [PMID: 30240703 PMCID: PMC6592617 DOI: 10.1016/j.jaci.2018.07.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/29/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Transmaternal exposure to tobacco, microbes, nutrients, and other environmental factors shapes the fetal immune system through epigenetic processes. The gastric microbe Helicobacter pylori represents an ancestral constituent of the human microbiota that causes gastric disorders on the one hand and is inversely associated with allergies and chronic inflammatory conditions on the other. OBJECTIVE Here we investigate the consequences of transmaternal exposure to H pylori in utero and/or during lactation for susceptibility to viral and bacterial infection, predisposition to allergic airway inflammation, and development of immune cell populations in the lungs and lymphoid organs. METHODS We use experimental models of house dust mite- or ovalbumin-induced airway inflammation and influenza A virus or Citrobacter rodentium infection along with metagenomics analyses, multicolor flow cytometry, and bisulfite pyrosequencing, to study the effects of H pylori on allergy severity and immunologic and microbiome correlates thereof. RESULTS Perinatal exposure to H pylori extract or its immunomodulator vacuolating cytotoxin confers robust protective effects against allergic airway inflammation not only in first- but also second-generation offspring but does not increase susceptibility to viral or bacterial infection. Immune correlates of allergy protection include skewing of regulatory over effector T cells, expansion of regulatory T-cell subsets expressing CXCR3 or retinoic acid-related orphan receptor γt, and demethylation of the forkhead box P3 (FOXP3) locus. The composition and diversity of the gastrointestinal microbiota is measurably affected by perinatal H pylori exposure. CONCLUSION We conclude that exposure to H pylori has consequences not only for the carrier but also for subsequent generations that can be exploited for interventional purposes.
Collapse
Affiliation(s)
- Andreas Kyburz
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland
| | - Xiaozhou Zhang
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland
| | - Aleksandra Altobelli
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland
| | - Mariela Artola-Boran
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland
| | - Timothy Borbet
- Human Microbiome Program, New York University Langone Medical Center, New York, NY
| | - Sabine Urban
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland
| | - Petra Paul
- Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Timothy L Cover
- Vanderbilt University Medical Center and Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tenn
| | - Martin J Blaser
- Human Microbiome Program, New York University Langone Medical Center, New York, NY
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zurich, Switzerland.
| |
Collapse
|
13
|
Yao F, Yin L, Feng S, Wang X, Zhang A, Zhou H. Functional characterization of grass carp runt-related transcription factor 3: Involvement in TGF-β1-mediated c-Myc transcription in fish cells. FISH & SHELLFISH IMMUNOLOGY 2018; 82:130-135. [PMID: 30099141 DOI: 10.1016/j.fsi.2018.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 06/08/2023]
Abstract
In mammals, both runt-related transcription factor 3 (RUNX3) and c-Myc are the downstream effectors of transforming growth factor-β1 (TGF-β1) signaling to mediate various cellular responses. However, information of their interaction especially in fish is lacking. In the present study, grass carp (Ctenopharyngodon idella) runx3 (gcrunx3) cDNA was cloned and identified. Interestingly, opposing effects of recombinant grass carp TGF-β1 (rgcTGF-β1) on c-myc and runx3 mRNA expression were observed in grass carp periphery blood lymphocytes (PBLs). Parallelly, Runx3 protein levels were enhanced by rgcTGF-β1 in the cells. These findings prompted us to examine whether Runx3 can mediate the inhibition of TGF-β1 on c-myc expression in fish cells. In line with this, overexpression of grass carp Runx3 and Runx3 DN (a dominant-negative form of Runx3) in grass carp kidney cell line (CIK) cells decreased and increased c-myc transcript levels, respectively. Particularly, the regulation of Runx3 and Runx3 DN on c-myc mRNA expression was direct since they were presented in the nucleus without any stimulation. In addition, rgcTGF-β1 alone suppressed c-myc mRNA expression in CIK cells as in PBLs. Moreover, this inhibitory effect was also observed when grass carp Runx3 and Runx3 DN were overexpressed. These results strengthened the role of TGF-β1 signaling in controlling c-myc transcription. Taken together, TGF-β1-mediated c-myc expression was affected at least in part by Runx3, thereby firstly exploring the functional role of Runx3 in TGF-β1 down-regulation on c-myc mRNA expression in fish.
Collapse
Affiliation(s)
- Fuli Yao
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China; Department of Biochemistry and Molecular Biology, College of Preclinical Medicine, Southwest Medical University, Luzhou, People's Republic of China
| | - Licheng Yin
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Shiyu Feng
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xinyan Wang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Anying Zhang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Hong Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| |
Collapse
|
14
|
Mittal P, Abblett R, Ryan JM, Hagymasi AT, Agyekum-Yamoah A, Svedova J, Reiner SL, St Rose MC, Hanley MP, Vella AT, Adler AJ. An Immunotherapeutic CD137 Agonist Releases Eomesodermin from ThPOK Repression in CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:1513-1526. [PMID: 29305435 DOI: 10.4049/jimmunol.1701039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Agonists to the TNF/TNFR costimulatory receptors CD134 (OX40) and CD137 (4-1BB) elicit antitumor immunity. Dual costimulation with anti-CD134 plus anti-CD137 is particularly potent because it programs cytotoxic potential in CD8+ and CD4+ T cells. Cytotoxicity in dual-costimulated CD4 T cells depends on the T-box transcription factor eomesodermin (Eomes), which we report is induced via a mechanism that does not rely on IL-2, in contrast to CD8+ CTL, but rather depends on the CD8 T cell lineage commitment transcription factor Runx3, which supports Eomes expression in mature CD8+ CTLs. Further, Eomes and Runx3 were indispensable for dual-costimulated CD4 T cells to mediate antitumor activity in an aggressive melanoma model. Runx3 is also known to be expressed in standard CD4 Th1 cells where it fosters IFN-γ expression; however, the CD4 T cell lineage commitment factor ThPOK represses transcription of Eomes and other CD8 lineage genes, such as Cd8a Hence, CD4 T cells can differentiate into Eomes+ cytotoxic CD4+CD8+ double-positive T cells by terminating ThPOK expression. In contrast, dual-costimulated CD4 T cells express Eomes, despite the continued expression of ThPOK and the absence of CD8α, indicating that Eomes is selectively released from ThPOK repression. Finally, although Eomes was induced by CD137 agonist, but not CD134 agonist, administered individually, CD137 agonist failed to induce CD134-/- CD4 T cells to express Eomes or Runx3, indicating that both costimulatory pathways are required for cytotoxic Th1 programming, even when only CD137 is intentionally engaged with a therapeutic agonist.
Collapse
Affiliation(s)
- Payal Mittal
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Rebecca Abblett
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Joseph M Ryan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam T Hagymasi
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | | | - Julia Svedova
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Steven L Reiner
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Matthew P Hanley
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam J Adler
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030;
| |
Collapse
|
15
|
Afaloniati H, Karagiannis GS, Hardas A, Poutahidis T, Angelopoulou K. Inflammation-driven colon neoplasmatogenesis in uPA-deficient mice is associated with an increased expression of Runx transcriptional regulators. Exp Cell Res 2017; 361:257-264. [DOI: 10.1016/j.yexcr.2017.10.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
|
16
|
Abstract
In this chapter we summarize the pros and cons of the notion that Runx3 is a major tumor suppressor gene (TSG). Inactivation of TSGs in normal cells provides a viability/growth advantage that contributes cell-autonomously to cancer. More than a decade ago it was suggested that RUNX3 is involved in gastric cancer development, a postulate extended later to other epithelial cancers portraying RUNX3 as a major TSG. However, evidence that Runx3 is not expressed in normal gastric and other epithelia has challenged the RUNX3-TSG paradigm. In contrast, RUNX3 is overexpressed in a significant fraction of tumor cells in various human epithelial cancers and its overexpression in pancreatic cancer cells promotes their migration, anchorage-independent growth and metastatic potential. Moreover, recent high-throughput quantitative genome-wide studies on thousands of human samples of various tumors and new investigations of the role of Runx3 in mouse cancer models have unequivocally demonstrated that RUNX3 is not a bona fide cell-autonomous TSG. Importantly, accumulating data demonstrated that RUNX3 functions in control of immunity and inflammation, thereby indirectly influencing epithelial tumor development.
Collapse
|
17
|
Fan L, Wang X, Fan L, Chen Q, Zhang H, Pan H, Xu A, Wang H, Yu Y. MicroRNA-145 influences the balance of Th1/Th2 via regulating RUNX3 in asthma patients. Exp Lung Res 2016; 42:417-424. [PMID: 27902892 DOI: 10.1080/01902148.2016.1256452] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIM OF THE STUDY To delineate the underlying mechanism of microRNA-145 modulate the balance of Th1/Th2 via targeting RUNX3 in asthma patients. MATERIALS AND METHODS Peripheral blood samples were collected from asthma patients and healthy controls. CD4+ T cells were isolated and cultured. Using quantitative PCR detect, the level of microRNA-145 and RUNX3 mRNA level in the CD4+ T cells from asthma patients and healthy controls, meanwhile, western blot was used to detect the RUNX3 protein level. Th1 or Th2 related cytokines were measured by enzyme-linked immunosorbent assay. Dual-Luciferase Reporter Assay was performed to confirm the correlation between microRNA-145 and RUNX3. MicroRNA-145 mimic or inhibitor was transfected in the CD4+ T cells and the changes of RUNX3 level, Th1 or Th2 related cytokines and the percentage of Th1 and Th2 were observed after transfection. RESULTS MicroRNA-145 level of CD4+ T cells was higher with a lower RUNX3 expression in asthma patients. There is negative correlation between microRNA-145 and RUNX3. Th2 hyperactivity and Th1 deficiency was detected in the CD4+ T cells of asthma patients. Dual-Luciferase Reporter Assay has shown that RUNX3 is a target of microRNA. Up-regulation or down-regulation of miR-145 level caused RUNX3 expression changes in CD4+ T cells and influence the related cytokines. Inhibition of microRNA-145 may reverse the imbalance of Th1/Th2 in asthma patients. CONCLUSION MicroRNA-145 could regulate the balance of Th1/Th2 through targeting the RUNX3 in asthma patients. MicroRNA-145 and RUNX3 may be used as biomarkers or targets in the diagnosis or therapy of asthma.
Collapse
Affiliation(s)
- Linxia Fan
- a Respiratory Department in the Cadre Ward , Gansu Provincial Hospital , Lanzhou , China
| | - Xiaojun Wang
- b Respiratory Department , Gansu Provincial Hospital , Lanzhou , China
| | - Linlan Fan
- c Medical Experimental Center of Lanzhou University , Lanzhou , China
| | - Qizhang Chen
- a Respiratory Department in the Cadre Ward , Gansu Provincial Hospital , Lanzhou , China
| | - Hong Zhang
- d Asthma Center of the Gansu Provincial Hospital , Lanzhou , China
| | - Hui Pan
- e Department of Internet Information , Gansu Provincial Hospital , Lanzhou , China
| | - Aixia Xu
- f Department of Pharmacy , Gansu Provincial Hospital , Lanzhou , China
| | - Hongjuan Wang
- a Respiratory Department in the Cadre Ward , Gansu Provincial Hospital , Lanzhou , China
| | - Yang Yu
- g Department of Internal Medicine , Traditional Chinese Medicine Hospital of Lintao County , Lintao , China
| |
Collapse
|
18
|
Mizoguchi A, Takeuchi T, Himuro H, Okada T, Mizoguchi E. Genetically engineered mouse models for studying inflammatory bowel disease. J Pathol 2015; 238:205-19. [PMID: 26387641 DOI: 10.1002/path.4640] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/05/2015] [Accepted: 09/14/2015] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition that is mediated by very complex mechanisms controlled by genetic, immune, and environmental factors. More than 74 kinds of genetically engineered mouse strains have been established since 1993 for studying IBD. Although mouse models cannot fully reflect human IBD, they have provided significant contributions for not only understanding the mechanism, but also developing new therapeutic means for IBD. Indeed, 20 kinds of genetically engineered mouse models carry the susceptibility genes identified in human IBD, and the functions of some other IBD susceptibility genes have also been dissected out using mouse models. Cutting-edge technologies such as cell-specific and inducible knockout systems, which were recently employed to mouse IBD models, have further enhanced the ability of investigators to provide important and unexpected rationales for developing new therapeutic strategies for IBD. In this review article, we briefly introduce 74 kinds of genetically engineered mouse models that spontaneously develop intestinal inflammation.
Collapse
Affiliation(s)
- Atsushi Mizoguchi
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Takahito Takeuchi
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Hidetomo Himuro
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Toshiyuki Okada
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Emiko Mizoguchi
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Fruit Street, Boston, MA, 02114, USA
| |
Collapse
|
19
|
Vecellio M, Roberts AR, Cohen CJ, Cortes A, Knight JC, Bowness P, Wordsworth BP. The genetic association of RUNX3 with ankylosing spondylitis can be explained by allele-specific effects on IRF4 recruitment that alter gene expression. Ann Rheum Dis 2015; 75:1534-40. [PMID: 26452539 PMCID: PMC4975853 DOI: 10.1136/annrheumdis-2015-207490] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 08/19/2015] [Indexed: 12/18/2022]
Abstract
Objectives To identify the functional basis for the genetic association of single nucleotide polymorphisms (SNP), upstream of the RUNX3 promoter, with ankylosing spondylitis (AS). Methods We performed conditional analysis of genetic association data and used ENCODE data on chromatin remodelling and transcription factor (TF) binding sites to identify the primary AS-associated regulatory SNP in the RUNX3 region. The functional effects of this SNP were tested in luciferase reporter assays. Its effects on TF binding were investigated by electrophoretic mobility gel shift assays and chromatin immunoprecipitation. RUNX3 mRNA levels were compared in primary CD8+ T cells of AS risk and protective genotypes by real-time PCR. Results The association of the RUNX3 SNP rs4648889 with AS (p<7.6×10−14) was robust to conditioning on all other SNPs in this region. We identified a 2 kb putative regulatory element, upstream of RUNX3, containing rs4648889. In reporter gene constructs, the protective rs4648889 ‘G’ allele increased luciferase activity ninefold but significantly less activity (4.3-fold) was seen with the AS risk ‘A’ allele (p≤0.01). The binding of Jurkat or CD8+ T-cell nuclear extracts to the risk allele was decreased and IRF4 recruitment was reduced. The AS-risk allele also affected H3K4Me1 histone methylation and associated with an allele-specific reduction in RUNX3 mRNA (p<0.05). Conclusion We identified a regulatory region upstream of RUNX3 that is modulated by rs4648889. The risk allele decreases TF binding (including IRF4) and reduces reporter activity and RUNX3 expression. These findings may have important implications for understanding the role of T cells and other immune cells in AS.
Collapse
Affiliation(s)
- Matteo Vecellio
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK National Institute for Health Research Oxford Musculoskeletal Biomedical Research Unit, Oxford, UK National Institute for Health Research Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - Amity R Roberts
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK National Institute for Health Research Oxford Musculoskeletal Biomedical Research Unit, Oxford, UK National Institute for Health Research Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - Carla J Cohen
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK National Institute for Health Research Oxford Musculoskeletal Biomedical Research Unit, Oxford, UK National Institute for Health Research Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - Adrian Cortes
- Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julian C Knight
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Paul Bowness
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK National Institute for Health Research Oxford Musculoskeletal Biomedical Research Unit, Oxford, UK National Institute for Health Research Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - B Paul Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK National Institute for Health Research Oxford Musculoskeletal Biomedical Research Unit, Oxford, UK National Institute for Health Research Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| |
Collapse
|
20
|
Tang RJ, Shen SN, Zhao XY, Nie YZ, Xu YJ, Ren J, Lv MM, Hou YY, Wang TT. Mesenchymal stem cells-regulated Treg cells suppress colitis-associated colorectal cancer. Stem Cell Res Ther 2015; 6:71. [PMID: 25889203 PMCID: PMC4414289 DOI: 10.1186/s13287-015-0055-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 08/30/2014] [Accepted: 03/18/2015] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Previous studies have produced controversial results regarding whether mesenchymal stem cells (MSCs) promote or inhibit tumor development. Given the dual role of MSCs in inflammation and cancer, in this study the colitis-associated colorectal cancer (CAC) model was used to examine whether umbilical cord tissue-derived MSCs could prevent neoplasm by inhibiting chronic inflammation. METHODS MSCs were obtained and identified using flow cytometry. Colitis-associated colorectal cancer model was induced using azoxymethane (AOM) and dextran sulfate sodium (DSS) and MSCs were injected intravenously twice. Levels of immune cells in mesenteric lymph node including regulatory T (Treg) cells were detected using flow cytometry. Naïve T cells and Jurkat cells were co-cultured with MSCs and the effect of MSCs on Treg cells differentiation was evaluated. RESULTS After injection through tail vein, MSCs could migrate to colon and suppress colitis-related neoplasm. This tumor suppressive effect was characterized by longer colon length, decreased tumor numbers and decreased expression of Ki-67. Moreover, MSCs alleviated the pathology of inflammation in the colitis stage of CAC model and inhibited inflammation cytokines both in colon and serum. Furthermore, Treg cells were accumulated in mesenteric lymph node of MSCs-treated mice while the percentage of T helper cells 2 (Th2) and Th17 were not changed. Of note, MSCs secreted transforming growth factor-β (TGF-β) enhanced the induction of Treg cells from naïve T cells. The conditioned medium of MSCs also activated Smad2 signaling, which has been reported to regulate Treg cells. CONCLUSIONS These results proved that MSCs could migrate to colon tissues and induce the differentiation of Treg cells via Smad2 as so to inhibit the colitis and suppress the development of CAC.
Collapse
Affiliation(s)
- Rui-jing Tang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Su-nan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Xiao-yin Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yun-zhong Nie
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yu-jun Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Jing Ren
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Ming-ming Lv
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Ya-yi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Ting-ting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| |
Collapse
|
21
|
Lotem J, Levanon D, Negreanu V, Bauer O, Hantisteanu S, Dicken J, Groner Y. Runx3 at the interface of immunity, inflammation and cancer. Biochim Biophys Acta Rev Cancer 2015; 1855:131-43. [PMID: 25641675 DOI: 10.1016/j.bbcan.2015.01.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 02/06/2023]
Abstract
Inactivation of tumor suppressor genes (TSG) in normal cells provides a viability/growth advantage that contributes cell-autonomously to cancer. More than a decade ago claims arose that the RUNX3 member of the RUNX transcription factor family is a major TSG inactivated in gastric cancer, a postulate extended later to other cancers. However, evidence that Runx3 is not expressed in normal gastric and other epithelia has challenged the RUNX3-TSG paradigm. Here we critically re-appraise this paradigm in light of recent high-throughput, quantitative genome-wide studies on thousands of human samples of various tumors and new investigations of the role of Runx3 in mouse cancer models. Collectively, these studies unequivocally demonstrate that RUNX3 is not a bona fide cell-autonomous TSG. Accordingly, RUNX3 is not recognized as a TSG and is not included among the 2000 cancer genes listed in the "Cancer Gene Census" or "Network for Cancer Genes" repositories. In contrast, RUNX3 does play important functions in immunity and inflammation and may thereby indirectly influence epithelial tumor development.
Collapse
Affiliation(s)
- Joseph Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Ditsa Levanon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Negreanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Omri Bauer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Shay Hantisteanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Joseph Dicken
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoram Groner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
22
|
Wu HZ, Cai MN, An Y, Lan C, Wei JL, Sun XN. Runx3 might participate in regulating dendriti cell function in patients with irritable bowel syndrome. ASIAN PAC J TROP MED 2014. [DOI: 10.1016/s1995-7645(14)60130-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
23
|
Kurklu B, Whitehead RH, Ong EK, Minamoto T, Fox JG, Mann JR, Judd LM, Giraud AS, Menheniott TR. Lineage-specific RUNX3 hypomethylation marks the preneoplastic immune component of gastric cancer. Oncogene 2014; 34:2856-66. [PMID: 25088199 DOI: 10.1038/onc.2014.233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/20/2014] [Indexed: 12/22/2022]
Abstract
Runt domain transcription factor 3 (RUNX3) is widely regarded as a tumour-suppressor gene inactivated by DNA hypermethylation of its canonical CpG (cytidine-phosphate-guanidine) island (CGI) promoter in gastric cancer (GC). Absence of RUNX3 expression from normal gastric epithelial cells (GECs), the progenitors to GC, coupled with frequent RUNX3 overexpression in GC progression, challenge this longstanding paradigm. However, epigenetic models to better describe RUNX3 deregulation in GC have not emerged. Here, we identify lineage-specific DNA methylation at an alternate, non-CGI promoter (P1) as a new mechanism of RUNX3 epigenetic control. In normal GECs, P1 was hypermethylated and repressed, whereas in immune lineages P1 was hypomethylated and widely expressed. In human GC development, we detected aberrant P1 hypomethylation signatures associated with the early inflammatory, preneoplastic and tumour stages. Aberrant P1 hypomethylation was fully recapitulated in mouse models of gastric inflammation and tumorigenesis. Cell sorting showed that P1 hypomethylation reflects altered cell-type composition of the gastric epithelium/tumour microenvironment caused by immune cell recruitment, not methylation loss. Finally, via long-term culture of gastric tumour epithelium, we revealed that de novo methylation of the RUNX3 canonical CGI promoter is a bystander effect of oncogenic immortalization and not likely causal in GC pathogenesis as previously argued. We propose a new model of RUNX3 epigenetic control in cancer, based on immune-specific, non-CGI promoter hypomethylation. This novel epigenetic signature may have utility in early detection of GC and possibly other epithelial cancers with premalignant immune involvement.
Collapse
Affiliation(s)
- B Kurklu
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - R H Whitehead
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - E K Ong
- Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - T Minamoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - J G Fox
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J R Mann
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Zoology, University of Melbourne, Melbourne, Victoria, Australia
| | - L M Judd
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - A S Giraud
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - T R Menheniott
- 1] Infection and Immunity, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia [2] Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Ondondo BO. Fallen angels or risen apes? A tale of the intricate complexities of imbalanced immune responses in the pathogenesis and progression of immune-mediated and viral cancers. Front Immunol 2014; 5:90. [PMID: 24639678 PMCID: PMC3944202 DOI: 10.3389/fimmu.2014.00090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 02/20/2014] [Indexed: 12/13/2022] Open
Abstract
Excessive immune responses directed against foreign pathogens, self-antigens, or commensal microflora can cause cancer establishment and progression if the execution of tight immuno-regulatory mechanisms fails. On the other hand, induction of potent tumor antigen-specific immune responses together with stimulation of the innate immune system is a pre-requisite for effective anti-tumor immunity, and if suppressed by the strong immuno-regulatory mechanisms can lead to cancer progression. Therefore, it is crucial that the inevitable co-existence of these fundamental, yet conflicting roles of immune-regulatory cells is carefully streamlined as imbalances can be detrimental to the host. Infection with chronic persistent viruses is characterized by severe immune dysfunction resulting in T cell exhaustion and sometimes deletion of antigen-specific T cells. More often, this is due to increased immuno-regulatory processes, which are triggered to down-regulate immune responses and limit immunopathology. However, such heightened levels of immune disruption cause a concomitant loss of tumor immune-surveillance and create a permissive microenvironment for cancer establishment and progression, as demonstrated by increased incidences of cancer in immunosuppressed hosts. Paradoxically, while some cancers arise as a consequence of increased immuno-regulatory mechanisms that inhibit protective immune responses and impinge on tumor surveillance, other cancers arise due to impaired immuno-regulatory mechanisms and failure to limit pathogenic inflammatory responses. This intricate complexity, where immuno-regulatory cells can be beneficial in certain immune settings but detrimental in other settings underscores the need for carefully formulated interventions to equilibrate the balance between immuno-stimulatory and immuno-regulatory processes.
Collapse
|
25
|
Lotem J, Levanon D, Negreanu V, Groner Y. The False Paradigm of RUNX3 Function as Tumor Suppressor in Gastric Cancer. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jct.2013.41a003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Műzes G, Molnár B, Sipos F. Regulatory T cells in inflammatory bowel diseases and colorectal cancer. World J Gastroenterol 2012; 18:5688-94. [PMID: 23155308 PMCID: PMC3484336 DOI: 10.3748/wjg.v18.i40.5688] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 04/06/2012] [Accepted: 04/12/2012] [Indexed: 02/06/2023] Open
Abstract
Regulatory T cells (T(regs)) are key elements in immunological self-tolerance. The number of T(regs) may alter in both peripheral blood and in colonic mucosa during pathological circumstances. The local cellular, microbiological and cytokine milieu affect immunophenotype and function of T(regs). Forkhead box P3+ T(regs) function shows altered properties in inflammatory bowel diseases (IBDs). This alteration of T(regs) function can furthermore be observed between Crohn's disease and ulcerative colitis, which may have both clinical and therapeutical consequences. Chronic mucosal inflammation may also influence T(regs) function, which together with the intestinal bacterial flora seem to have a supporting role in colitis-associated colorectal carcinogenesis. T(regs) have a crucial role in the immunoevasion of cancer cells in sporadic colorectal cancer. Furthermore, their number and phenotype correlate closely with the clinical outcome of the disease, even if their contribution to carcinogenesis has previously been controversial. Despite knowledge of the clinical relationship between IBD and colitis-associated colon cancer, and the growing number of immunological aspects encompassing sporadic colorectal carcinogenesis, the molecular and cellular links amongst T(regs), regulation of the inflammation, and cancer development are still not well understood. In this paper, we aimed to review the current data surrounding the role of T(regs) in the pathogenesis of IBD, colitis-associated colon cancer and sporadic colorectal cancer.
Collapse
Affiliation(s)
- Györgyi Műzes
- 2nd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary
| | | | | |
Collapse
|
27
|
Nambu Y, Hayashi T, Jang KJ, Aoki K, Mano H, Nakano K, Osato M, Takahashi K, Itoh K, Teramukai S, Komori T, Fujita J, Ito Y, Shimizu A, Sugai M. In situ differentiation of CD8αα Τ cells from CD4 T cells in peripheral lymphoid tissues. Sci Rep 2012; 2:642. [PMID: 22962636 PMCID: PMC3435563 DOI: 10.1038/srep00642] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/23/2012] [Indexed: 12/24/2022] Open
Abstract
Mutually exclusive cell fate determination of CD4 helper or CD8 killer T cells occurs in the thymus. These T-cell subsets are not believed to redirect other lineages. Here we showed that retinoic acid and transforming growth factor-β1 promoted the differentiation of CD8αα T cells from CD4 T cells in a Runx3-dependent manner. These cells were inferred to belong to immunoregulatory populations because subpopulations of CD8αα+TCRαβ T cells are known to suppress activated T cells, and mice with Runx3(-/-) T cells showed defects during recovery from experimental allergic encephalomyelitis. Our results demonstrate that CD4 T cells play fundamental roles in controlling immune reactions through promotion and attenuation. We accordingly anticipate that clarifying the mechanisms underlying this process will provide insights leading to autoimmune and immunodeficiency disease therapies.
Collapse
Affiliation(s)
- Yukiko Nambu
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, 54 Shogoin-Kawahara-cho,Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
The dual role of inflammation in colon carcinogenesis. Int J Mol Sci 2012; 13:11071-11084. [PMID: 23109839 PMCID: PMC3472731 DOI: 10.3390/ijms130911071] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 08/28/2012] [Accepted: 08/28/2012] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation characterizing patients with inflammatory bowel disease (IBD) represents a major risk factor for the development of colorectal cancer. Mechanisms underlying this neoplastic transformation are not fully understood though studies in experimental models of colon carcinogenesis suggest that inflammatory cell-derived cytokines either directly or indirectly stimulate the uncontrolled growth of cancer cells. Nevertheless, under specific inflammatory conditions, immune cells can boost an anti-tumor immune response with the down-stream effect of eliminating dysplastic and cancerous cells. This review outlines the beneficial and detrimental role of inflammation in colon carcinogenesis.
Collapse
|
29
|
Fijneman RJA, Anderson RA, Richards E, Liu J, Tijssen M, Meijer GA, Anderson J, Rod A, O'Sullivan MG, Scott PM, Cormier RT. Runx1 is a tumor suppressor gene in the mouse gastrointestinal tract. Cancer Sci 2012; 103:593-9. [PMID: 22171576 DOI: 10.1111/j.1349-7006.2011.02189.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Runx1 transcription factor plays an important role in tissue homeostasis through its effects on stem/progenitor cell populations and differentiation. The effect of Runx1 on epithelial differentiation of the secretory cell lineage of the colon was recently demonstrated. This study aimed to examine the role of Runx1 in tumor development in epithelial cells of the gastrointestinal tract. Conditional knockout mice that lacked Runx1 expression in epithelial cells of the GI tract were generated. These mice were crossed onto the Apc(Min) background, killed and their intestinal tumor phenotypes were compared with Apc(Min) Runx1 wild-type control mice. Apc-wild-type Runx1-mutant mice were also examined for tumor development. Colons from Runx1 knockout and wild-type mice were used for genome-wide mRNA expression analyses followed by gene-specific quantitative RT-PCR of whole colon and colon epithelium to identify Runx1 target genes. Runx1 deficiency in intestinal epithelial cells significantly enhanced tumorigenesis in Apc(Min) mice. Notably, epithelial Runx1 deficiency in Apc-wild-type mice was sufficient to cause tumor development. Absence of Runx1 was associated with global changes in the expression of genes involved in inflammation and intestinal metabolism, and with gene sets indicative of a metastatic phenotype and poor prognosis. Gene-specific analysis of Runx1-deficient colon epithelium revealed increased expression of genes linked to an expansion of the stem/progenitor cell population. These results identify Runx1 as a novel tumor suppressor gene for gastrointestinal tumors and support a role for Runx1 in maintaining the balance between the intestinal stem/progenitor cell population and epithelial differentiation of the GI tract.
Collapse
Affiliation(s)
- Remond J A Fijneman
- Department of Pathology, VU University Medical Center, Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mizoguchi A. Animal models of inflammatory bowel disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 105:263-320. [PMID: 22137435 DOI: 10.1016/b978-0-12-394596-9.00009-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition that is medicated by genetic, immune, and environmental factors. At least 66 different kinds of animal models have been established to study IBD, which are classified primarily into chemically induced, cell-transfer, congenial mutant, and genetically engineered models. These IBD models have provided significant contributions to not only dissect the mechanism but also develop novel therapeutic strategies for IBD. In addition, recent advances on genetically engineered techniques such as cell-specific and inducible knockout as well as knockin mouse systems have brought novel concepts on IBD pathogenesis to the fore. Further, mouse models, which lack some IBD susceptibility genes, have suggested more complicated mechanism of IBD than previously predicted. This chapter summarizes the distinct feature of each murine IBD model and discusses the previous and current lessons from the IBD models.
Collapse
Affiliation(s)
- Atsushi Mizoguchi
- Department of Pathology, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|