1
|
Zivotic I, Kolic I, Cvetkovic M, Spasojevic-Dimitrijeva B, Zivkovic M, Stankovic A, Jovanovic I. Copy number variation analysis identifies MIR9-3 and MIR1299 as novel miRNA candidate genes for CAKUT. Pediatr Nephrol 2024; 39:2655-2665. [PMID: 38656454 DOI: 10.1007/s00467-024-06381-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Congenital anomalies of the kidney and urinary tract (CAKUT) represent a frequent cause of pediatric kidney failure. CNVs, as a major class of genomic variations, can also affect miRNA regions. Common CNV corresponding miRNAs (cCNV-miRNAs) are functional variants regulating crucial processes which could affect urinary system development. Thus, we hypothesize that cCNV-miRNAs are associated with CAKUT occurrence and its expressivity. METHODS The extraction and filtering of common CNVs, identified in control samples deposited in publicly available databases gnomAD v2.1 and dbVar, were coupled with mapping of miRNA sequences using UCSC Genome Browser. After verification of the mapped miRNAs using referent miRBase V22.1, prioritization of cCNV-miRNA candidates has been performed using bioinformatic annotation and literature research. Genotyping of miRNA gene copy numbers for MIR9-3, MIR511, and MIR1299, was conducted on 221 CAKUT patients and 192 controls using TaqMan™ technology. RESULTS We observed significantly different MIR9-3 and MIR1299 gene copy number distribution between CAKUT patients and controls (Chi-square, P = 0.006 and P = 0.0002, respectively), while difference of MIR511 copy number distribution showed nominal significance (Chi-square, P = 0.027). The counts of less and more than two of MIR1299 copy numbers were more frequent within CAKUT patients compared to controls (P = 0.01 and P = 0.008, respectively) and also in cohort of patients with anomalies of the urinary tract compared to controls (P = 0.016 and P = 0.003, respectively). CONCLUSIONS Copy number variations of miRNA genes represent a novel avenue in clarification of the inheritance complexity in CAKUT and provide potential evidence about the association of common genetic variation with CAKUT phenotypes.
Collapse
Affiliation(s)
- Ivan Zivotic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001, Belgrade, Serbia
| | - Ivana Kolic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001, Belgrade, Serbia
| | - Mirjana Cvetkovic
- Nephrology and Urology Departments, University Children's Hospital, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Brankica Spasojevic-Dimitrijeva
- Nephrology and Urology Departments, University Children's Hospital, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Maja Zivkovic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001, Belgrade, Serbia
| | - Aleksandra Stankovic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001, Belgrade, Serbia
| | - Ivan Jovanovic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11001, Belgrade, Serbia.
| |
Collapse
|
2
|
Alhosseini MN, Ebadi P, Karimi MH, Migliorati G, Cari L, Nocentini G, Heidari M, Soleimanian S. Therapy with regulatory T-cell infusion in autoimmune diseases and organ transplantation: A review of the strengths and limitations. Transpl Immunol 2024; 85:102069. [PMID: 38844002 DOI: 10.1016/j.trim.2024.102069] [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: 11/10/2023] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/17/2024]
Abstract
In the last decade, cell therapies have revolutionized the treatment of some diseases, earning the definition of being the "third pillar" of therapeutics. In particular, the infusion of regulatory T cells (Tregs) is explored for the prevention and control of autoimmune reactions and acute/chronic allograft rejection. Such an approach represents a promising new treatment for autoimmune diseases to recover an immunotolerance against autoantigens, and to prevent an immune response to alloantigens. The efficacy of the in vitro expanded polyclonal and antigen-specific Treg infusion in the treatment of a large number of autoimmune diseases has been extensively demonstrated in mouse models. Similarly, experimental work documented the efficacy of Treg infusions to prevent acute and chronic allograft rejections. The Treg therapy has shown encouraging results in the control of type 1 diabetes (T1D) as well as Crohn's disease, systemic lupus erythematosus, autoimmune hepatitis and delaying graft rejection in clinical trials. However, the best method for Treg expansion and the advantages and pitfalls with the different types of Tregs are not fully understood in terms of how these therapeutic treatments can be applied in the clinical setting. This review provides an up-to-date overview of Treg infusion-based treatments in autoimmune diseases and allograft transplantation, the current technical challenges, and the highlights and disadvantages of this therapeutic approaches."
Collapse
Affiliation(s)
| | - Padideh Ebadi
- Islamic Azad University, Department of Biochemistry, Kazerun, Iran
| | | | - Graziella Migliorati
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, Perugia, Italy
| | - Luigi Cari
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, Perugia, Italy
| | - Giuseppe Nocentini
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, Perugia, Italy
| | - Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Xu SJ, Chen JH, Chang S, Li HL. The role of miRNAs in T helper cell development, activation, fate decisions and tumor immunity. Front Immunol 2024; 14:1320305. [PMID: 38264670 PMCID: PMC10803515 DOI: 10.3389/fimmu.2023.1320305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
T helper (Th) cells are central members of adaptive immunity and comprise the last line of defense against pathogen infection and malignant cell invasion by secreting specific cytokines. These cytokines then attract or induce the activation and differentiation of other immune cells, including antibody-producing B cells and cytotoxic CD8+ T cells. Therefore, the bidirectional communication between Th cells and tumor cells and their positioning within the tumor microenvironment (TME), especially the tumor immune microenvironment (TIME), sculpt the tumor immune landscape, which affects disease initiation and progression. The type, number, and condition of Th cells in the TME and TIME strongly affect tumor immunity, which is precisely regulated by key effectors, such as granzymes, perforins, cytokines, and chemokines. Moreover, microRNAs (miRNAs) have emerged as important regulators of Th cells. In this review, we discuss the role of miRNAs in regulating Th cell mediated adaptive immunity, focusing on the development, activation, fate decisions, and tumor immunity.
Collapse
Affiliation(s)
- Shi-Jun Xu
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- Henan Medical Device Engineering Research Center of Interventional Therapy for Non-vascular Tumors, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jin-Hua Chen
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Suhwan Chang
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hai-Liang Li
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- Henan Medical Device Engineering Research Center of Interventional Therapy for Non-vascular Tumors, Henan Cancer Hospital, Zhengzhou, Henan, China
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
4
|
South AP, Laimer M, Gueye M, Sui JY, Eichenfield LF, Mellerio JE, Nyström A. Type VII Collagen Deficiency in the Oncogenesis of Cutaneous Squamous Cell Carcinoma in Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2023; 143:2108-2119. [PMID: 37327859 DOI: 10.1016/j.jid.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Dystrophic epidermolysis bullosa is a rare genetic skin disorder caused by COL7A1 sequence variations that result in type VII collagen deficits and cutaneous and extracutaneous manifestations. One serious complication of dystrophic epidermolysis bullosa is cutaneous squamous cell carcinoma, a leading driver of morbidity and mortality, especially among patients with recessive dystrophic epidermolysis bullosa. Type VII collagen deficits alter TGFβ signaling and evoke multiple other cutaneous squamous cell carcinoma progression-promoting activities within epidermal microenvironments. This review examines cutaneous squamous cell carcinoma pathophysiology in dystrophic epidermolysis bullosa with a focus on known oncogenesis pathways at play and explores the idea that therapeutic type VII collagen replacement may reduce cutaneous squamous cell carcinoma risk.
Collapse
Affiliation(s)
- Andrew P South
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | | | - Jennifer Y Sui
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Jemima E Mellerio
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
5
|
Fayyad-Kazan M, Rouas R, Merimi M, Najar M, Badran B, Lewalle P, Fayyad-Kazan H. Human CD4 +CD25 +CD127 lowFOXP3 + regulatory T lymphocytes and CD4 +CD25 -FOXP3 - conventional T lymphocytes: a differential transcriptome profile. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:919-929. [PMID: 37246921 DOI: 10.1080/15257770.2023.2216226] [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: 02/15/2022] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/30/2023]
Abstract
CD4+CD25+ FOXP3+ regulatory T cells (Tregs) represent a subpopulation of CD4+ T cells central for the suppression of physiological and pathological immune reactions. Although distinct cell surface antigens are expressed in regulatory T cells, those components are also present on the surface of activated CD4+CD25- FOXP3-T cells, thus making the discrimination between Tregs and conventional CD4+ T difficult and isolation of Tregs complex. Yet, the molecular components driving Tregs' function are still not fully characterized. Aiming at unraveling molecular components specifically marking Tregs, and upon using quantitative real-time PCR (qRT-PCR) followed by bioinformatics analysis, we identified, in this study, differential transcriptional profiles, in peripheral blood CD4 + CD25 + CD127low FOXP3+ Tregs versus CD4 + CD25-FOXP3- conventional T cells, for set of genes with distinct immunological roles. In conclusion, this study identifies some novel genes that appeared to be differentially transcribed in CD4+ Tregs versus conventional T cells. The identified genes could serve as novel molecular targets relevant to Tregs' function and isolation.
Collapse
Affiliation(s)
- Mohammad Fayyad-Kazan
- College of Arts and Sciences, Department of Natural and Applied Sciences, The American University of Iraq-Baghdad (AUIB), Baghdad, Iraq
| | - Redouane Rouas
- Laboratory of Experimental Hematology, Hematology Department, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Makram Merimi
- Laboratory of Experimental Hematology, Hematology Department, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
- Genetics and Immune-Cell therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Mehdi Najar
- Laboratory of Experimental Hematology, Hematology Department, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Hematology Department, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| |
Collapse
|
6
|
Jiang P, Zheng C, Xiang Y, Malik S, Su D, Xu G, Zhang M. The involvement of TH17 cells in the pathogenesis of IBD. Cytokine Growth Factor Rev 2023; 69:28-42. [PMID: 35871978 DOI: 10.1016/j.cytogfr.2022.07.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 02/07/2023]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) is still unclear. Immune dysfunction may play a key role in the pathogenesis of IBD, in which the role of CD4+ T helper (Th) cells is particularly important. Th17 cells are a major component of CD4+ T cells, and their differentiation is regulated by a variety of extracellular signals, transcription factors, RNA, and posttranslational modifications. Th17 cells specifically produce IL-17 and play an important role in the protection of mucous membranes and epithelial tissues against infection by extracellular microbes. However, when immune regulation is dysfunctional, Th17 cells abnormally proliferate and produce large amounts of proinflammatory cytokines that can recruit other inflammatory cells, which together induce abnormal immune responses and result in the development of many autoimmune diseases. In recent years, studies have confirmed that Th17 cells play an important role in the pathogenesis of IBD, which makes it a possible target for IBD therapy. This article reviews the recent progress of Th17 cells involved in the pathogenesis of IBD and its targeted therapy.
Collapse
Affiliation(s)
- Ping Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Chang Zheng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Ying Xiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Sara Malik
- Northwestern University Feinberg School of Medicine, Chicago 60611, IL, USA
| | - Dan Su
- FUJIFILM Diosynth Biotechnologies, Watertown 02472, MA, USA
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China.
| | - Mingming Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China; Department of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai 200001, China.
| |
Collapse
|
7
|
Li X, Wang W, Chen J, Xie B, Luo S, Chen D, Cai C, Li C, Li W. The potential role of exosomal miRNAs and membrane proteins in acute HIV-infected people. Front Immunol 2022; 13:939504. [PMID: 36032099 PMCID: PMC9411714 DOI: 10.3389/fimmu.2022.939504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/21/2022] [Indexed: 11/23/2022] Open
Abstract
Exosomes play an important role during human immunodeficiency virus (HIV) acute infection. Yet, information regarding its cargo and its association with HIV rapid progressors (RPs) and typical progressors (TPs) remain largely unknown. In this study, exosomal miRNAs sequencing and mass cytometry were used to identify differential exosomal miRNAs and membrane proteins that participate in the pathogenesis of TPs and RPs. We discovered that miR-144-5p, miR-1180-3p, miR-451a, miR-362-5p, and miR-625-5p are associated with the TPs and miR-362-5p with the RPs. Decreased autophagy, amino acid metabolism, immune response, and IL-6 are closely related to RPs. In addition, SP1 was selected as the most significant transcription factor (TF) associated with disease progression. CD49D, CD5, CCR5, CD40, CD14, and CD86 were selected as the differential exosomal membrane proteins between TPs and RPs. This study provides valuable information for clarifying the mechanism in people with acute HIV infection.
Collapse
Affiliation(s)
- Xin Li
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- General Surgery Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jing Chen
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Bangxiang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Shumin Luo
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chao Cai
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Weihua Li, ; Chao Cai, ; Chuanyun Li,
| | - Chuanyun Li
- General Surgery Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Weihua Li, ; Chao Cai, ; Chuanyun Li,
| | - Weihua Li
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Weihua Li, ; Chao Cai, ; Chuanyun Li,
| |
Collapse
|
8
|
Targeting TGF-β: triumphs and challenges. Future Med Chem 2022; 14:455-458. [DOI: 10.4155/fmc-2021-0344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
9
|
Liu X, Li H. A Systematic Review and Meta-Analysis on Multiple Cytokine Gene Polymorphisms in the Pathogenesis of Periodontitis. Front Immunol 2022; 12:713198. [PMID: 35046930 PMCID: PMC8761621 DOI: 10.3389/fimmu.2021.713198] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/10/2021] [Indexed: 12/30/2022] Open
Abstract
Aim Periodontitis is an inflammatory disease that destroys both soft and hard periodontal tissues. However, a complex periodontal cytokine network remains unclear. This systematic review explored multiple cytokine gene polymorphisms in the pathogenesis of periodontitis. Material and Methods A systematic search was performed using the databases from previous publications, which indicated the association between cytokine polymorphisms and periodontitis pathogenesis. Meta-analysis was conducted using fixed or randomized models to calculate the significance of multiple cytokine polymorphisms. A total of 147 articles were analyzed with polymorphisms in 12 interleukins [Th1 (IL-2, IFN-γ, and TNF-α), Th2 (IL-4 and IL-13), Th17 (IL-1α, IL-1β, IL-6, and IL-17), and Treg cytokines (IL-10 and TGF-β)]. Doi plot was used to probe the occurrence of publication bias. Results The polymorphisms of IL-2 and TNF-α of Th1 cytokine family may be associated with the pathogenesis or the prevention of periodontitis risk, while the polymorphism of IFN-γ is not related to periodontitis risk. The polymorphisms for IL-4 and IL-13 of Th2 cytokine family are not found to be associated with the pathogenesis of periodontitis. For the polymorphisms of the members of Th17 cytokine family, different IL-1α polymorphisms may have inverse actions in the pathogenesis of periodontitis. IL-1β is a noteworthy cytokine biomarker in periodontitis development and progression. IL-6 may have a protective function in the inflammatory responses of periodontitis, and IL-17 has a weak relationship the inflammatory responses. The polymorphisms for the members of Treg cell cytokines may have a protective function against periodontitis risk. LFK indexes show the major asymmetry due to publication bias. Conclusion IL-1β is a notable cytokine biomarker in periodontitis risk. Treg cytokines favor an anti-inflammatory and protective environment. Further data are needed to confirm the present conclusion due to publication bias.
Collapse
Affiliation(s)
- Xin Liu
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hui Li
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Bermick J, Schaller M. Epigenetic regulation of pediatric and neonatal immune responses. Pediatr Res 2022; 91:297-327. [PMID: 34239066 DOI: 10.1038/s41390-021-01630-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Epigenetic regulation of transcription is a collective term that refers to mechanisms known to regulate gene transcription without changing the underlying DNA sequence. These mechanisms include DNA methylation and histone tail modifications which influence chromatin accessibility, and microRNAs that act through post-transcriptional gene silencing. Epigenetics is known to regulate a variety of biological processes, and the role of epigtenetics in immunity and immune-mediated diseases is becoming increasingly recognized. While DNA methylation is the most widely studied, each of these systems play an important role in the development and maintenance of appropriate immune responses. There is clear evidence that epigenetic mechanisms contribute to developmental stage-specific immune responses in a cell-specific manner. There is also mounting evidence that prenatal exposures alter epigenetic profiles and subsequent immune function in exposed offspring. Early life exposures that are associated with poor long-term health outcomes also appear to impact immune specific epigenetic patterning. Finally, each of these epigenetic mechanisms contribute to the pathogenesis of a wide variety of diseases that manifest during childhood. This review will discuss each of these areas in detail. IMPACT: Epigenetics, including DNA methylation, histone tail modifications, and microRNA expression, dictate immune cell phenotypes. Epigenetics influence immune development and subsequent immune health. Prenatal, perinatal, and postnatal exposures alter immune cell epigenetic profiles and subsequent immune function. Numerous pediatric-onset diseases have an epigenetic component. Several successful strategies for childhood diseases target epigenetic mechanisms.
Collapse
Affiliation(s)
- Jennifer Bermick
- Department of Pediatrics, Division of Neonatology, University of Iowa, Iowa City, IA, USA. .,Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA.
| | - Matthew Schaller
- Department of Pulmonary, Critical Care & Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
11
|
Rossi ÁD, Higa LM, Herlinger AL, Ribeiro-Alves M, de Menezes MT, Giannini ALM, Cardoso CC, Da Poian AT, Tanuri A, Aguiar RS. Differential Expression of Human MicroRNAs During Dengue Virus Infection in THP-1 Monocytes. Front Cell Infect Microbiol 2021; 11:714088. [PMID: 34568093 PMCID: PMC8455953 DOI: 10.3389/fcimb.2021.714088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/25/2021] [Indexed: 12/31/2022] Open
Abstract
Dengue virus (DENV) is the most widespread arbovirus, responsible for a wide range of clinical manifestations, varying from self-limited illness to severe hemorrhagic fever. Dengue severity is associated with host intense proinflammatory response and monocytes have been considered one of the key cell types involved in the early steps of DENV infection and immunopathogenesis. To better understand cellular mechanisms involved in monocyte infection by DENV, we analyzed the expression levels of 754 human microRNAs in DENV-infected THP-1 cells, a human monocytic cell line. Eleven human microRNAs showed differential expression after DENV infection and gene ontology and enrichment analysis revealed biological processes potentially affected by these molecules. Five downregulated microRNAs were significantly linked to cellular response to stress, four to cell death/apoptosis, two to innate immune responses and one upregulated to vesicle mediated, TGF-β signaling, phosphatidylinositol mediated signaling, lipid metabolism process and blood coagulation.
Collapse
Affiliation(s)
- Átila Duque Rossi
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiza Mendonça Higa
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Bioquímica de Vírus, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alice Laschuk Herlinger
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Ribeiro-Alves
- Laboratório de Pesquisa Clínica em DST/AIDS, Instituto Nacional de Infectologia Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Mariane Talon de Menezes
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Lucia Moraes Giannini
- Laboratório de Genômica Funcional e Transdução de Sinal, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea T Da Poian
- Laboratório de Bioquímica de Vírus, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Santana Aguiar
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
12
|
The circular RNA circINPP4B acts as a sponge of miR-30a to regulate Th17 cell differentiation during progression of experimental autoimmune encephalomyelitis. Cell Mol Immunol 2021; 18:2177-2187. [PMID: 34363030 PMCID: PMC8429594 DOI: 10.1038/s41423-021-00748-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/18/2021] [Indexed: 02/06/2023] Open
Abstract
Circular RNAs (circRNAs) regulate gene expression and participate in various biological and pathological processes. However, little is known about the effects of specific circRNAs on T helper cell 17 (Th17) differentiation and related autoimmune diseases, such as multiple sclerosis (MS). Here, using transcriptome microarray analysis at different stages of experimental autoimmune encephalomyelitis (EAE), we identified the EAE progression-related circINPP4B, which showed upregulated expression in Th17 cells from mice with EAE and during Th17 differentiation in vitro. Silencing of circINPP4B inhibited Th17 differentiation and alleviated EAE, characterized by less demyelination and Th17 infiltration in the spinal cord. Mechanistically, circINPP4B served as a sponge that directly targeted miR-30a to regulate Th17 differentiation. Furthermore, circINPP4B levels were associated with the developing phases of clinical relapsing-remitting MS patients. Our results indicate that circINPP4B plays an important role in promoting Th17 differentiation and progression of EAE by targeting miR-30a, which provides a potential diagnostic and therapeutic target for Th17-mediated MS.
Collapse
|
13
|
Wang H, Fang K, Yan W, Chang X. T-Cell Immune Imbalance in Rheumatoid Arthritis Is Associated with Alterations in NK Cells and NK-Like T Cells Expressing CD38. J Innate Immun 2021; 14:148-166. [PMID: 34428762 DOI: 10.1159/000516642] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 04/18/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND CD38+ NK (CD3- CD16+ CD38+ CD56+) cells were increased in rheumatoid arthritis (RA), which suppressed Treg cell differentiation. This study explored how CD38+ NK cells regulated CD4+ T-cell differentiation into Treg cells in RA. METHODS Proportions of CD38+ NK cells and their counterpart CD38+ NK-like T (CD3+ CD16+ CD38+ CD56+) cells were measured in RA and rats with collagen-induced arthritis (CIA). CD38+ NK cells and CD38+ NK-like T cells were cocultured with CD4+ T cells, respectively. RESULTS A significantly increased proportion of CD38+ NK cells and a decreased proportion of CD38+ NK-like T cells were detected in RA and CIA blood and synovial fluids. When CD4+ T cells were cocultured with CD38+ NK cells, mammalian target of rapamycin (mTOR) signaling was activated, and Th1/Th2 and Th17/Treg ratios were increased. When CD38+ NK cells were pretreated with anti-CD38 antibody, Treg cell proportion was increased, and Th1/Th2 and Th17/Treg ratios were decreased. CD38+ NK-like T cells showed the opposite results. CD38+ NK cells and CD38+ NK-like-T cells activated differential gene expressions and pathways in CD4+ T cells and initiated Th1 and Th2 cell differentiation by differential gene nodes. CONCLUSIONS This study suggest that the high CD38+ NK cell proportion and low CD38+ NK-like T cell proportion in RA suppress Treg cell differentiation by stimulating mTOR signaling in CD4+ T cells, which consequentially disturbs the immune tolerance.
Collapse
Affiliation(s)
- Hongxing Wang
- Medical Research Center of The Affiliated Hospital of Qingdao University, Qingdao, China.,Clinical Laboratory of Qilu Hospital, Shandong University, Jinan, China
| | - Kehua Fang
- Clinical Laboratory of The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weining Yan
- Joint Surgery Department of The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotian Chang
- Medical Research Center of The Affiliated Hospital of Qingdao University, Qingdao, China.,Qingdao Engineering Technology Center for Major Disease Marker, Qingdao, China
| |
Collapse
|
14
|
Made to Measure: Patient-Tailored Treatment of Multiple Sclerosis Using Cell-Based Therapies. Int J Mol Sci 2021; 22:ijms22147536. [PMID: 34299154 PMCID: PMC8304207 DOI: 10.3390/ijms22147536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, there is still no cure for multiple sclerosis (MS), which is an autoimmune and neurodegenerative disease of the central nervous system. Treatment options predominantly consist of drugs that affect adaptive immunity and lead to a reduction of the inflammatory disease activity. A broad range of possible cell-based therapeutic options are being explored in the treatment of autoimmune diseases, including MS. This review aims to provide an overview of recent and future advances in the development of cell-based treatment options for the induction of tolerance in MS. Here, we will focus on haematopoietic stem cells, mesenchymal stromal cells, regulatory T cells and dendritic cells. We will also focus on less familiar cell types that are used in cell therapy, including B cells, natural killer cells and peripheral blood mononuclear cells. We will address key issues regarding the depicted therapies and highlight the major challenges that lie ahead to successfully reverse autoimmune diseases, such as MS, while minimising the side effects. Although cell-based therapies are well known and used in the treatment of several cancers, cell-based treatment options hold promise for the future treatment of autoimmune diseases in general, and MS in particular.
Collapse
|
15
|
Luo Y, Guo J, Zhang P, Cheuk YC, Jiang Y, Wang J, Xu S, Rong R. Mesenchymal Stem Cell Protects Injured Renal Tubular Epithelial Cells by Regulating mTOR-Mediated Th17/Treg Axis. Front Immunol 2021; 12:684197. [PMID: 34122446 PMCID: PMC8194268 DOI: 10.3389/fimmu.2021.684197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
The increase in T helper 17 cell (Th17)-mediated pro-inflammatory response and decrease in regulatory T cell (Treg)-mediated anti-inflammatory effect aggravate renal tubular epithelial cell (RTEC) injury. However, increasing evidence indicated that mesenchymal stem cell (MSC) possessed the ability to control the imbalance between Th17 and Treg. Given that Th17 and Treg are derived from a common CD4+ T cell precursor, we summarize the current knowledge of MSC-mediated inhibition of the mammalian target of rapamycin (mTOR), which is a master regulator of CD4+ T cell polarization. During CD4+ T cell differentiation, mTOR signaling mediates Th17 and Treg differentiation via hypoxia-inducible factor-1α (HIF-1α)-dependent metabolic regulation and signaling pathway, as well as mTOR-mediated phosphorylation of signal transducer and activator of transcription (STAT) 3 and 5. Through interfering with mTOR signaling, MSC restrains CD4+ T cell differentiation into Th17, but in turn promotes Treg generation. Thus, this review indicates that MSC-mediated Th17-to-Treg polarization is expected to act as new immunotherapy for kidney injury.
Collapse
Affiliation(s)
- Yongsheng Luo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jingjing Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Pingbao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yamei Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jiyan Wang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Shanghai Medical College, Fudan University, Shanghai, China
| | - Shihao Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
16
|
Fu J, Zang Y, Zhou Y, Chen C, Shao S, Shi G, Wu L, Zhu G, Sun T, Zhang D, Zhang T. Exploring a novel triptolide derivative possess anti-colitis effect via regulating T cell differentiation. Int Immunopharmacol 2021; 94:107472. [PMID: 33611058 DOI: 10.1016/j.intimp.2021.107472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/08/2021] [Accepted: 02/03/2021] [Indexed: 11/16/2022]
Abstract
Inflammatory bowel disease (IBD) is generally characterized by chronic inflammatory disorders of the gastrointestinal tract that are known as ulcerative colitis (UC) or Crohn's disease (CD). Although the underlying mechanism of action of IBD is unclear and because of the lack of satisfactory treatment, increasing evidence has indicated that pro-inflammatory cytokines that activate JAK-STAT signaling pathway regulate the differentiation of naïve T cells towards T helper (Th)1 and Th17 cell subsets and contribute to the development of IBD. ZT01 is a newly obtained triptolide derivative with strong anti-inflammatory effects and low toxicity. In this study, we evaluated the effects of ZT01 on DSS-induced colitis and investigated the underlying mechanism of action involved. Mice with DSS-induced acute or chronic colitis were used to assess the efficacy of ZT01 treatment, and T cells were cultured to analyze the differentiation of Th1 and Th17 cell by flow cytometry. In addition, intestinal epithelial barrier function, macrophage polarization, activation of the JAK-STAT signaling pathway, and the expression of cytokines and transcription factors were measured to assess the possible mechanisms of ZT01. We found that ZT01 had an obviously beneficial effect on DSS-induced colitis by improving the symptoms of bloody diarrhea, weight loss, and a shortened colon, thereby preserving the epithelial barrier function in the mouse colon. Furthermore, ZT01 significantly inhibited T cell differentiation into Th1 and/or Th17 cell subsets and macrophage polarization towards into an inflammatory phenotype via regulating the JAK-STAT signaling pathway. Thus, our findings suggested that ZT01 might be a potential pharmaceutical candidate that deserves to be further investigated as a treatment for IBD patients.
Collapse
Affiliation(s)
- Junmin Fu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yingda Zang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuai Shao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Gaona Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Guoyuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Tao Sun
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Dongming Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
17
|
Huang J, Xu X, Yang J. miRNAs Alter T Helper 17 Cell Fate in the Pathogenesis of Autoimmune Diseases. Front Immunol 2021; 12:593473. [PMID: 33968012 PMCID: PMC8096907 DOI: 10.3389/fimmu.2021.593473] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/18/2021] [Indexed: 01/05/2023] Open
Abstract
T helper 17 (Th17) cells are characterized by the secretion of the IL-17 cytokine and are essential for the immune response against bacterial and fungal infections. Despite the beneficial roles of Th17 cells, unrestrained IL-17 production can contribute to immunopathology and inflammatory autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease. Although these diverse outcomes are directed by the activation of Th17 cells, the regulation of Th17 cells is incompletely understood. The discovery that microRNAs (miRNAs) are involved in the regulation of Th17 cell differentiation and function has greatly improved our understanding of Th17 cells in immune response and disease. Here, we provide an overview of the biogenesis and function of miRNA and summarize the role of miRNAs in Th17 cell differentiation and function. Finally, we focus on recent advances in miRNA-mediated dysregulation of Th17 cell fate in autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Ji Yang
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Abstract
A type of evolutionarily conserved, noncoding, small, endogenous, single-stranded RNA, miRNAs are widely distributed in eukaryotes, where they participate in various biological processes as critical regulatory molecules. miR-1299 has mainly been investigated in cancers. miR-1299 is a tumor suppressor that regulates the expression of its target genes, activating or inhibiting the transcription of genes regulating biological activities including cell proliferation, migration, survival and programmed cell death. miR-1299 has become a hotspot in research of disease mechanisms and biomarkers; elucidation of the regulatory roles of miR-1299 in tumorigenesis, proliferation, apoptosis, invasion, migration and angiogenesis may provide a new perspective for understanding its biological functions as a tumor suppressor. As key regulatory molecules, microRNAs participate in various biological processes and have become a widespread research focus. This article discusses how the microRNA miR-1299 plays a role as a tumor suppressor and participates in the regulation of tumor pathogenesis. We provide an overview of the role of miR-1299 in tumor diseases and discuss the pathogenesis and regulation mechanisms of miR-1299 in different specific cancers.
Collapse
Affiliation(s)
- Deng Kaiyuan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, 150086, China
| | - Huang Lijuan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, 150086, China
| | - Sun Xueyuan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, 150086, China
| | - Zang Yunhui
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, 150086, China
| |
Collapse
|
19
|
Wang J, Zhang Y, Li H, Chen G, Zou Y, Rin K. Immune effects of miRNA and Th17 cells on β-Lg allergy in dietary milk based on mouse model. Saudi J Biol Sci 2020; 27:3442-3448. [PMID: 33304154 PMCID: PMC7715044 DOI: 10.1016/j.sjbs.2020.08.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/02/2022] Open
Abstract
β-lactoglobulin (β-Lg) allergy in dietary milk seriously affects the use of high-quality milk protein in infants. In order to solve this problem, the expression of miRNA and Th17 cells in milk β-Lg allergic reaction of children's diet was studied. Method: female BALB/c mice aged 5–6 weeks were selected as the subjects and randomly divided into blank group and β-Lg sensitized group, with 10 mice in each group. On the 1st, 7th and 14th day, the mice in the β-Lg sensitized group were intraperitoneally injected with allergen (Freund's adjuvant + β-Lg). Mice in the blank group were given the same amount of normal saline. Blood samples were collected from the eyeballs of mice to determine the number of inflammatory cells. The contents of Th17 related cytokines and transcription factors in spleen were detected by RT-PCR. Results: 1. the number of eosinophils and neutrophils in the β-Lg sensitized group were 15.76/mL and 24.36/mL, respectively, which were significantly higher than those in the blank group (P < 0.05); 2. in the mice of β-Lg sensitized group, the expression of miR-146a and miR-155 was abnormal, the number of Th17 cells was abnormally increased, and the expression levels of IL-17 and RORγt were significantly increased; 3. the abnormal expression of miR-146a and miR-155 in the mice of β-Lg sensitized group was positively correlated with the secretion of Th17 related cytokines, which could be used as one of the biological indexes to evaluate allergic reaction. Conclusion: the number of Thl7 increased abnormally in dietary milk allergy patients. miRNA gene expression and IL-17 expression could be used as one of the biological indicators to evaluate the allergic reaction of β-Lg.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Ying Zhang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Hong Li
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Guoqing Chen
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yuqiong Zou
- Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Kathe Rin
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester 01609, United States
| |
Collapse
|
20
|
Janssens I, Cools N. Regulating the regulators: Is introduction of an antigen-specific approach in regulatory T cells the next step to treat autoimmunity? Cell Immunol 2020; 358:104236. [PMID: 33137651 DOI: 10.1016/j.cellimm.2020.104236] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022]
Abstract
In autoimmunity, the important and fragile balance between immunity and tolerance is disturbed, resulting in abnormal immune responses to the body's own tissues and cells. CD4+CD25hiFoxP3+ regulatory T cells (Tregs) induce peripheral tolerance in vivo by means of direct cell-cell contact and release of soluble factors, or indirectly through antigen-presenting cells (APC), thereby controlling auto-reactive effector T cells. Based on these unique capacities of Tregs, preclinical studies delivered proof-of-principle for the clinical use of Tregs for the treatment of autoimmune diseases. To date, the first clinical trials using ex vivo expanded polyclonal Tregs have been completed. These pioneering studies demonstrate the feasibility of generating large numbers of polyclonal Tregs in a good manufacturing practices (GMP)-compliant manner, and that infusion of Tregs is well tolerated by patients with no evidence of general immunosuppression. Nonetheless, only modest clinical results were observed, arguing that a more antigen-specific approach might be needed to foster a durable patient-specific clinical cell therapy without the risk for general immunosuppression. In this review, we discuss current knowledge, applications and future goals of adoptive immune-modulatory Treg therapy for the treatment of autoimmune disease and transplant rejection. We describe the key advances and prospects of the potential use of T cell receptor (TCR)- and chimeric antigen receptor (CAR)-engineered Tregs in future clinical applications. These approaches could deliver the long-awaited breakthrough in stopping undesired autoimmune responses and transplant rejections.
Collapse
Affiliation(s)
- Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
21
|
Dai Z, Zhang J, Wu Q, Fang H, Shi C, Li Z, Lin C, Tang D, Wang D. Intestinal microbiota: a new force in cancer immunotherapy. Cell Commun Signal 2020; 18:90. [PMID: 32522267 PMCID: PMC7288675 DOI: 10.1186/s12964-020-00599-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer displays high levels of heterogeneity and mutation potential, and curing cancer remains a challenge that clinicians and researchers are eager to overcome. In recent years, the emergence of cancer immunotherapy has brought hope to many patients with cancer. Cancer immunotherapy reactivates the immune function of immune cells by blocking immune checkpoints, thereby restoring the anti-tumor activity of immune cells. However, immune-related adverse events are a common complication of checkpoint blockade, which might be caused by the physiological role of checkpoint pathways in regulating adaptive immunity and preventing autoimmunity. In this context, the intestinal microbiota has shown great potential in the immunotherapy of cancer. The intestinal microbiota not only regulates the immune function of the body, but also optimizes the therapeutic effect of immune checkpoint inhibitors, thus reducing the occurrence of complications. Therefore, manipulating the intestinal microbiota is expected to enhance the effectiveness of immune checkpoint inhibitors and reduce adverse reactions, which will lead to new breakthroughs in immunotherapy and cancer management. Video abstract.
Collapse
Affiliation(s)
- Zhujiang Dai
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Jingqiu Zhang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou, 225001 P. R. China
| | - Qi Wu
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Huiwen Fang
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Chunfeng Shi
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Zhen Li
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Chaobiao Lin
- Clinical Medical college, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou, 225001 P. R. China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou, 225001 P. R. China
| |
Collapse
|
22
|
Wang Y, Zhang J, Su Y, Wang C, Zhang G, Liu X, Chen Q, Lv M, Chang Y, Peng J, Hou M, Huang X, Zhang X. miRNA-98-5p Targeting IGF2BP1 Induces Mesenchymal Stem Cell Apoptosis by Modulating PI3K/Akt and p53 in Immune Thrombocytopenia. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:764-776. [PMID: 32428701 PMCID: PMC7232042 DOI: 10.1016/j.omtn.2020.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Immune thrombocytopenia (ITP) is a common hematological autoimmune disease, in which defective mesenchymal stem cells (MSCs) are potentially involved. Our previous study suggested that MSCs in ITP patients displayed enhanced apoptosis. MicroRNAs (miRNAs) play important roles in ITP by affecting megakaryopoiesis, platelet production and immunoregulation, whereas the roles of miRNAs in ITP-MSCs remain unknown. In a previous study, we performed microarray analysis to obtain mRNA and miRNA profiles of ITP-MSCs. In the present study, we reanalyze the data and identify miR-98-5p as a candidate miRNA contributing to MSC deficiency in ITP. miR-98-5p acts through targeting insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), and the subsequent downregulation of insulin-like growth factor 2 (IGF-2) causes inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which is involved in the process of MSC deficiency. Furthermore, miR-98-5p upregulates p53 by inhibiting β-transducin repeat-containing protein (β-TrCP)-dependent p53 ubiquitination. Moreover, miR-98-5p overexpression impairs the therapeutic effect of MSCs in ITP mice. All-trans retinoic acid (ATRA) protects MSCs from apoptosis by downregulating miR-98-5p, thus providing a potential therapeutic approach for ITP. Our findings demonstrate that miR-98-5p is a critical regulator of ITP-MSCs, which will help us thoroughly understand the pathogenesis of ITP.
Collapse
Affiliation(s)
- Yanan Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Jiamin Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Yan Su
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Chencong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Gaochao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Xiao Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Meng Lv
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China.
| |
Collapse
|
23
|
Looman KIM, Meel ER, Grosserichter‐Wagener C, Vissers FJM, Klingenberg JH, Jong NW, Jongste JC, Pasmans SGMA, Duijts L, Zelm MC, Moll HA. Associations of Th2, Th17, Treg cells, and IgA + memory B cells with atopic disease in children: The Generation R Study. Allergy 2020; 75:178-187. [PMID: 31385614 DOI: 10.1111/all.14010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/10/2019] [Accepted: 06/18/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND New insights into immune cells could contribute to treatment and monitoring of atopic disease. Because nongenetic factors shape the human immune system, we here studied these immune cells in a large cohort with atopic children with adjustment for prenatal and postnatal confounders. METHODS Information on atopic dermatitis, inhalant- and food-allergic sensitization, asthma lung function scores was obtained from 855 10-year-old children within the Generation R cohort. 11-color flow cytometry was performed to determine CD27+ and CD27- IgG+ , IgE+ and IgA+ memory B cells, Th1, Th2, Th17, and Treg-memory cells from venous blood. Associations between any atopic disease, the individual atopic diseases, and immune cell numbers were determined. RESULTS Children with any atopic disease had higher Th2, Treg, Treg-memory, and CD27+ IgA+ memory B-cell numbers compared to children without atopic disease. When studying the individual diseases compared to children without the individual diseases, children with atopic dermatitis, inhalant-, and food-allergic sensitization had higher memory Treg cell numbers 12.3% (95% CI 4.2; 21.0), (11.1% (95% CI 3.0; 19.8), (23.7% (95% CI 7.9; 41.8), respectively. Children with food-allergic sensitization had higher total B and CD27+ IgA+ memory B-cell numbers (15.2% [95% CI 3.2; 28.7], 22.5% [95% CI 3.9; 44.3], respectively). No associations were observed between asthma and B- or T-cell numbers. CONCLUSION Children with any atopic disease and children with inhalant- and food-allergic sensitization or atopic dermatitis had higher circulating memory Treg cells, but not higher IgE+ B-cell numbers. The associations of higher Treg and CD27+ IgA+ B-cell numbers in children with food-allergic sensitization are suggestive of TGF-β-mediated compensation for chronic inflammation.
Collapse
Affiliation(s)
- Kirsten I. M. Looman
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Evelien R. Meel
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam the Netherlands
| | | | - Floor J. M. Vissers
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Janice H. Klingenberg
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Nicolette W. Jong
- Department of Internal Medicine, Division of Allergology Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Johan C. Jongste
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | | | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Pediatrics, Division of Neonatology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Menno C. Zelm
- Department of Immunology and Pathology, Central Clinical School Monash University and Alfred Hospital Melbourne Victoria Australia
| | - Henriëtte A. Moll
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| |
Collapse
|
24
|
Nocentini G, Cari L, Riccardi C. Novel Immune Targets in Melanoma-Letter. Clin Cancer Res 2019; 25:5422-5423. [PMID: 31481484 DOI: 10.1158/1078-0432.ccr-19-1140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/09/2019] [Accepted: 06/19/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Giuseppe Nocentini
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy.
| | - Luigi Cari
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
25
|
Feng L, Yang X, Liang S, Xu Q, Miller MR, Duan J, Sun Z. Silica nanoparticles trigger the vascular endothelial dysfunction and prethrombotic state via miR-451 directly regulating the IL6R signaling pathway. Part Fibre Toxicol 2019; 16:16. [PMID: 30975181 PMCID: PMC6460825 DOI: 10.1186/s12989-019-0300-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/01/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Safety evaluation is a prerequisite for nanomaterials in a wide range of fields, including chemical industries, medicine or food sciences. Previously, we had demonstrated that SiNPs could trigger the thrombotic effects in vivo, but the underlying mechanisms remain unknown. This study was aimed to explore and verify the role of miR-451a on SiNPs-induced vascular endothelial dysfunction and pre-thrombotic state. RESULTS The color doppler ultrasound results showed that SiNPs had the inhibitory effects on aorta velocity and cardiac output. The histological and ultrastructural analysis manifested that SiNPs could induce the vascular endothelial damage. In addition, the expression level of MDA was elevated while the activity of SOD and GSH-Px were decreased in aortic arch triggered by SiNPs, accompanied with the release of iNOS and decline of eNOS in blood serum. The immunohistochemistry results showed that the positive staining of TF and PECAM-1 were elevated in a dose-dependent manner induced by SiNPs. The activation of coagulation function occurred via shortened TT, PT and APTT while the FIB was elevated markedly induced by SiNPs. Coagulant factors (TF, FXa and vWF) and PLT numbers were increased whereas the levels of anticoagulant factors (ATIII, TFPI and t-PA) were decreased. Microarray analysis showed that the down-regulated miR-451a could target the gene expression of IL6R, which further activated the JAK/STAT signaling pathway triggered by SiNPs. Dual-luciferase reporter gene assay confirmed the directly target relationship between miR-451a and IL6R. Additionally, the chemical mimics of miR-451a led to attenuate the expression of IL6R/STAT/TF signaling pathway in vitro and in vivo induced by SiNPs, while the inhibitor of miR-451a enhanced the activation of IL6R/STAT/TF signaling pathway. CONCLUSIONS In summary, SiNPs could accelerate the vascular endothelial dysfunction and prethrombotic state via miR-451a negative regulating the IL6R/STAT/TF signaling pathway.
Collapse
Affiliation(s)
- Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xiaozhe Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Qing Xu
- Core Facilities for Electrophysiology, Core Facility Center, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Mark R Miller
- University/BHF Centre for Cardiovascular Science, Queens Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People's Republic of China.
| |
Collapse
|
26
|
Romano M, Fanelli G, Albany CJ, Giganti G, Lombardi G. Past, Present, and Future of Regulatory T Cell Therapy in Transplantation and Autoimmunity. Front Immunol 2019; 10:43. [PMID: 30804926 PMCID: PMC6371029 DOI: 10.3389/fimmu.2019.00043] [Citation(s) in RCA: 368] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) are important for the induction and maintenance of peripheral tolerance therefore, they are key in preventing excessive immune responses and autoimmunity. In the last decades, several reports have been focussed on understanding the biology of Tregs and their mechanisms of action. Preclinical studies have demonstrated the ability of Tregs to delay/prevent graft rejection and to control autoimmune responses following adoptive transfer in vivo. Due to these promising results, Tregs have been extensively studied as a potential new tool for the prevention of graft rejection and/or the treatment of autoimmune diseases. Currently, solid organ transplantation remains the treatment of choice for end-stage organ failure. However, chronic rejection and the ensuing side effects of immunosuppressants represent the main limiting factors for organ acceptance and patient survival. Autoimmune disorders are chronic diseases caused by the breakdown of tolerance against self-antigens. This is triggered either by a numerical or functional Treg defect, or by the resistance of effector T cells to suppression. In this scenario, patients receiving high doses of immunosuppressant are left susceptible to life-threatening opportunistic infections and have increased risk of malignancies. In the last 10 years, a few phase I clinical trials aiming to investigate safety and feasibility of Treg-based therapy have been completed and published, whilst an increasing numbers of trials are still ongoing. The first results showed safety and feasibility of Treg therapy and phase II clinical trials are already enrolling. In this review, we describe our understanding of Tregs focussing primarily on their ontogenesis, mechanisms of action and methods used in the clinic for isolation and expansion. Furthermore, we will describe the ongoing studies and the results from the first clinical trials with Tregs in the setting of solid organ transplantation and autoimmune disorders. Finally, we will discuss strategies to further improve the success of Treg therapy.
Collapse
Affiliation(s)
- Marco Romano
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giorgia Fanelli
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Caraugh Jane Albany
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giulio Giganti
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Scuola di Specializzazione in Medicina Interna, Universita' degli Studi di Milano, Milan, Italy
| | - Giovanna Lombardi
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
27
|
Cafferata EA, Alvarez C, Diaz KT, Maureira M, Monasterio G, González FE, Covarrubias C, Vernal R. Multifunctional nanocarriers for the treatment of periodontitis: Immunomodulatory, antimicrobial, and regenerative strategies. Oral Dis 2019; 25:1866-1878. [PMID: 30565778 DOI: 10.1111/odi.13023] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/31/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022]
Abstract
Periodontitis is an inflammatory disease, in which the host immuno-inflammatory response against the dysbiotic subgingival biofilm leads to the breakdown of periodontal tissues. Most of the available treatments seem to be effective in the short-term; nevertheless, permanent periodical controls and patient compliance compromise long-term success. Different strategies have been proposed for the modulation of the host immune response as potential therapeutic tools to take a better care of most susceptible periodontitis patients, such as drug local delivery approaches. Though, maintaining an effective drug concentration for a prolonged period of time has not been achieved yet. In this context, advanced drug delivery strategies using biodegradable nanocarriers have been proposed to avoid toxicity and frequency-related problems of treatment. The versatility of distinct nanocarriers allows the improvement of their loading and release capabilities and could be potentially used for microbiological control, periodontal regeneration, and/or immunomodulation. In the present review, we revise and discuss the most frequent biodegradable nanocarrier strategies proposed for the treatment of periodontitis, including polylactic-co-glycolic acid (PLGA), chitosan, and silica-derived nanoparticles, and further suggest novel therapeutic strategies.
Collapse
Affiliation(s)
- Emilio A Cafferata
- Periodontal Biology Laboratory, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Carla Alvarez
- Periodontal Biology Laboratory, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Karla T Diaz
- School of Public Health, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Miguel Maureira
- Laboratory of Nanobiomaterials, ICOD, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Gustavo Monasterio
- Periodontal Biology Laboratory, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Fermín E González
- Laboratory of Experimental Immunology and Cancer, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Cristian Covarrubias
- Laboratory of Nanobiomaterials, ICOD, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Dentistry Unit, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
28
|
Zhou Y, Xia Z, Cheng Z, Xu G, Yang X, Liu S, Zhu Y. Inducible microRNA-590-5p inhibits host antiviral response by targeting the soluble interleukin-6 (IL6) receptor. J Biol Chem 2018; 293:18168-18179. [PMID: 30291142 DOI: 10.1074/jbc.ra118.005057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/21/2018] [Indexed: 01/09/2023] Open
Abstract
MicroRNA (miR)-590-5p has been identified as an important regulator of some signaling pathways such as cell proliferation and tumorigenesis. However, little is known about its role during viral infection. Here, we report that miR-590-5p was significantly induced by various viruses and effectively potentiated virus replication in different viral infection systems. Furthermore, miR-590-5p substantially attenuated the virus-induced expression of type I and type III interferons (IFNs) and inflammatory cytokines, resulting in impaired downstream antiviral signaling. Interleukin-6 receptor (IL6R) was identified as a target of miR-590-5p. Interestingly, the role of miR-590-5p in virus-triggered signaling was abolished in IL6R knockout cells, and this could be rescued by restoring the expression of the soluble IL6R (sIL6R) but not the membrane-bound IL6R (mIL6R), suggesting that sIL6R is indispensable for miR-590-5p in modulating the host antiviral response. Furthermore, miR-590-5p down-regulated endogenous sIL6R and mIL6R expression through a translational repression mechanism. These findings thus uncover a previously uncharacterized role and the underlying mechanism of miR-590-5p in the innate immune response to viral infection.
Collapse
Affiliation(s)
- Yaqin Zhou
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhangchuan Xia
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhikui Cheng
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Gang Xu
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Xiaodan Yang
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shi Liu
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ying Zhu
- From the State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| |
Collapse
|
29
|
Th17 and Treg lymphocytes in obesity and Type 2 diabetic patients. Clin Immunol 2018; 197:77-85. [PMID: 30218707 DOI: 10.1016/j.clim.2018.09.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/09/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022]
Abstract
Assumption that the pathogenesis of obesity-associated type 2 diabetes (T2DM) encompasses inflammation and autoimmune aspects is increasingly recognized. In the state of obesity and T2DM, the imbalance of T helper 17 (Th17) cells and regulatory T (Treg) cells are observed. These alterations reflect a loss of T cell homeostasis, which may contribute to tissue and systemic inflammation and immunity in T2DM. In this review we will discuss the accumulating data supporting the concept that Th17/Treg mediated immune responses are present in obesity-related T2DM pathogenesis, and provide evidences that restoration of Th17/Treg imbalance may be a possible therapeutic avenue for the prevention and treatment of T2DM and its complications.
Collapse
|
30
|
Inácio DP, Amado T, Silva-Santos B, Gomes AQ. Control of T cell effector functions by miRNAs. Cancer Lett 2018; 427:63-73. [PMID: 29679611 DOI: 10.1016/j.canlet.2018.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/23/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022]
Abstract
The differentiation of effector T cells is a tightly regulated process that relies on the selective expression of lineage-defining master regulators that orchestrate unique transcriptional programs, including the production of distinct sets of effector cytokines. miRNAs are post-transcriptional regulators that are now viewed as critical players in these gene expression networks and help defining cell identity and function. This review summarises the role of individual miRNAs in the regulation of the differentiation of effector T cell subsets, including CD4+ T helper cells, cytotoxic CD8+ T cells and innate-like NKT cells. Moreover, we refer to miRNAs that have been identified to affect simultaneously two or more effector T cell populations, impacting on the balance between effector T cells in vivo, thus constituting potential biomarkers or targets for therapies aiming at boosting immunity or controlling autoimmunity.
Collapse
Affiliation(s)
- Daniel P Inácio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Tiago Amado
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Anita Q Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal; Escola Superior de Tecnologia da Saúde de Lisboa, 1990-096, Lisboa, Portugal.
| |
Collapse
|
31
|
Yang JH, Eun SC. Therapeutic application of T regulatory cells in composite tissue allotransplantation. J Transl Med 2017; 15:218. [PMID: 29073905 PMCID: PMC5658973 DOI: 10.1186/s12967-017-1322-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
With growing number of cases in recent years, composite tissue allotransplantation (CTA) has been improving the quality of life of patient who seeks reconstruction and repair of damaged tissues. Composite tissue allografts are heterogeneous. They are composed of a variety of tissue types, including skin, muscle, vessel, bone, bone marrow, lymph nodes, nerve, and tendon. As a primary target of CTA, skin has high antigenicity with a rich repertoire of resident cells that play pivotal roles in immune surveillance. In this regard, understanding the molecular mechanisms involved in immune rejection in the skin would be essential to achieve successful CTA. Although scientific evidence has proved the necessity of immunosuppressive drugs to prevent rejection of allotransplanted tissues, there remains a lingering dilemma due to the lack of specificity of targeted immunosuppression and risks of side effects. A cumulative body of evidence has demonstrated T regulatory (Treg) cells have critical roles in induction of immune tolerance and immune homeostasis in preclinical and clinical studies. Presently, controlling immune susceptible characteristics of CTA with adoptive transfer of Treg cells is being considered promising and it has drawn great interests. This updated review will focus on a dominant form of Treg cells expressing CD4+CD25+ surface molecules and a forkhead box P3 transcription factor with immune tolerant and immune homeostasis activities. For future application of Treg cells as therapeutics in CTA, molecular and cellular characteristics of CTA and immune rejection, Treg cell development and phenotypes, Treg cell plasticity and stability, immune tolerant functions of Treg cells in CTA in preclinical studies, and protocols for therapeutic application of Treg cells in clinical settings are addressed in this review. Collectively, Treg cell therapy in CTA seems feasible with promising perspectives. However, the extreme high immunogenicity of CTA warrants caution.
Collapse
Affiliation(s)
- Jeong-Hee Yang
- Department of Plastic and Reconstructive Surgery, Composite Tissue Allotransplantation Immunology Laboratory, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seok-Chan Eun
- Department of Plastic and Reconstructive Surgery, Composite Tissue Allotransplantation Immunology Laboratory, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
32
|
Song Y, Yang JM. Role of interleukin (IL)-17 and T-helper (Th)17 cells in cancer. Biochem Biophys Res Commun 2017; 493:1-8. [PMID: 28859982 DOI: 10.1016/j.bbrc.2017.08.109] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/27/2017] [Indexed: 12/18/2022]
Abstract
Interleukin-17 (IL-17), a pleiotropic proinflammatory cytokine, is reported to be significantly generated by a distinct subset of CD4+ T-cells, upgrading cancer-elicited inflammation and preventing cancer cells from immune surveillance. T-helper (Th)17 cells produced from naive CD4+ T cells have recently been renowned and generally accepted, gaining eminence in cancer studies and playing the effective role in context of cancer. Th17 cells are the main source of IL-17-secreting cells, It was found that other cell types produced this cytokine as well, including Group 3 innate lymphoid cells (ILC3), δγT cells, invariant natural killer T (iNKT) cells, lymphoid-tissue inducer (LTi)-like cells and Natural killer (NK) cells. Th17-associated cytokines give impetus to tumor progression, or inducing angiogenesis and metastasis. This review demonstrates an understanding on how the pro- or antitumor function of Th17 cells and IL-17 may change cancer progression, leading to the appearance of complex and pivotal biologic activities in tumor.
Collapse
Affiliation(s)
- Yang Song
- Department of Otorhinolaryngology, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China.
| | - Jian Ming Yang
- Department of Otorhinolaryngology, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| |
Collapse
|