51
|
Abstract
Hundreds of microRNAs (miRNAs) are expressed in distinct spatial and temporal patterns during embryonic and postnatal mouse development. The loss of all miRNAs through the deletion of critical miRNA biogenesis factors results in early lethality. The function of each miRNA stems from their cumulative negative regulation of multiple mRNA targets expressed in a particular cell type. During development, miRNAs often coordinate the timing and direction of cell fate transitions. In adults, miRNAs frequently contribute to organismal fitness through homeostatic roles in physiology. Here, we review how the recent dissection of miRNA-knockout phenotypes in mice as well as advances related to their targets, dosage, and interactions have collectively informed our understanding of the roles of miRNAs in mammalian development and adaptive responses.
Collapse
|
52
|
Papadaki C, Thomopoulou K, Monastirioti A, Koronakis G, Papadaki MA, Rounis K, Vamvakas L, Nikolaou C, Mavroudis D, Agelaki S. MicroRNAs Regulating Tumor and Immune Cell Interactions in the Prediction of Relapse in Early Stage Breast Cancer. Biomedicines 2021; 9:biomedicines9040421. [PMID: 33924670 PMCID: PMC8069787 DOI: 10.3390/biomedicines9040421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/10/2021] [Accepted: 04/11/2021] [Indexed: 11/20/2022] Open
Abstract
MicroRNAs (miRNAs) are involved in the regulation of immune response and hold an important role in tumor immune escape. We investigated the differential expression of the immunomodulatory miR-10b, miR-19a, miR-20a, miR-126, and miR-155 in the plasma of healthy women and patients with early stage breast cancer and interrogated their role in the prediction of patients’ relapse. Blood samples were obtained from healthy women (n = 20) and patients with early stage breast cancer (n = 140) before adjuvant chemotherapy. Plasma miRNA expression levels were assessed by RT-qPCR. Relapse predicting models were developed using binary logistic regression and receiver operating curves (ROC) were constructed to determine miRNA sensitivity and specificity. Only miR-155 expression was lower in patients compared with healthy women (p = 0.023), whereas miR-155 and miR-10b were lower in patients who relapsed compared with healthy women (p = 0.039 and p = 0.002, respectively). MiR-155 expression combined with axillary lymph node infiltration and tumor grade demonstrated increased capability in distinguishing relapsed from non-relapsed patients [(area under the curve, (AUC = 0.861; p < 0.001)]. Combined miR-19a and miR-20a expression had the highest performance in discriminating patients with early relapse (AUC = 0.816; p < 0.001). Finally, miR-10b in combination with lymph node status and grade had the highest accuracy to discriminate patients with late relapse (AUC = 0.971; p < 0.001). The robustness of the relapse predicting models was further confirmed in a 10-fold cross validation. Deregulation of circulating miRNAs involved in tumor-immune interactions may predict relapse in early stage breast cancer. Their successful clinical integration could potentially address the significance challenge of treatment escalation or de-escalation according to the risk of recurrence.
Collapse
Affiliation(s)
- Chara Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Vassilika Vouton, 71003 Crete, Greece; (C.P.); (A.M.); (M.A.P.); (D.M.)
| | - Konstantina Thomopoulou
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Crete, Greece; (K.T.); (G.K.); (K.R.); (L.V.)
| | - Alexia Monastirioti
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Vassilika Vouton, 71003 Crete, Greece; (C.P.); (A.M.); (M.A.P.); (D.M.)
| | - George Koronakis
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Crete, Greece; (K.T.); (G.K.); (K.R.); (L.V.)
| | - Maria A. Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Vassilika Vouton, 71003 Crete, Greece; (C.P.); (A.M.); (M.A.P.); (D.M.)
| | - Konstantinos Rounis
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Crete, Greece; (K.T.); (G.K.); (K.R.); (L.V.)
| | - Lambros Vamvakas
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Crete, Greece; (K.T.); (G.K.); (K.R.); (L.V.)
| | - Christoforos Nikolaou
- Department of Biology, University of Crete, Heraklion, Vassilika Vouton, 70013 Crete, Greece;
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation of Research and Technology (FORTH), Heraklion, Vassilika Vouton, 70013 Crete, Greece
- Biomedical Science Research Center “Alexander Fleming”, Institute of Bioinnovation, 16672 Athens, Greece
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Vassilika Vouton, 71003 Crete, Greece; (C.P.); (A.M.); (M.A.P.); (D.M.)
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Crete, Greece; (K.T.); (G.K.); (K.R.); (L.V.)
| | - Sofia Agelaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Vassilika Vouton, 71003 Crete, Greece; (C.P.); (A.M.); (M.A.P.); (D.M.)
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Crete, Greece; (K.T.); (G.K.); (K.R.); (L.V.)
- Correspondence: ; Tel.: +30-281-039-2438
| |
Collapse
|
53
|
Abstract
B cells constitute a main branch adaptive immune system. They mediate host defence through the production of high-affinity antibodies against an enormous diversity of foreign antigens. Remarkably, B cells undergo multiple types of somatic DNA mutation to achieve this effector function, including class switch recombination (CSR) and somatic hypermutation (SHM). These processes occur in response to antigen recognition and inflammatory signals, and require strict biological control at multiple levels. Transcription within the locus that encodes antibodies plays direct roles in CSR. Additional non-coding RNAs (ncRNAs), including both microRNAs (miRNAs) and long ncRNAs (lncRNAs), also play pivotal roles in B cell activation and terminal effector function through post-transcriptional gene regulation and chromatin remodelling, respectively.
Collapse
Affiliation(s)
- Eric J Wigton
- Department of Microbiology & Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA, USA
| | - K Mark Ansel
- Department of Microbiology & Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
54
|
Yan J, Zhang Y, Su Y, Tian L, Qin P, Xu X, Zhou Y. microRNA-125a targets MAVS and TRAF6 to modulate interferon signaling and promote HCV infection. Virus Res 2021; 296:198336. [PMID: 33577860 DOI: 10.1016/j.virusres.2021.198336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/30/2020] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
Hepatitis C virus (HCV) can cause chronic lifelong infections in humans, resulting in sustained hepatic inflammation, liver cirrhosis, and hepatocellular carcinoma. The clearance of HCV infections is dependent upon effective and coordinated innate and adaptive antiviral immune responses. However, HCV has evolved a range of strategies that enable it to evade or overcome the host immune response, enabling the virus to persist in susceptible hosts through mechanisms that remain to be fully clarified. Herein, we describe a novel mechanism whereby HCV can evade immune surveillance by activating microRNA (miR)-125a. Hepatocytes upregulate miR-125a following HCV infection, and serum from HCV-infected patients similarly exhibits the upregulation of this miRNA. We found that miR-125a is able to target and suppress the expression of two key genes associated with the interferon (IFN) signaling pathway - mitochondrial antiviral signaling (MAVS) and TNF receptor-associated factor 6 (TRAF6). Disrupting the expression of these genes can in turn compromise type I IFN responses to HCV. Together, our data reveal that HCV infection results in the upregulation of miR-125a, which negatively regulates IFN signaling via inhibiting the expression of MAVS and TRAF6, thereby enabling the virus to evade innate antiviral immunity. Targeting this pathway may thus represent an efficient approach to treating HCV and bolstering antiviral immune responses in infected patients.
Collapse
Affiliation(s)
- Jianguo Yan
- Department of Physiology, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Yuting Zhang
- Department of Microbiology, Guilin Medical University, Guilin, 541004, Guangxi, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Yan Su
- Department of Physiology, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Linlin Tian
- Department of Microbiology, Guilin Medical University, Guilin, 541004, Guangxi, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Peifang Qin
- Department of Microbiology, Guilin Medical University, Guilin, 541004, Guangxi, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Xingfeng Xu
- Department of Physiology, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Yali Zhou
- Department of Microbiology, Guilin Medical University, Guilin, 541004, Guangxi, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541004, Guangxi, China.
| |
Collapse
|
55
|
Increased let-7b-5p is associated with enhanced BAFF-R expression and B cell survival in immune thrombocytopenia. Int Immunopharmacol 2021; 93:107393. [PMID: 33529914 DOI: 10.1016/j.intimp.2021.107393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND B cells play a key role in the pathogenesis of immune thrombocytopenia (ITP) by producing platelet autoantibodies. Accumulating evidence suggest that microRNA (miRNA) is a critical regulator in B cells. The contribution of miRNA to B cell dysfunction in ITP has not been described. The aim of this study was to examine the expression of miRNA let-7b-5p in B cells of ITP patients and investigate its possible association with B cell function in ITP. METHODS The CD19+ cells were isolated from peripheral mononuclear cells of ITP patients and healthy controls using immunomagnetic microbeads. B cell survival in vitro was evaluated by cell counting. The level of let-7b-5p was quantified by quantitative PCR. The surface expression of B cell activating factor receptor (BAFF-R) was detected by flow cytometry. The role of let-7b-5p was examined in isolated B cells by transfecting miRNA mimics or inhibitors. RESULTS The results showed that let-7b-5p in B cells was elevated, and B cell survival was enhanced in ITP patients compared with healthy controls. BAFF and B cell receptor stimulation can induce the expression of let-7b-5p in vitro. Overexpression of let-7b-5p in B cells enhanced the expression of surface BAFF-R and promoted B cell survival. Moreover, let-7b-5p enhanced the phosphorylation of NF-κB2 p100 and upregulated the expression of survival factor Bcl-xL after BAFF induction. CONCLUSION Let-7b-5p is a pro-survival miRNA in B cells and increased let-7b-5p is associated with enhanced surface BAFF-R in ITP.
Collapse
|
56
|
Anderson G, Maes M. Mitochondria and immunity in chronic fatigue syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2020; 103:109976. [PMID: 32470498 DOI: 10.1016/j.pnpbp.2020.109976] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
It is widely accepted that the pathophysiology and treatment of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) could be considerably improved. The heterogeneity of ME/CFS and the confusion over its classification have undoubtedly contributed to this, although this would seem a consequence of the complexity of the array of ME/CFS presentations and high levels of diverse comorbidities. This article reviews the biological underpinnings of ME/CFS presentations, including the interacting roles of the gut microbiome/permeability, endogenous opioidergic system, immune cell mitochondria, autonomic nervous system, microRNA-155, viral infection/re-awakening and leptin as well as melatonin and the circadian rhythm. This details not only relevant pathophysiological processes and treatment options, but also highlights future research directions. Due to the complexity of interacting systems in ME/CFS pathophysiology, clarification as to its biological underpinnings is likely to considerably contribute to the understanding and treatment of other complex and poorly managed conditions, including fibromyalgia, depression, migraine, and dementia. The gut and immune cell mitochondria are proposed to be two important hubs that interact with the circadian rhythm in driving ME/CFS pathophysiology.
Collapse
Affiliation(s)
- G Anderson
- CRC Scotland & London, Eccleston Square, London, UK.
| | - M Maes
- Dept Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Dept Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria.; IMPACT Research Center, Deakin University, Geelong, Australia
| |
Collapse
|
57
|
Hines MJ, Coffre M, Mudianto T, Panduro M, Wigton EJ, Tegla C, Osorio-Vasquez V, Kageyama R, Benhamou D, Perez O, Bajwa S, McManus MT, Ansel KM, Melamed D, Koralov SB. miR-29 Sustains B Cell Survival and Controls Terminal Differentiation via Regulation of PI3K Signaling. Cell Rep 2020; 33:108436. [PMID: 33264610 DOI: 10.1016/j.celrep.2020.108436] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 10/01/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling cascade downstream of the B cell receptor (BCR) signalosome is essential for B cell maturation. Proper signaling strength is maintained through the PI3K negative regulator phosphatase and tensin homolog (PTEN). Although a role for microRNA (miRNA)-dependent control of the PTEN-PI3K axis has been described, the contribution of individual miRNAs to the regulation of this crucial signaling modality in mature B lymphocytes remains to be elucidated. Our analyses reveal that ablation of miR-29 specifically in B lymphocytes results in an increase in PTEN expression and dampening of the PI3K pathway in mature B cells. This dysregulation has a profound impact on the survival of B lymphocytes and results in increased class switch recombination and decreased plasma cell differentiation. Furthermore, we demonstrate that ablation of one copy of Pten is sufficient to ameliorate the phenotypes associated with miR-29 loss. Our data suggest a critical role for the miR-29-PTEN-PI3K regulatory axis in mature B lymphocytes.
Collapse
Affiliation(s)
- Marcus J Hines
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Maryaline Coffre
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Tenny Mudianto
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Marisella Panduro
- Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, UCSF, San Francisco, CA 94143, USA
| | - Eric J Wigton
- Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, UCSF, San Francisco, CA 94143, USA
| | - Cosmin Tegla
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Robin Kageyama
- Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, UCSF, San Francisco, CA 94143, USA
| | - David Benhamou
- Department of Immunology, Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Oriana Perez
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Sofia Bajwa
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Michael T McManus
- Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA; Diabetes Center, UCSF, San Francisco, CA 94143, USA
| | - K Mark Ansel
- Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, UCSF, San Francisco, CA 94143, USA
| | - Doron Melamed
- Department of Immunology, Faculty of Medicine, Technion, Haifa 31096, Israel.
| | - Sergei B Koralov
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
58
|
Singh A, Srivastava N, Yadav A, Ateeq B. Targeting AGTR1/NF-κB/CXCR4 axis by miR-155 attenuates oncogenesis in glioblastoma. Neoplasia 2020; 22:497-510. [PMID: 32896760 PMCID: PMC7481885 DOI: 10.1016/j.neo.2020.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM) represents the most aggressive malignancy of the central nervous system. Increased expression of Angiotensin II Receptor Type 1 (AGTR1) has been associated with proliferative and infiltrative properties of glioma cells. However, the underlying mechanism of AGTR1 upregulation in GBM is still unexplored. To understand the post-transcriptional regulation of AGTR1 in GBM, we screened 3'untranslated region (3'UTR) of AGTR1 for putative miRNA binding by using prediction algorithms. Interestingly, miR-155 showed conserved binding on the 3'UTR of AGTR1, subsequently confirmed by luciferase reporter assay. Furthermore, miR-155 overexpressing GBM cells show decrease in AGTR1 expression accompanied with reduced cell proliferation, invasion, foci formation and anchorage-independent growth. Strikingly, immunodeficient mice implanted with stable miR-155 overexpressing SNB19 cells show negligible tumor growth. Notably, miR-155 attenuates NF-κB signaling downstream of AGTR1 leading to reduced CXCR4 as well as AGTR1 levels. Mechanistically, miR-155 mitigates AGTR1-mediated angiogenesis, epithelial-to-mesenchymal transition, stemness, and MAPK signaling. Similar effects were observed by using pharmacological inhibitor of IκB Kinase (IKK) complex in multiple cell-based assays. Taken together, we established that miRNA-155 post-transcriptionally regulates AGTR1 expression, abrogates AGTR1/NF-κB/CXCR4 signaling axis and elicits pleiotropic anticancer effects in GBM. This study opens new avenues for using IKK inhibitors and miRNA-155 replacement therapies for the treatment of AGTR1-positive malignancies.
Collapse
MESH Headings
- Animals
- Apoptosis
- Brain Neoplasms/genetics
- Brain Neoplasms/metabolism
- Brain Neoplasms/pathology
- Cell Movement
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Glioblastoma/genetics
- Glioblastoma/metabolism
- Glioblastoma/pathology
- Humans
- Mice
- Mice, Inbred NOD
- Mice, SCID
- MicroRNAs/genetics
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Anukriti Singh
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India; Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Nidhi Srivastava
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Anjali Yadav
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India
| | - Bushra Ateeq
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India; Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, U.P., India.
| |
Collapse
|
59
|
Kunze-Schumacher H, Krueger A. The Role of MicroRNAs in Development and Function of Regulatory T Cells - Lessons for a Better Understanding of MicroRNA Biology. Front Immunol 2020; 11:2185. [PMID: 33013919 PMCID: PMC7509487 DOI: 10.3389/fimmu.2020.02185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as critical posttranscriptional regulators of the immune system, including function and development of regulatory T (Treg) cells. Although this critical role has been firmly demonstrated through genetic models, key mechanisms of miRNA function in vivo remain elusive. Here, we review the role of miRNAs in Treg cell development and function. In particular, we focus on the question what the study of miRNAs in this context reveals about miRNA biology in general, including context-dependent function and the role of individual targets vs. complex co-targeting networks. In addition, we highlight potential technical pitfalls and state-of-the-art approaches to improve the mechanistic understanding of miRNA biology in a physiological context.
Collapse
Affiliation(s)
- Heike Kunze-Schumacher
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Andreas Krueger
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
60
|
Fuertes T, Ramiro AR, de Yebenes VG. miRNA-Based Therapies in B Cell Non-Hodgkin Lymphoma. Trends Immunol 2020; 41:932-947. [PMID: 32888820 DOI: 10.1016/j.it.2020.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
Abstract
Non-Hodgkin lymphoma (NHL) is a diverse class of hematological cancers, many of which arise from germinal center (GC)-experienced B cells. Thus GCs, the sites of antibody affinity maturation triggered during immune responses, also provide an environment that facilitates B cell oncogenic transformation. miRNAs provide attractive and mechanistically different strategies to treat these malignancies based on their potential for simultaneous modulation of multiple targets. Here, we discuss the scientific rationale for miRNA-based therapeutics in B cell neoplasias and review recent advances that may help establish a basis for novel candidate miRNA-based therapies for B cell-NHL (B-NHL).
Collapse
Affiliation(s)
- Teresa Fuertes
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Virginia G de Yebenes
- Universidad Complutense de Madrid School of Medicine, Department of Immunology, Ophthalmology and ENT, 12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| |
Collapse
|
61
|
Labi V, Derudder E. Cell signaling and the aging of B cells. Exp Gerontol 2020; 138:110985. [PMID: 32504658 DOI: 10.1016/j.exger.2020.110985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/17/2020] [Accepted: 05/29/2020] [Indexed: 12/24/2022]
Abstract
The uniqueness of each B cell lies in the structural diversity of the B-cell antigen receptor allowing the virtually limitless recognition of antigens, a necessity to protect individuals against a range of challenges. B-cell development and response to stimulation are exquisitely regulated by a group of cell surface receptors modulating various signaling cascades and their associated genetic programs. The effects of these signaling pathways in optimal antibody-mediated immunity or the aberrant promotion of immune pathologies have been intensely researched in the past in young individuals. In contrast, we are only beginning to understand the contribution of these pathways to the changes in B cells of old organisms. Thus, critical transcription factors such as E2A and STAT5 show differential expression or activity between young and old B cells. As a result, B-cell physiology appears altered, and antibody production is impaired. Here, we discuss selected phenotypic changes during B-cell aging and attempt to relate them to alterations of molecular mechanisms.
Collapse
Affiliation(s)
- Verena Labi
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria.
| | - Emmanuel Derudder
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
62
|
Anderson G, Carbone A, Mazzoccoli G. Aryl Hydrocarbon Receptor Role in Co-Ordinating SARS-CoV-2 Entry and Symptomatology: Linking Cytotoxicity Changes in COVID-19 and Cancers; Modulation by Racial Discrimination Stress. BIOLOGY 2020; 9:E249. [PMID: 32867244 PMCID: PMC7564943 DOI: 10.3390/biology9090249] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
There is an under-recognized role of the aryl hydrocarbon receptor (AhR) in co-ordinating the entry and pathophysiology of the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) that underpins the COVID-19 pandemic. The rise in pro-inflammatory cytokines during the 'cytokine storm' induce indoleamine 2,3-dioxygenase (IDO), leading to an increase in kynurenine that activates the AhR, thereby heightening the initial pro-inflammatory cytokine phase and suppressing the endogenous anti-viral response. Such AhR-driven changes underpin the heightened severity and fatality associated with pre-existent high-risk medical conditions, such as type II diabetes, as well as to how racial discrimination stress contributes to the raised severity/fatality in people from the Black Asian and Minority Ethnic (BAME) communities. The AhR is pivotal in modulating mitochondrial metabolism and co-ordinating specialized, pro-resolving mediators (SPMs), the melatonergic pathways, acetyl-coenzyme A, and the cyclooxygenase (COX) 2-prostaglandin (PG) E2 pathway that underpin 'exhaustion' in the endogenous anti-viral cells, paralleling similar metabolic suppression in cytolytic immune cells that is evident across all cancers. The pro-inflammatory cytokine induced gut permeability/dysbiosis and suppression of pineal melatonin are aspects of the wider pathophysiological underpinnings regulated by the AhR. This has a number of prophylactic and treatment implications for SARS-CoV-2 infection and cancers and future research directions that better investigate the biological underpinnings of social processes and how these may drive health disparities.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London SW1V 1PB, UK;
| | - Annalucia Carbone
- Division of Internal Medicine and Chronobiology Laboratory, Department of Medical Sciences, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Gianluigi Mazzoccoli
- Division of Internal Medicine and Chronobiology Laboratory, Department of Medical Sciences, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, 71013 Foggia, Italy;
| |
Collapse
|
63
|
MicroRNA miR-181-A Rheostat for TCR Signaling in Thymic Selection and Peripheral T-Cell Function. Int J Mol Sci 2020; 21:ijms21176200. [PMID: 32867301 PMCID: PMC7503384 DOI: 10.3390/ijms21176200] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/17/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
The selection of T cells during intra-thymic d evelopment is crucial to obtain a functional and simultaneously not self-reactive peripheral T cell repertoire. However, selection is a complex process dependent on T cell receptor (TCR) thresholds that remain incompletely understood. In peripheral T cells, activation, clonal expansion, and contraction of the active T cell pool, as well as other processes depend on TCR signal strength. Members of the microRNA (miRNA) miR-181 family have been shown to be dynamically regulated during T cell development as well as dependent on the activation stage of T cells. Indeed, it has been shown that expression of miR-181a leads to the downregulation of multiple phosphatases, implicating miR-181a as ‘‘rheostat’’ of TCR signaling. Consistently, genetic models have revealed an essential role of miR-181a/b-1 for the generation of unconventional T cells as well as a function in tuning TCR sensitivity in peripheral T cells during aging. Here, we review these broad roles of miR-181 family members in T cell function via modulating TCR signal strength.
Collapse
|
64
|
Nelson MC, O'Connell RM. MicroRNAs: At the Interface of Metabolic Pathways and Inflammatory Responses by Macrophages. Front Immunol 2020; 11:1797. [PMID: 32922393 PMCID: PMC7456828 DOI: 10.3389/fimmu.2020.01797] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages are key cells of the innate immune system with functional roles in both homeostatic maintenance of self-tissues and inflammatory responses to external stimuli, including infectious agents. Recent advances in metabolic research have revealed that macrophage functions rely upon coordinated metabolic programs to regulate gene expression, inflammation, and other important cellular processes. Polarized macrophages adjust their use of nutrients such as glucose and amino acids to meet their changing metabolic needs, and this in turn supports the functions of the activated macrophage. Metabolic and inflammatory processes have been widely studied, and a crucial role for their regulation at the post-transcriptional level by microRNAs (miRNAs) has been identified. miRNAs govern many facets of macrophage biology, including direct targeting of metabolic regulators and inflammatory pathways. This review will integrate emerging data that support an interplay between miRNAs and metabolism during macrophage inflammatory responses, highlighting critical miRNAs and miRNA families. Additionally, we will address the implications of these networks for human disease and discuss emerging areas of research in this field.
Collapse
Affiliation(s)
- Morgan C Nelson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
65
|
Gomes C, Sequeira C, Barbosa M, Cunha C, Vaz AR, Brites D. Astrocyte regional diversity in ALS includes distinct aberrant phenotypes with common and causal pathological processes. Exp Cell Res 2020; 395:112209. [PMID: 32739211 DOI: 10.1016/j.yexcr.2020.112209] [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: 06/08/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022]
Abstract
Astrocytes are major contributors of motor neuron (MN) degeneration in amyotrophic lateral sclerosis (ALS). We investigated whether regional and cell maturation differences influence ALS astrocyte malfunction. Spinal and cortical astrocytes from SOD1G93A (mSOD1) 7-day-old mice were cultured for 5 and 13 days in vitro (DIV). Astrocyte aberrancies predominated in 13DIV cells with region specificity. 13DIV cortical mSOD1 astrocytes showed early morphological changes and a predominant reactive and inflammatory phenotype, while repressed proteins and genes were found in spinal cells. Inflammatory-associated miRNAs, e.g. miR-155/miR-21/miR-146a, were downregulated in the first and upregulated in the later ones. Interestingly, depleted miR-155/miR-21/miR-146a in small extracellular vesicles (sEVs/exosomes) was a common pathological feature. Cortical mSOD1 astrocytes induced late apoptosis and kinesin-1 downregulation in mSOD1 NSC-34 MNs, whereas spinal cells upregulated dynein, while decreased nNOS and synaptic-related genes. Both regional-distinct mSOD1 astrocytes enhanced iNOS gene expression in mSOD1 MNs. We provide information on the potential contribution of astrocytes to ALS bulbar-vs. spinal-onset pathology, local influence on neuronal dysfunction and their shared miRNA-depleted exosome trafficking. These causal and common features may have potential therapeutic implications in ALS. Future studies should clarify if astrocyte-derived sEVs are active players in ALS-related neuroinflammation and glial activation.
Collapse
Affiliation(s)
- Cátia Gomes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Catarina Sequeira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Marta Barbosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Carolina Cunha
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Ana Rita Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
66
|
MicroRNAs as Guardians of the Prostate: Those Who Stand before Cancer. What Do We Really Know about the Role of microRNAs in Prostate Biology? Int J Mol Sci 2020; 21:ijms21134796. [PMID: 32645914 PMCID: PMC7370012 DOI: 10.3390/ijms21134796] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/29/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is the second leading cause of cancer-related deaths of men in the Western world. Despite recent advancement in genomics, transcriptomics and proteomics to understand prostate cancer biology and disease progression, castration resistant metastatic prostate cancer remains a major clinical challenge and often becomes incurable. MicroRNAs (miRNAs), about 22-nucleotide-long non-coding RNAs, are a group of regulatory molecules that mainly work through post-transcriptional gene silencing via translational repression. Expression analysis studies have revealed that miRNAs are aberrantly expressed in cancers and have been recognized as regulators of prostate cancer progression. In this critical review, we provide an analysis of reported miRNA functions and conflicting studies as they relate to expression levels of specific miRNAs and prostate cancer progression; oncogenic and/or tumor suppressor roles; androgen receptor signaling; epithelial plasticity; and the current status of diagnostic and therapeutic applications. This review focuses on select miRNAs, highly expressed in normal and cancer tissue, to emphasize the current obstacles faced in utilizing miRNA data for significant impacts on prostate cancer therapeutics.
Collapse
|
67
|
Yi L, Chen Y, Jin Q, Deng C, Wu Y, Li H, Liu T, Li Y, Yang Y, Wang J, Lv Q, Zhang L, Xie M. Antagomir-155 Attenuates Acute Cardiac Rejection Using Ultrasound Targeted Microbubbles Destruction. Adv Healthc Mater 2020; 9:e2000189. [PMID: 32548962 DOI: 10.1002/adhm.202000189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/27/2020] [Indexed: 12/22/2022]
Abstract
Antagomir-155 is an artificial inhibitor of miRNA-155, which is expected to be a promising therapeutic target to attenuate acute cardiac rejection (ACR). However, its vulnerability of being degraded by endogenous nuclease and potential off-target effect make the authors seek for a more suitable way to delivery it. In attribution of efficiency and safety, ultrasound targeted microbubbles destruction (UTMD) turns out to be an appropriate method to deliver gene to target tissues. Here, cationic microbubbles to deliver antagomir-155 downregulating miRNA-155 in murine allograft hearts triggered by UTMD are synthesized. The viability of this therapy is verified by fluorescent microscopy. The biodistribution of antagomir-155 is analyzed by optical imaging system. The results show antagomir-155 delivered by UTMD which significantly decreases the levels of miR-155. Also, this therapy downregulates the expression of cytokines and inflammation infiltration. And allograft survival time is significantly prolonged. Therefore, antagomir-loaded microbubbles trigged by UTMD may provide a novel platform for ACR target treatment.
Collapse
Affiliation(s)
- Luyang Yi
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Yihan Chen
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Qiaofeng Jin
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Cheng Deng
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Ya Wu
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Huiling Li
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Tianshu Liu
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Yuman Li
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Yali Yang
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Jing Wang
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Qing Lv
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Li Zhang
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Mingxing Xie
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| |
Collapse
|
68
|
Bhargava A, Kumari R, Khare S, Shandilya R, Gupta PK, Tiwari R, Rahman A, Chaudhury K, Goryacheva IY, Mishra PK. Mapping the Mitochondrial Regulation of Epigenetic Modifications in Association With Carcinogenic and Noncarcinogenic Polycyclic Aromatic Hydrocarbon Exposure. Int J Toxicol 2020; 39:465-476. [PMID: 32588678 DOI: 10.1177/1091581820932875] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) refer to a ubiquitous group of anthropogenic air pollutants that are generated through incomplete carbon combustion. Although the immunotoxic nature of PAHs has been previously reported, the underlying molecular mechanisms of this effect are not fully understood. In the present study, we investigated the mitochondrial-mediated epigenetic regulation of 2 PAHs, carcinogenic (benzo[a]pyrene; BaP) and noncarcinogenic (anthracene [ANT]), in peripheral lymphocytes. While ANT exposure triggered mitochondrial oxidative damage, no appreciable epigenetic modifications were observed. On the other hand, exposure to BaP perturbed the mitochondrial redox machinery and initiated cascade of epigenetic modifications. Cells exposed to BaP showed prominent changes in the expression of mitochondrial microRNAs (miR-24, miR-34a, miR-150, and miR-155) and their respective gene targets (NF-κβ, MYC, and p53). The exposure of BaP also caused significant alterations in the expression of epigenetic modifiers (DNMT1, HDAC1, HDAC7, KDM3a, EZH2, and P300) and hypomethylation within nuclear and mitochondrial DNA. This further induced methylation of histone tails, which play a crucial role in the regulation of chromatin structure. Overall, our study provides novel mechanistic insights into the mitochondrial regulation of epigenetic modifications in association with PAH-induced immunotoxicity.
Collapse
Affiliation(s)
- Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Roshani Kumari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Surbhi Khare
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Ruchita Shandilya
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pushpendra Kumar Gupta
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Rajnarayan Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Akhlaqur Rahman
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Koel Chaudhury
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, India
| | - Irina Yu Goryacheva
- Department of General and Inorganic Chemistry, Saratov State University, Saratov, Russia
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
69
|
Chillappagari S, Garapati V, Mahavadi P, Naehrlich L, Schmeck BT, Schmitz ML, Guenther A. Defective BACH1/HO-1 regulatory circuits in cystic fibrosis bronchial epithelial cells. J Cyst Fibros 2020; 20:140-148. [PMID: 32534959 DOI: 10.1016/j.jcf.2020.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/01/2020] [Accepted: 05/23/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The stress-regulated enzyme hemeoxygenase-1 (HO-1) contributes to the cell response towards inflammation and oxidative stress. We previously reported on curtailed HO-1 expression in cystic fibrosis (CF) bronchial epithelial (CFBE41o-) cells and CF-mice, but the molecular mechanisms for this are not known. Here, we compared healthy and CF bronchial epithelial cells for regulatory circuits controlling HO-1 protein levels. METHODS In this study, we employed immunohistochemistry on CF and healthy lung sections to examine the BACH1 protein expression. Alteration of BACH1 protein levels in 16HBE14o- and CFBE41o- cells was achieved by using either siRNA-mediated knockdown of BACH1 or by increasing miRNA-155 levels. HO-1 luciferase reporter assay was chosen to examine the downstream affects after BACH1 modulation. RESULTS Human CF lungs and cells showed increased levels of the HO-1 transcriptional repressor, BACH1, and increased miR-155 expression. Knockdown studies using BACH1 siRNA and overexpression of miR-155 did not significantly rescue HO-1 expression in CFBE41o- cells. Elevated BACH1 expression detected in CF cells was refractory to the inhibitory function of miR-155 and was instead due to increased protein stability. CONCLUSION We observed defects in the inhibitory activities of miR-155 and BACH1 on HO-1 expression in CF cells. Thus various defective regulatory loops account for dysregulated BACH1 expression in CF, which in turn may contribute to low HO-1 levels.
Collapse
Affiliation(s)
- Shashipavan Chillappagari
- Institute of Biochemistry, Justus-Liebig-University, D-35392, Giessen, Germany; Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany.
| | - Virajith Garapati
- Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Lutz Naehrlich
- University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany; Department of Pediatrics, Justus Liebig University, Giessen, Feulgenstrasse 12, 35392 Giessen, Germany
| | - Bernd T Schmeck
- University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany; Institute for Lung Research, Department of Respiratory and Critical Care Medicine, University Medical Center Marburg, Center for Synthetic Microbiology (SYNMIKRO), Philipps-University, Marburg, Germany, Member of the German Center for Lung Research (DZL), and the German Center of Infection Research (DZIF), Marburg, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University, D-35392, Giessen, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany; Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| |
Collapse
|
70
|
Li X, Pritykin Y, Concepcion CP, Lu Y, La Rocca G, Zhang M, King B, Cook PJ, Au YW, Popow O, Paulo JA, Otis HG, Mastroleo C, Ogrodowski P, Schreiner R, Haigis KM, Betel D, Leslie CS, Ventura A. High-Resolution In Vivo Identification of miRNA Targets by Halo-Enhanced Ago2 Pull-Down. Mol Cell 2020; 79:167-179.e11. [PMID: 32497496 DOI: 10.1016/j.molcel.2020.05.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 12/19/2022]
Abstract
The identification of microRNA (miRNA) targets by Ago2 crosslinking-immunoprecipitation (CLIP) methods has provided major insights into the biology of this important class of non-coding RNAs. However, these methods are technically challenging and not easily applicable to an in vivo setting. To overcome these limitations and facilitate the investigation of miRNA functions in vivo, we have developed a method based on a genetically engineered mouse harboring a conditional Halo-Ago2 allele expressed from the endogenous Ago2 locus. By using a resin conjugated to the HaloTag ligand, Ago2-miRNA-mRNA complexes can be purified from cells and tissues expressing the endogenous Halo-Ago2 allele. We demonstrate the reproducibility and sensitivity of this method in mouse embryonic stem cells, developing embryos, adult tissues, and autochthonous mouse models of human brain and lung cancers. This method and the datasets we have generated will facilitate the characterization of miRNA-mRNA networks in vivo under physiological and pathological conditions.
Collapse
Affiliation(s)
- Xiaoyi Li
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yuri Pritykin
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carla P Concepcion
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuheng Lu
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gaspare La Rocca
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Minsi Zhang
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Bryan King
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Peter J Cook
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Yu Wah Au
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Internal Medicine (Nephrology), Leiden University Medical Center, Zuid-Holland, 2333 ZA, the Netherlands
| | - Olesja Popow
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hannah G Otis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Chiara Mastroleo
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paul Ogrodowski
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ryan Schreiner
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kevin M Haigis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Doron Betel
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Andrea Ventura
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
71
|
miR-155 indicates the fate of CD4 + T cells. Immunol Lett 2020; 224:40-49. [PMID: 32485191 DOI: 10.1016/j.imlet.2020.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/14/2020] [Accepted: 05/24/2020] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) are a class of short noncoding RNAs that regulate the translation of target messenger RNA (mRNA) and consequently participate in a variety of biological processes at the posttranscriptional level. miR-155, encoded within a region known as the B cell integration cluster (BIC), plays multifunctional roles in shaping lymphocytes ranging from biological development to adaptive immunity. It has been revealed that miR-155 plays a key role in fine-tuning the regulation of lymphocyte subsets, including dendritic cells (DCs), macrophages, B cells, and CD8+ and CD4+ T cells. Antigen-specific CD4+ T lymphocytes are critical for host defense against pathogens and prevention of damage resulting from excessive inflammation. Over the past years, various studies have shown that miR-155 plays a critical role in CD4+ T cells function. Therefore, we summarize multiple target genes of miR-155 that regulate aspects of CD4+ T cells immunity, particularly CD4+ T cells differentiation, in this review. In addition, we also focus on the role of miR-155 in the regulation of immunological diseases, suggesting it as a potential disease biomarker and therapeutic target.
Collapse
|
72
|
Arroyo M, Salka K, Chorvinsky E, Xuchen X, Abutaleb K, Perez GF, Weinstock J, Gaviria S, Gutierrez MJ, Nino G. Airway mir-155 responses are associated with TH1 cytokine polarization in young children with viral respiratory infections. PLoS One 2020; 15:e0233352. [PMID: 32442188 PMCID: PMC7244143 DOI: 10.1371/journal.pone.0233352] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/04/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND MicroRNAs (miRs) control gene expression and the development of the immune system and antiviral responses. MiR-155 is an evolutionarily-conserved molecule consistently induced during viral infections in different cell systems. Notably, there is still an unresolved paradox for the role of miR-155 during viral respiratory infections. Despite being essential for host antiviral TH1 immunity, miR-155 may also contribute to respiratory disease by enhancing allergic TH2 responses and NFkB-mediated inflammation. The central goal of this study was to define how airway miR-155 production is related to TH1, TH2, and pro-inflammatory cytokine responses during naturally occurring viral respiratory infections in young children. METHODS Normalized nasal airway levels of miR-155 and nasal protein levels of IFN-γ, TNF-α, IL-1β, IL-13, IL-4 were quantified in young children (≤2 years) hospitalized with viral respiratory infections and uninfected controls. These data were linked to individual characteristics and respiratory disease parameters. RESULTS A total of 151 subjects were included. Increased miR-155 levels were observed in nasal samples from patients with rhinovirus, RSV and all respiratory viruses analyzed. High miR-155 levels were strongly associated with high IFN-γ production, increased airway TH1 cytokine polarization (IFN-γ/IL-4 ratios) and increased pro-inflammatory responses. High airway miR-155 levels were linked to decreased respiratory disease severity in individuals with high airway TH1 antiviral responses. CONCLUSIONS The airway secretion of miR-155 during viral respiratory infections in young children is associated with enhanced antiviral immunity (TH1 polarization). Further studies are needed to define additional physiological roles of miR-155 in the respiratory tract of human infants and young children during health and disease.
Collapse
Affiliation(s)
- Maria Arroyo
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Kyle Salka
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Elizabeth Chorvinsky
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Xilei Xuchen
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Karima Abutaleb
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Geovanny F. Perez
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Jered Weinstock
- Department of Pediatrics, Division of Pediatric Pulmonology, University at Buffalo, The State University of New York, Buffalo, NY, United States of America
| | - Susana Gaviria
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Maria J. Gutierrez
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
| | - Gustavo Nino
- Department of Pediatrics, Division of Pediatric Pulmonary and Sleep Medicine, Children’s National Medical Center, George Washington University, Washington, DC, United States of America
- Division of Pediatric Allergy and Immunology, Johns Hopkins University, Baltimore, MD, United States of America
| |
Collapse
|
73
|
Cordeiro B, Jeon P, Boukhaled GM, Corrado M, Lapohos O, Roy DG, Williams K, Jones RG, Gruenheid S, Sagan SM, Krawczyk CM. MicroRNA-9 Fine-Tunes Dendritic Cell Function by Suppressing Negative Regulators in a Cell-Type-Specific Manner. Cell Rep 2020; 31:107585. [PMID: 32375032 DOI: 10.1016/j.celrep.2020.107585] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 08/08/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells, cells of the innate immune system, are found in a steady state poised to respond to activating stimuli. Once stimulated, they rapidly undergo dynamic changes in gene expression to adopt an activated phenotype capable of stimulating immune responses. We find that the microRNA miR-9 is upregulated in both bone marrow-derived DCs and conventional DC1s but not in conventional DC2s following stimulation. miR-9 expression in BMDCs and conventional DC1s promotes enhanced DC activation and function, including the ability to stimulate T cell activation and control tumor growth. We find that miR-9 regulated the expression of several negative regulators of transcription, including the transcriptional repressor Polycomb group factor 6 (Pcgf6). These findings demonstrate that miR-9 facilitates the transition of DCs from steady state to mature state by regulating the expression of several negative regulators of DC function in a cell-type-specific manner.
Collapse
Affiliation(s)
- Brendan Cordeiro
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Peter Jeon
- Goodman Cancer Research Center, Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Giselle M Boukhaled
- Goodman Cancer Research Center, Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Mario Corrado
- Goodman Cancer Research Center, Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Orsolya Lapohos
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Dominic G Roy
- Goodman Cancer Research Center, Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Kelsey Williams
- Metabolic and Nutritional Programming Group, Van Andel Institute, 333 Bostwick Avenue NE, Grand Rapids, MI 49503, USA
| | - Russell G Jones
- Goodman Cancer Research Center, Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada; Metabolic and Nutritional Programming Group, Van Andel Institute, 333 Bostwick Avenue NE, Grand Rapids, MI 49503, USA
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Selena M Sagan
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Connie M Krawczyk
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Center, Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada; Metabolic and Nutritional Programming Group, Van Andel Institute, 333 Bostwick Avenue NE, Grand Rapids, MI 49503, USA.
| |
Collapse
|
74
|
Ekiz HA, Ramstead AG, Lee SH, Nelson MC, Bauer KM, Wallace JA, Hu R, Round JL, Rutter J, Drummond MJ, Rao DS, O'Connell RM. T Cell-Expressed microRNA-155 Reduces Lifespan in a Mouse Model of Age-Related Chronic Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2064-2075. [PMID: 32161096 PMCID: PMC7325601 DOI: 10.4049/jimmunol.1901484] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/31/2020] [Indexed: 01/13/2023]
Abstract
Aging-related chronic inflammation is a risk factor for many human disorders through incompletely understood mechanisms. Aged mice deficient in microRNA (miRNA/miR)-146a succumb to life-shortening chronic inflammation. In this study, we report that miR-155 in T cells contributes to shortened lifespan of miR-146a-/- mice. Using single-cell RNA sequencing and flow cytometry, we found that miR-155 promotes the activation of effector T cell populations, including T follicular helper cells, and increases germinal center B cells and autoantibodies in mice aged over 15 months. Mechanistically, aerobic glycolysis genes are elevated in T cells during aging, and upon deletion of miR-146a, in a T cell miR-155-dependent manner. Finally, skewing T cell metabolism toward aerobic glycolysis by deleting mitochondrial pyruvate carrier recapitulates age-dependent T cell phenotypes observed in miR-146a-/- mice, revealing the sufficiency of metabolic reprogramming to influence immune cell functions during aging. Altogether, these data indicate that T cell-specific miRNAs play pivotal roles in regulating lifespan through their influences on inflammaging.
Collapse
Affiliation(s)
- H Atakan Ekiz
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Andrew G Ramstead
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Soh-Hyun Lee
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Morgan C Nelson
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Kaylyn M Bauer
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Jared A Wallace
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Ruozhen Hu
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - June L Round
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Jared Rutter
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Micah J Drummond
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84112
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112; and
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Ryan M O'Connell
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112;
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
75
|
Su YC, Huang YF, Wu YW, Chen HF, Wu YH, Hsu CC, Hsu YC, Lee JC. MicroRNA-155 inhibits dengue virus replication by inducing heme oxygenase-1-mediated antiviral interferon responses. FASEB J 2020; 34:7283-7294. [PMID: 32277848 DOI: 10.1096/fj.201902878r] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) have been reported to directly alter the virus life cycle and virus-host interactions, and so are considered promising molecules for controlling virus infection. In the present study, we observed that miR-155 time-dependently downregulated upon dengue virus (DENV) infection. In contrast, exogenous overexpression of miR-155 appeared to limit viral replication in vitro, suggesting that the low levels of miR-155 would be beneficial for DENV replication. In vivo, overexpression of miR-155 protected ICR suckling mice from the life-threatening effects of DENV infection and reduced virus propagation. Further investigation revealed that the anti-DENV activity of miR-155 was due to target Bach1, resulting in the induction of the heme oxygenase-1 (HO-1)-mediated inhibition of DENV NS2B/NS3 protease activity, ultimately leading to induction of antiviral interferon responses, including interferon-induced protein kinase R (PKR), 2'-5'-oligoadenylate synthetase 1 (OAS1), OAS2, and OAS3 expression, against DENV replication. Collectively, our results provide a promising new strategy to manage DENV infection by modulation of miR-155 expression.
Collapse
Affiliation(s)
- Yu-Chieh Su
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan.,School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Fang Huang
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Wen Wu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Feng Chen
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Hsuan Wu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Chun Hsu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Chin Hsu
- Department of Chinese medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Jin-Ching Lee
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine and Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
76
|
Zhao N, Wang G, Long S, Hu M, Gao J, Ran X, Wang J, Su Y, Wang T. MicroRNA-34a deficiency leads to impaired wound closure by augmented inflammation in mice. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:447. [PMID: 32395491 PMCID: PMC7210195 DOI: 10.21037/atm.2020.03.161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Proper inflammation resolution is critical for cutaneous wound healing and disordered inflammation resolution results in chronic nonhealing wounds. However, the cellular and molecular mechanisms for resolution of inflammation during skin wound healing are not well understood. MicroRNA-34a is regarded as one tumor suppressor with complexed immune regulatory effects, yet its role during skin wound repair is still unclear. Methods Circular full thickness excisional wounds were made on the dorsal skin of C57 mice and miR-34a expression pattern was examined by real time RT-PCR and in situ hybridization. The wound healing rates and histologic morphometric analysis were quantified and compared between wounds treated with antagomir-34a and autologous control antagomir-NC wounds, as well as wounds between miR-34a knockout (KO) and wild type (WT) mice. Immunohistochemistry (IHC) for both MPO and F4/80 were performed to examine the infiltrative neutrophils and macrophages in wounds from miR-34a KO and WT mice. Cytokines including IL-1β, IL-6, TNF-α and IL-10, were detected and analyzed by real time RT-PCR during wound healing. IHC for IL-6 and p-STAT3 were quantified, and WB for p-STAT3 and IL-6R were examined in wounds of miR-34a KO and WT mice. Results We found miR-34a was significantly downregulated in the inflammatory phase and back to normal levels in the proliferative phase. Both topical knockdown wounds miR-34a levels by antagomir gel and systematic knockout miR-34a using KO mice resulted in impaired wound healing with delayed re-epithelialization and augmented inflammation. IHC results indicated that there were more residual infiltrative inflammatory cells in the proliferative phase. Moreover, over-activated IL-6/STAT3 signal pathway was identified in the wounds of miR-34a KO mice. Conclusions Our findings reveal that miR-34a deficiency augments skin wound inflammation response and leads to impaired wound healing, which suggest that targeted inhibition of miR-34a for tissue repair/regeneration should be with serious consideration.
Collapse
Affiliation(s)
- Na Zhao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Guojian Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Shuang Long
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Mengjia Hu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Jining Gao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Xinze Ran
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Junping Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Yongping Su
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Tao Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
77
|
Jia Y, Wei Y. Modulators of MicroRNA Function in the Immune System. Int J Mol Sci 2020; 21:E2357. [PMID: 32235299 PMCID: PMC7177468 DOI: 10.3390/ijms21072357] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) play a key role in fine-tuning host immune homeostasis and responses through the negative regulation of mRNA stability and translation. The pathways regulated by miRNAs are well characterized, but the precise mechanisms that control the miRNA-mediated regulation of gene expression during immune cell-development and immune responses to invading pathogens are incompletely understood. Context-specific interactions of miRNAs with other RNA species or proteins may modulate the function of a given miRNA. Dysregulation of miRNA function is associated with various human diseases, such as cardiovascular diseases and cancers. Here, we review the potential modulators of miRNA function in the immune system, including the transcription regulators of miRNA genes, miRNA-processing enzymes, factors affecting miRNA targeting, and intercellular communication.
Collapse
Affiliation(s)
- Yunhui Jia
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuanyuan Wei
- Department of Immunology, Shanghai Key laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
78
|
MiR-574–5p promotes the differentiation of human cardiac fibroblasts via regulating ARID3A. Biochem Biophys Res Commun 2020; 521:427-433. [DOI: 10.1016/j.bbrc.2019.09.107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/25/2019] [Indexed: 12/22/2022]
|
79
|
Labi V, Peng S, Klironomos F, Munschauer M, Kastelic N, Chakraborty T, Schoeler K, Derudder E, Martella M, Mastrobuoni G, Hernandez-Miranda LR, Lahmann I, Kocks C, Birchmeier C, Kempa S, Quintanilla-Martinez de Fend L, Landthaler M, Rajewsky N, Rajewsky K. Context-specific regulation of cell survival by a miRNA-controlled BIM rheostat. Genes Dev 2019; 33:1673-1687. [PMID: 31699777 PMCID: PMC6942046 DOI: 10.1101/gad.330134.119] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022]
Abstract
Knockout of the ubiquitously expressed miRNA-17∼92 cluster in mice produces a lethal developmental lung defect, skeletal abnormalities, and blocked B lymphopoiesis. A shared target of miR-17∼92 miRNAs is the pro-apoptotic protein BIM, central to life-death decisions in mammalian cells. To clarify the contribution of miR-17∼92:Bim interactions to the complex miR-17∼92 knockout phenotype, we used a system of conditional mutagenesis of the nine Bim 3' UTR miR-17∼92 seed matches. Blocking miR-17∼92:Bim interactions early in development phenocopied the lethal lung phenotype of miR-17∼92 ablation and generated a skeletal kinky tail. In the hematopoietic system, instead of causing the predicted B cell developmental block, it produced a selective inability of B cells to resist cellular stress; and prevented B and T cell hyperplasia caused by Bim haploinsufficiency. Thus, the interaction of miR-17∼92 with a single target is essential for life, and BIM regulation by miRNAs serves as a rheostat controlling cell survival in specific physiological contexts.
Collapse
Affiliation(s)
- Verena Labi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Siying Peng
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Filippos Klironomos
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Mathias Munschauer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Nicolai Kastelic
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Tirtha Chakraborty
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Katia Schoeler
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Emmanuel Derudder
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Institute for Biomedical Ageing Research, University of Innsbruck, Innsbruck 6020, Austria
| | - Manuela Martella
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center Tübingen, Eberhard-Karls-University, Tübingen 72076, Germany
| | - Guido Mastrobuoni
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Luis R Hernandez-Miranda
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Ines Lahmann
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Christine Kocks
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Carmen Birchmeier
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | | | - Markus Landthaler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Nikolaus Rajewsky
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Klaus Rajewsky
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
80
|
Polina ER, Oliveira FM, Sbruzzi RC, Crispim D, Canani LH, Santos KG. Gene polymorphism and plasma levels of miR-155 in diabetic retinopathy. Endocr Connect 2019; 8:1591-1599. [PMID: 31751306 PMCID: PMC6933831 DOI: 10.1530/ec-19-0446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023]
Abstract
Circulating microRNA-155 (miR-155) is associated with type 2 diabetes mellitus (T2DM) and the rs767649 polymorphism in the pre-MIR155 gene is associated with miR-155 expression. However, their relationship with diabetic retinopathy (DR) is still unknown. Therefore, the aim of this case-control study was to test the hypothesis that the rs767649 polymorphism in the pre-MIR155 gene is associated with DR in South Brazilians with T2DM. We also evaluated the association of plasma levels of miR-155 with DR and the rs767649 polymorphism in a subgroup of subjects. The rs767649 polymorphism was genotyped in 139 blood donors and 546 T2DM patients (244 had no DR, 161 had non-proliferative DR and 141 had proliferative DR). miR-155 expression was quantified in 20 blood donors and 60 T2DM patients (20 from each group). Among T2DM patients, the carriership of the A allele and the A allele were more frequent in subjects with DR than in those without it (P < 0.05), and the A allele was independently associated with an increased risk of DR (adjusted OR = 2.12, 95% CI = 1.12-4.01). The plasma levels of miR-155 were lower in T2DM patients than in blood donors (P < 0.001). However, the miR-155 levels did not differ according to the presence and severity of DR or according to rs767649 genotypes among T2DM patients. These findings support that the rs767649 polymorphism in the pre-MIR155 gene is associated with DR in T2DM and that the miR-155 plasma levels might be associated with T2DM. Additional studies are needed to further investigate their clinical significance in DR and T2DM.
Collapse
Affiliation(s)
- E R Polina
- Laboratory of Human Molecular Genetics, Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | - F M Oliveira
- Laboratory of Human Molecular Genetics, Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | - R C Sbruzzi
- Laboratory of Human Molecular Genetics, Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
| | - D Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - L H Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Department of Internal Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - K G Santos
- Laboratory of Human Molecular Genetics, Universidade Luterana do Brasil (ULBRA), Canoas, Brazil
- Cardiovascular Research Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- Correspondence should be addressed to K G Santos:
| |
Collapse
|
81
|
Bogusławska J, Popławski P, Alseekh S, Koblowska M, Iwanicka-Nowicka R, Rybicka B, Kędzierska H, Głuchowska K, Hanusek K, Tański Z, Fernie AR, Piekiełko-Witkowska A. MicroRNA-Mediated Metabolic Reprograming in Renal Cancer. Cancers (Basel) 2019; 11:cancers11121825. [PMID: 31756931 PMCID: PMC6966432 DOI: 10.3390/cancers11121825] [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: 10/25/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of renal cell cancer (RCC). We hypothesized that altered metabolism of RCC cells results from dysregulation of microRNAs targeting metabolically relevant genes. Combined large-scale transcriptomic and metabolic analysis of RCC patients tissue samples revealed a group of microRNAs that contribute to metabolic reprogramming in RCC. miRNAs expressions correlated with their predicted target genes and with gas chromatography-mass spectrometry (GC-MS) metabolome profiles of RCC tumors. Assays performed in RCC-derived cell lines showed that miR-146a-5p and miR-155-5p targeted genes of PPP (the pentose phosphate pathway) (G6PD and TKT), the TCA (tricarboxylic acid cycle) cycle (SUCLG2), and arginine metabolism (GATM), respectively. miR-106b-5p and miR-122-5p regulated the NFAT5 osmoregulatory transcription factor. Altered expressions of G6PD, TKT, SUCLG2, GATM, miR-106b-5p, miR-155-5p, and miR-342-3p correlated with poor survival of RCC patients. miR-106b-5p, miR-146a-5p, and miR-342-3p stimulated proliferation of RCC cells. The analysis involving >6000 patients revealed that miR-34a-5p, miR-106b-5p, miR-146a-5p, and miR-155-5p are PanCancer metabomiRs possibly involved in global regulation of cancer metabolism. In conclusion, we found that microRNAs upregulated in renal cancer contribute to disturbed expression of key genes involved in the regulation of RCC metabolome. miR-146a-5p and miR-155-5p emerge as a key “metabomiRs” that target genes of crucial metabolic pathways (PPP (the pentose phosphate pathway), TCA cycle, and arginine metabolism).
Collapse
Affiliation(s)
- Joanna Bogusławska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
| | - Piotr Popławski
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
| | - Saleh Alseekh
- Max-Planck Institute of Molecular Plant Physiology, 14476 Potsdam-Golm, Germany; (S.A.); (A.R.F.)
- Center for Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
| | - Marta Koblowska
- Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland; (M.K.); (R.I.-N.)
- Laboratory for Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland; (M.K.); (R.I.-N.)
- Laboratory for Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Beata Rybicka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
| | - Hanna Kędzierska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
| | - Katarzyna Głuchowska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
| | - Karolina Hanusek
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
| | - Zbigniew Tański
- Masovian Specialist Hospital in Ostroleka, 07-410 Ostroleka, Poland;
| | - Alisdair R. Fernie
- Max-Planck Institute of Molecular Plant Physiology, 14476 Potsdam-Golm, Germany; (S.A.); (A.R.F.)
- Center for Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
| | - Agnieszka Piekiełko-Witkowska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (P.P.); (B.R.); (H.K.); (K.G.); (K.H.)
- Correspondence: ; Tel.: +48-22-5693810
| |
Collapse
|
82
|
Hua Y, Zhang J, Jia Z, Li J, Xiong X, Xiong Y. Immune-related genes response to stimulation of miR-155 overexpression in CIK (ctenopharyngodon idella kidney) cells and zebrafish. FISH & SHELLFISH IMMUNOLOGY 2019; 94:142-148. [PMID: 31487536 DOI: 10.1016/j.fsi.2019.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/28/2019] [Accepted: 09/01/2019] [Indexed: 06/10/2023]
Abstract
MiR-155 regulates the development of germinal-center and the generation of immunoglobulin class-switched plasma cells. However, whether miR-155 is involved in immune response in fish is still unclear. Here, CIK cells transfected with miR-155 overexpressed plasmid inhibited mRNA expression of mIg and Rag2 (P < 0.05). Interestingly, mIg was predicted as a potential target gene of miR-155 by RNAhybrid, with a putative binding site in its CDS. Further, mIg luciferase reporter vectors with successive deletions of mIg cDNA sequence were constructed and dual luciferase reporter assay showed that vectors containing the sequence from 318 to 347 in CDS exhibited lower relative luciferase activity than others without predicted binding region (P < 0.05), which indicated mIg is the target gene of miR-155 and reveal bona fide targeted binding site of mIg for miR-155 in fish. In vivo, the zebrafish were respectively injected with miR-155 overexpressed and empty vector, and showed that miR-155 efficiently expressed in zebrafish (P < 0.01), which consistently decreased mRNA level of immune-related genes, including mIg (P < 0.01), sIg (P < 0.05), AID (P < 0.01), PU.1 (P < 0.05) and Rag2 (P < 0.05) at d 3 and d 6 post injection, comparing to control. Collectively, this work indicates that overexpression of miR-155 suppresses the mRNA level of immune-related genes in CIK cells and zebrafish, and mIg is a novel target gene of miR-155 in fish. These findings provide an insight into the miR-155 modulating adaptive immunity in grass carp and zebrafish.
Collapse
Affiliation(s)
- Yonglin Hua
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, 610041, China; College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China
| | - Jing Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, 610041, China; College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China
| | - Zhihao Jia
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, 610041, China; College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu, China.
| | - Xianrong Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, 610041, China; College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, 610041, China; College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China; Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu, China.
| |
Collapse
|
83
|
Winter SJ, Krueger A. Development of Unconventional T Cells Controlled by MicroRNA. Front Immunol 2019; 10:2520. [PMID: 31708931 PMCID: PMC6820353 DOI: 10.3389/fimmu.2019.02520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/10/2019] [Indexed: 12/21/2022] Open
Abstract
Post-transcriptional gene regulation through microRNA (miRNA) has emerged as a major control mechanism of multiple biological processes, including development and function of T cells. T cells are vital components of the immune system, with conventional T cells playing a central role in adaptive immunity and unconventional T cells having additional functions reminiscent of both innate and adaptive immunity, such as involvement in stress responses and tissue homeostasis. Unconventional T cells encompass cells expressing semi-invariant T cell receptors (TCRs), such as invariant Natural Killer T (iNKT) and Mucosal-Associated Invariant T (MAIT) cells. Additionally, some T cells with diverse TCR repertoires, including γδT cells, intraepithelial lymphocytes (IEL) and regulatory T (Treg) cells, share some functional and/or developmental features with their semi-invariant unconventional counterparts. Unconventional T cells are particularly sensitive to disruption of miRNA function, both globally and on the individual miRNA level. Here, we review the role of miRNA in the development and function of unconventional T cells from an iNKT-centric point of view. The function of single miRNAs can provide important insights into shared and individual pathways for the formation of different unconventional T cell subsets.
Collapse
Affiliation(s)
- Samantha J Winter
- Institute for Molecular Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Andreas Krueger
- Institute for Molecular Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
84
|
Du L, Lin L, Li Q, Liu K, Huang Y, Wang X, Cao K, Chen X, Cao W, Li F, Shao C, Wang Y, Shi Y. IGF-2 Preprograms Maturing Macrophages to Acquire Oxidative Phosphorylation-Dependent Anti-inflammatory Properties. Cell Metab 2019; 29:1363-1375.e8. [PMID: 30745181 DOI: 10.1016/j.cmet.2019.01.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/14/2018] [Accepted: 01/14/2019] [Indexed: 12/24/2022]
Abstract
Recent investigations revealed that macrophages could be trained with an altered responsiveness, raising the possibility of combating autoimmune diseases by imparting anti-inflammatory capabilities to these cells. While investigating the effect of mesenchymal stem cells on experimental autoimmune encephalomyelitis (EAE), we found a critical role of insulin-like growth factor 2 (IGF-2) in training macrophages to become anti-inflammatory during their maturation. IGF-2 exerts its effects by preprogramming maturing macrophages to commit oxidative phosphorylation (OXPHOS). IGF-2-preprogrammed macrophages maintained the mitochondrial complex V activities even upon pro-inflammation stimulation, thus enabling an elevated programmed death-ligand 1 (PD-L1) expression. PD-L1 neutralization abolished the beneficial effect of IGF-2 on EAE. Furthermore, adoptive transfer of IGF-2-preprogrammed macrophages to EAE mice increased Tregs and alleviated the diseases. Our results demonstrate that shaping macrophage responsiveness by IGF-2 is effective in managing inflammatory diseases, and the OXPHOS commitment can be preset to determine the anti-inflammatory fate of macrophages.
Collapse
Affiliation(s)
- Liming Du
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China; The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Liangyu Lin
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China; The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Qing Li
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Keli Liu
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Yin Huang
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Xuefeng Wang
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Kai Cao
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Xiaodong Chen
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Wei Cao
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Fengying Li
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China
| | - Ying Wang
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China.
| | - Yufang Shi
- Shanghai Jiao Tong University School of Medicine and Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China; The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
85
|
Martinez-Usatorre A, Sempere LF, Carmona SJ, Carretero-Iglesia L, Monnot G, Speiser DE, Rufer N, Donda A, Zehn D, Jandus C, Romero P. MicroRNA-155 Expression Is Enhanced by T-cell Receptor Stimulation Strength and Correlates with Improved Tumor Control in Melanoma. Cancer Immunol Res 2019; 7:1013-1024. [PMID: 31043416 DOI: 10.1158/2326-6066.cir-18-0504] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/24/2018] [Accepted: 04/26/2019] [Indexed: 11/16/2022]
Abstract
microRNAs are short noncoding RNAs that regulate protein expression posttranscriptionally. We previously showed that miR-155 promotes effector CD8+ T-cell responses. However, little is known about the regulation of miR-155 expression. Here, we report that antigen affinity and dose determine miR-155 expression in CD8+ T cells. In B16 tumors expressing a low-affinity antigen ligand, tumor-specific infiltrating CD8+ T cells showed variable miR-155 expression, whereby high miR-155 expression was associated with more cytokine-producing cells and tumor control. Moreover, anti-PD-1 treatment led to both increased miR-155 expression and tumor control by specific CD8+ T cells. In addition, miR-155 overexpression enhanced exhausted CD8+ T-cell persistence in the LCMV cl13 chronic viral infection model. In agreement with these observations in mouse models, miR-155 expression in human effector memory CD8+ T cells positively correlated with their frequencies in tumor-infiltrated lymph nodes of melanoma patients. Low miR-155 target gene signature in tumors was associated with prolonged overall survival in melanoma patients. Altogether, these results raise the possibility that high miR-155 expression in CD8+ tumor-infiltrating T cells may be a surrogate marker of the relative potency of in situ antigen-specific CD8+ T-cell responses.
Collapse
Affiliation(s)
| | - Lorenzo F Sempere
- Department of Radiology, Precision Health Program, Michigan State University, East Lansing, Michigan
| | - Santiago J Carmona
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Laura Carretero-Iglesia
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Gwennaëlle Monnot
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Daniel E Speiser
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.,Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Epalinges, Switzerland
| | - Alena Donda
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Dietmar Zehn
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Camilla Jandus
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Pedro Romero
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
86
|
Affiliation(s)
- Pengda Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kunyu Liao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
87
|
Labi V, Schoeler K, Melamed D. miR-17∼92 in lymphocyte development and lymphomagenesis. Cancer Lett 2019; 446:73-80. [PMID: 30660648 DOI: 10.1016/j.canlet.2018.12.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/06/2018] [Accepted: 12/31/2018] [Indexed: 01/07/2023]
Abstract
microRNAs (miRNAs) down-modulate the levels of proteins by sequence-specific binding to their respective target mRNAs, causing translational repression or mRNA degradation. The miR-17∼92 cluster encodes for six miRNAs whose target recognition specificities are determined by their distinct sequence. In mice, the four miRNA families generated from the miR-17∼92 cluster coordinate to allow for proper lymphocyte development and effective adaptive immune responses following infection or immunization. Lymphocyte development and homeostasis rely on tight regulation of PI3K signaling to avoid autoimmunity or immunodeficiency, and the miR-17∼92 miRNAs appear as key mediators to appropriately tune PI3K activity. On the other hand, in lymphoid tumors overexpression of the miR-17∼92 miRNAs is a common oncogenic event. In this review, we touch on what we have learned so far about the miR-17∼92 miRNAs, particularly with respect to their role in lymphocyte development, homeostasis and pathology.
Collapse
Affiliation(s)
- Verena Labi
- Division of Developmental Immunology, Biocenter, Innsbruck Medical University, Innsbruck, 6020, Austria.
| | - Katia Schoeler
- Division of Developmental Immunology, Biocenter, Innsbruck Medical University, Innsbruck, 6020, Austria
| | - Doron Melamed
- Department of Immunology, Technion-Israel Institute of Technology, Haifa, 31096, Israel.
| |
Collapse
|
88
|
Ekiz HA, Huffaker TB, Grossmann AH, Stephens WZ, Williams MA, Round JL, O'Connell RM. MicroRNA-155 coordinates the immunological landscape within murine melanoma and correlates with immunity in human cancers. JCI Insight 2019; 4:126543. [PMID: 30721153 DOI: 10.1172/jci.insight.126543] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/31/2019] [Indexed: 12/27/2022] Open
Abstract
miR-155 has recently emerged as an important promoter of antitumor immunity through its functions in T lymphocytes. However, the impact of T cell-expressed miR-155 on immune cell dynamics in solid tumors remains unclear. In the present study, we used single-cell RNA sequencing to define the CD45+ immune cell populations at different time points within B16F10 murine melanoma tumors growing in either wild-type or miR-155 T cell conditional knockout (TCKO) mice. miR-155 was required for optimal T cell activation and reinforced the T cell response at the expense of infiltrating myeloid cells. Further, myeloid cells from tumors growing in TCKO mice were defined by an increase in wound healing genes and a decreased IFN-γ-response gene signature. Finally, we found that miR-155 expression predicted a favorable outcome in human melanoma patients and was associated with a strong immune signature. Moreover, gene expression analysis of The Cancer Genome Atlas (TCGA) data revealed that miR-155 expression also correlates with an immune-enriched subtype in 29 other human solid tumors. Together, our study provides an unprecedented analysis of the cell types and gene expression signatures of immune cells within experimental melanoma tumors and elucidates the role of miR-155 in coordinating antitumor immune responses in mammalian tumors.
Collapse
Affiliation(s)
| | | | - Allie H Grossmann
- Division of Anatomic Pathology, Department of Pathology, University of Utah.,Huntsman Cancer Institute, University of Utah Health Sciences Center, and.,ARUP Laboratories, University of Utah, Salt Lake City, Utah, USA
| | | | | | | | | |
Collapse
|
89
|
Abstract
MicroRNAs (miRNAs) are naturally occurring, highly conserved families of transcripts (∼22 nucleotides in length) that are processed from larger hairpin precursors. miRNAs primarily regulate gene expression by promoting messenger RNA (mRNA) degradation or repressing mRNA translation. miRNAs have been shown to be important regulators of a variety of cellular processes involving development, differentiation, and signaling. Moreover, various human diseases, including cancer and immune dysfunction, are associated with aberrant expression of miRNAs. This review will focus on how the multifunctional miRNA, miR-155, regulates inflammatory diseases, including cancer and pulmonary disorders, and also how miR-155 expression and biogenesis are regulated. We will also provide examples of miR-155-regulated networks in coordination with other noncoding RNAs, including long noncoding RNAs as well as coding mRNAs acting as competing endogenous RNAs.
Collapse
Affiliation(s)
- Guruswamy Mahesh
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
90
|
Maul J, Alterauge D, Baumjohann D. Micro
RNA
‐mediated regulation of T follicular helper and T follicular regulatory cell identity. Immunol Rev 2019; 288:97-111. [DOI: 10.1111/imr.12735] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/21/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Julia Maul
- Institute for ImmunologyBiomedical CenterLudwig‐Maximilians‐Universität München Planegg‐Martinsried Germany
| | - Dominik Alterauge
- Institute for ImmunologyBiomedical CenterLudwig‐Maximilians‐Universität München Planegg‐Martinsried Germany
| | - Dirk Baumjohann
- Institute for ImmunologyBiomedical CenterLudwig‐Maximilians‐Universität München Planegg‐Martinsried Germany
| |
Collapse
|
91
|
Bondada MS, Yao Y, Nair V. Multifunctional miR-155 Pathway in Avian Oncogenic Virus-Induced Neoplastic Diseases. Noncoding RNA 2019; 5:ncrna5010024. [PMID: 30871221 PMCID: PMC6468363 DOI: 10.3390/ncrna5010024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/02/2019] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that fine-tune the responses of the cell by modulating the cell transcriptome and gene expression. MicroRNA 155 (miR-155) is a conserved multifunctional miRNA involved in multiple roles including the modulation of the immune responses. When deregulated, miR-155 can also contribute to cancer as has been demonstrated in several human malignancies such as diffuse large B cell lymphoma, chronic lymphocytic leukemia, as well as in Epstein⁻Barr virus (EBV)-induced B cell transformation. Avian oncogenic viruses such as Marek's disease virus (MDV), avian leukosis virus (ALV), and reticuloendotheliosis virus (REV) that account for more than 90% of cancers in avian species, also make use of the miR-155 pathway during oncogenesis. While oncogenic retroviruses, such as ALV, activate miR-155 by insertional activation, acutely transforming retroviruses use transduced oncogenes such as v-rel to upregulate miR-155 expression. MDV on the other hand, encodes a functional miR-155 ortholog mdv1-miR-M4, similar to the miR-155 ortholog kshv-miR-K11 present in Kaposi's sarcoma-associated herpesvirus (KSHV). We have shown that mdv1-miR-M4 is critical for the induction of MDV-induced lymphomas further demonstrating the oncogenic potential of miR-155 pathway in cancers irrespective of the diverse etiology. In this review, we discuss on our current understanding of miR-155 function in virus-induced lymphomas focusing primarily on avian oncogenic viruses.
Collapse
Affiliation(s)
- Megha Sravani Bondada
- Avian Oncogenic Viruses, The Pirbright Institute and the UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| | - Yongxiu Yao
- Avian Oncogenic Viruses, The Pirbright Institute and the UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
| | - Venugopal Nair
- Avian Oncogenic Viruses, The Pirbright Institute and the UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK.
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, United Kingdom..
| |
Collapse
|
92
|
Meldolesi J. Extracellular vesicles, news about their role in immune cells: physiology, pathology and diseases. Clin Exp Immunol 2019; 196:318-327. [PMID: 30756386 DOI: 10.1111/cei.13274] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
Two types of extracellular vesicles (EVs), exosomes and ectosomes, are generated and released by all cells, including immune cells. The two EVs appear different in many properties: size, mechanism and site of assembly, composition of their membranes and luminal cargoes, sites and processes of release. In functional terms, however, these differences are minor. Moreover, their binding to and effects on target cells appear similar, thus the two types are considered distinct only in a few cases, otherwise they are presented together as EVs. The EV physiology of the various immune cells differs as expected from their differential properties. Some properties, however, are common: EV release, taking place already at rest, is greatly increased upon cell stimulation; extracellular navigation occurs adjacent and at distance from the releasing cells; binding to and uptake by target cells are specific. EVs received from other immune or distinct cells govern many functions in target cells. Immune diseases in which EVs play multiple, often opposite (aggression and protection) effects, are numerous; inflammatory diseases; pathologies of various tissues; and brain diseases, such as multiple sclerosis. EVs also have effects on interactive immune and cancer cells. These effects are often distinct, promoting cytotoxicity or proliferation, the latter together with metastasis and angiogenesis. Diagnoses depend on the identification of EV biomarkers; therapies on various mechanisms such as (1) removal of aggression-inducing EVs; (2) EV manipulations specific for single targets, with insertion of surface peptides or luminal miRNAs; and (3) removal or re-expression of molecules from target cells.
Collapse
Affiliation(s)
- J Meldolesi
- Division of Neuroscience, Unit of Molecular and Cellular Neuroscience, San Raffaele Scientific Institute and San Raffaele University, Milan, Italy
| |
Collapse
|
93
|
Epigenetics of autoimmune liver diseases: current progress and future directions. JOURNAL OF BIO-X RESEARCH 2019. [DOI: 10.1097/jbr.0000000000000030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
94
|
Dual regulation of Arabidopsis AGO2 by arginine methylation. Nat Commun 2019; 10:844. [PMID: 30783097 PMCID: PMC6381116 DOI: 10.1038/s41467-019-08787-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/29/2019] [Indexed: 12/21/2022] Open
Abstract
Argonaute (AGO) proteins are core components of RNA interference (RNAi) but the mechanisms of their regulation, especially at the post-translational level, remain unclear. Among the ten AGOs in Arabidopsis, only AGO2 is induced by bacterial infection and is known to positively regulate immunity. Here we show that the N-terminal domain of AGO2 is enriched with arginine-glycine RG/GR repeats, which are methylated by protein arginine methyltransferase5 (PRMT5). Arginine methylation has dual functions in AGO2 regulation. Methylated arginine residues can promote AGO2 protein degradation and are also bound by Tudor-domain proteins (TSNs), which can degrade AGO2-associated small RNAs (sRNAs). PRMT5 is down-regulated during infection and the prmt5 mutant is more resistant to bacteria. We speculate that reduced PRMT5 expression during infection may lead to reduced arginine methylation of AGO2, resulting in accumulation of both AGO2 and, via reduced interaction with TSNs, accumulation of AGO2-associated sRNAs, to promote plant immunity. These results reveal that both the arginine methylation writer (PRMT5) and readers (TSNs) can regulate AGO2-mediated RNAi.
Collapse
|
95
|
Fu H, Cheng Y, Luo H, Rong Z, Li Y, Lu P, Ye X, Huang W, Qi Z, Li X, Cheng B, Wang X, Yao Y, Zhang YW, Zheng W, Zheng H. Silencing MicroRNA-155 Attenuates Kainic Acid-Induced Seizure by Inhibiting Microglia Activation. Neuroimmunomodulation 2019; 26:67-76. [PMID: 30928987 DOI: 10.1159/000496344] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/16/2018] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE(S) Neuroinflammation is an important contributor to the development of seizures and epilepsy. Micro-RNA-155 (miR-155) plays a critical role in immunity and -inflammation. This study aims to explore the function of miR-155 and miR-155-mediated inflammation in epilepsy. METHODS About 8-week-old male C57BL/6 mice were administered an intraperitoneal injection (i.p.) of kainic acid (KA) (15 mg/kg) or saline. The mice in the KA group developing acute seizure were further subjected to intracerebroventricular injection (i.c.v.) of antagomir negative control (NC) or miR-155 antagomir. Animal behavior was observed according to Racine's scale, and electroencephalographs were recorded. Primary microglia were cultured and treated with antagomir NC or antagomir. Whole-cell electrophysiological recording was conducted to detect the spontaneous EPSCs and IPSCs in the neurons treated with different conditioned medium from those microglia. miR-155 were detected by qRT-PCR in those models, as well as in the brain or blood from epileptic patients and healthy controls. RESULTS miR-155 was abundantly expressed in glial cells compared with neurons, and its expression was markedly elevated in the brain of epilepsy patients and KA-induced seizure mice. Silencing miR-155 attenuated KA-induced seizure, abnormal electroencephalography, proinflammatory cytokine expression, and microglia morphology change. Moreover, conditioned media from KA-treated microglia impaired neuron excitability, whereas conditioned media from KA and miR-155 antagomir co-treated microglia had no such effects. Finally, miR-155 levels were significantly higher in the blood of epilepsy patients than those of healthy controls. CONCLUSION(S) These findings demonstrate that aberrant upregulation of miR-155 contributes to epileptogenesis through inducing microglia neuroinflammation.
Collapse
Affiliation(s)
- Huajun Fu
- Department of Neurology, Affiliated Zhongshan Hospital, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Department of Neurology, The Affiliated Longyan First Hospital of Fujian Medical University, Longyan, China
| | - Yiyun Cheng
- Department of Neurology, Affiliated Zhongshan Hospital, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Graduate School of Fujian Medical University, Xiamen, China
| | - Haijuan Luo
- Department of Neurology, Affiliated Zhongshan Hospital, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Zhouyi Rong
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Ping Lu
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaowen Ye
- Department of Neurology, Affiliated Zhongshan Hospital, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Weiyan Huang
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Clinical Medicine of Grade 2014, School of Medicine, Xiamen University, Xiamen, China
| | - Ziguo Qi
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Clinical Medicine of Grade 2014, School of Medicine, Xiamen University, Xiamen, China
| | - Xiuying Li
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Clinical Medicine of Grade 2014, School of Medicine, Xiamen University, Xiamen, China
| | - Baoying Cheng
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Xintian Wang
- Department of Neurology, Affiliated Zhongshan Hospital, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Graduate School of Fujian Medical University, Xiamen, China
| | - Yi Yao
- Epilepsy Research Center, Department of Neurosurgery, Hongai Hospital, Xiamen, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Weihong Zheng
- Department of Neurology, Affiliated Zhongshan Hospital, Xiamen University, Xiamen, China
- Graduate School of Fujian Medical University, Xiamen, China
| | - Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Diseases and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China,
- Shenzhen Research Institute, Xiamen University, Shenzhen, China,
| |
Collapse
|
96
|
MicroRNAs as Regulators of Insulin Signaling: Research Updates and Potential Therapeutic Perspectives in Type 2 Diabetes. Int J Mol Sci 2018; 19:ijms19123705. [PMID: 30469501 PMCID: PMC6321520 DOI: 10.3390/ijms19123705] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/08/2018] [Accepted: 11/17/2018] [Indexed: 12/21/2022] Open
Abstract
The insulin signaling pathway is composed of a large number of molecules that positively or negatively modulate insulin specific signal transduction following its binding to the cognate receptor. Given the importance of the final effects of insulin signal transduction, it is conceivable that many regulators are needed in order to tightly control the metabolic or proliferative functional outputs. MicroRNAs (miRNAs) are small non-coding RNA molecules that negatively modulate gene expression through their specific binding within the 3′UTR sequence of messenger RNA (mRNA), thus causing mRNA decoy or translational inhibition. In the last decade, miRNAs have been addressed as pivotal cellular rheostats which control many fundamental signaling pathways, including insulin signal transduction. Several studies demonstrated that multiple alterations of miRNAs expression or function are relevant for the development of insulin resistance in type 2 diabetes (T2D); such alterations have been highlighted in multiple insulin target organs including liver, muscles, and adipose tissue. Indirectly, miRNAs have been identified as modulators of inflammation-derived insulin resistance, by controlling/tuning the activity of innate immune cells in insulin target tissues. Here, we review main findings on miRNA functions as modulators of insulin signaling in physiologic- or in T2D insulin resistance- status. Additionally, we report the latest hypotheses of prospective therapies involving miRNAs as potential targets for future drugs in T2D.
Collapse
|