1
|
Roy U, Desai SS, Kumari S, Bushra T, Choudhary B, Raghavan SC. Understanding the Role of miR-29a in the Regulation of RAG1, a Gene Associated with the Development of the Immune System. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1125-1138. [PMID: 39269689 DOI: 10.4049/jimmunol.2300344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/16/2024] [Indexed: 09/15/2024]
Abstract
The process of Ag receptor diversity is initiated by RAGs consisting of RAG1 and RAG2 in developing lymphocytes. Besides its role as a sequence-specific nuclease during V(D)J recombination, RAGs can also act as a structure-specific nuclease leading to genome instability. Thus, regulation of RAG expression is essential to maintaining genome stability. Previously, the role of miR29c in the regulation of RAG1 was identified. In this article, we report the regulation of RAG1 by miR-29a in the lymphocytes of both mice (Mus musculus) and humans (Homo sapiens). The level of RAG1 could be modulated by overexpression of miR-29a and inhibition using anti-miRs. Argonaute2-immunoprecipitation and high-throughput sequencing of RNA isolated by crosslinking immunoprecipitation studies established the association of miR-29a and RAG1 with Argonaute proteins. We observed a negative correlation between miR-29a and RAG1 levels in mouse B and T cells and leukemia patients. Overexpression of pre-miR-29a in the bone marrow cells of mice led to the generation of mature miR-29a transcripts and reduced RAG1 expression, which led to a significant reduction in V(D)J recombination in pro-B cells. Importantly, our studies are consistent with the phenotype reported in miR-29a knockout mice, which showed impaired immunity and survival defects. Finally, we show that although both miR-29c and miR-29a can regulate RAG1 at mRNA and protein levels, miR-29a substantially impacts immunity and survival. Our results reveal that the repression of RAG1 activity by miR-29a in B cells of mice and humans is essential to maintain Ig diversity and prevent hematological malignancies resulting from aberrant RAG1 expression in lymphocytes.
Collapse
Affiliation(s)
- Urbi Roy
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Sagar Sanjiv Desai
- Institute of Bioinformatics and Applied Biotechnology, Electronics City, Bangalore, India
| | - Susmita Kumari
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Tanzeem Bushra
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronics City, Bangalore, India
| | - Sathees C Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| |
Collapse
|
2
|
Tran V, de Oliveira‐Jr GP, Chidester S, Lu S, Pleet ML, Ivanov AR, Tigges J, Yang M, Jacobson S, Gonçalves MCB, Schmaier AA, Jones J, Ghiran IC. Choice of blood collection methods influences extracellular vesicles counts and miRNA profiling. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70008. [PMID: 39440167 PMCID: PMC11494683 DOI: 10.1002/jex2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/15/2024] [Accepted: 08/26/2024] [Indexed: 10/25/2024]
Abstract
Circulating RNAs have been investigated systematically for over 20 years, both as constituents of circulating extracellular vesicles (EVs) or, more recently, non-EV RNA carriers, such as exomeres and supermeres. The high level of variability and low reproducibility rate of EV/extracellular RNA (exRNA) results generated even on the same biofluids promoted several efforts to limit pre-analytical variability by standardizing sample collection and sample preparation, along with instrument validation, setup and calibration. Anticoagulants (ACs) are often chosen based on the initial goal of the study and not necessarily for the later EV and/or exRNA analyses. We show the effects of blood collection on EV size, abundance, and antigenic composition, as well on the miRNAs. Our focus of this work was on the effect of ACs on the number and antigenic composition of circulating EVs and on a set of circulating miRNA species, which were shown to be relevant as disease markers in several cancers and Alzheimer's disease. Results show that while the number of plasma EVs, their relative size, and post-fluorescence labeling profile varied with each AC, their overall antigenic composition, with few exceptions, did not change significantly. However, the number of EVs expressing platelet and platelet-activation markers increased in serum samples. For overall miRNA expression levels, EDTA was a better AC, although this may have been associated with stimulation of cells in the blood collection tube. Citrate and serum rendered better results for a set of miRNAs that were described as circulating markers for Alzheimer's disease, colon, and papillary thyroid cancers.
Collapse
Affiliation(s)
- Vivian Tran
- Department of Anesthesia, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Getulio Pereira de Oliveira‐Jr
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
- Department of Chemistry and Chemical Biology, The Barnett Institute of Chemical & Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - Stephanie Chidester
- Laboratory of Pathology, Center for Cancer ResearchNational Cancer InstituteBethesdaMassachusettsUSA
| | - Shulin Lu
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Michelle L. Pleet
- Neuroimmunology and Neurovirology Division, National Institute for Neurological Disease and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Alexander R. Ivanov
- Department of Chemistry and Chemical Biology, The Barnett Institute of Chemical & Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - John Tigges
- Nanoflow Cytometry Core Facility, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Moua Yang
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Steven Jacobson
- Neuroimmunology and Neurovirology Division, National Institute for Neurological Disease and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Maria C. B. Gonçalves
- Department of Anesthesia, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Alec A. Schmaier
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Jennifer Jones
- Laboratory of Pathology, Center for Cancer ResearchNational Cancer InstituteBethesdaMassachusettsUSA
| | - Ionita C. Ghiran
- Department of Anesthesia, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
3
|
Iacomino N, Tarasco MC, Berni A, Ronchi J, Mantegazza R, Cavalcante P, Foti M. Non-Coding RNAs in Myasthenia Gravis: From Immune Regulation to Personalized Medicine. Cells 2024; 13:1550. [PMID: 39329732 PMCID: PMC11430632 DOI: 10.3390/cells13181550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Myasthenia gravis (MG) is an antibody-mediated autoimmune disorder characterized by altered neuromuscular transmission, which causes weakness and fatigability in the skeletal muscles. The etiology of MG is complex, being associated with multiple genetic and environmental factors. Over recent years, progress has been made in understanding the immunological alterations implicated in the disease, but the exact pathogenesis still needs to be elucidated. A pathogenic interplay between innate immunity and autoimmunity contributes to the intra-thymic MG development. Epigenetic changes are critically involved in both innate and adaptive immune response regulation. They can act as (i) pathological factors besides genetic predisposition and (ii) co-factors contributing to disease phenotypes or patient-specific disease course/outcomes. This article reviews the role of non-coding RNAs (ncRNAs) as epigenetic factors implicated in MG. Particular attention is dedicated to microRNAs (miRNAs), whose expression is altered in MG patients' thymuses and circulating blood. The long ncRNA (lncRNA) contribution to MG, although not fully characterized yet, is also discussed. By summarizing the most recent and fast-growing findings on ncRNAs in MG, we highlight the therapeutic potential of these molecules for achieving immune regulation and their value as biomarkers for the development of personalized medicine approaches to improve disease care.
Collapse
Affiliation(s)
- Nicola Iacomino
- Neurology 4-Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Maria Cristina Tarasco
- Neurology 4-Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
- Ph.D. Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy
| | - Alessia Berni
- Neurology 4-Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Jacopo Ronchi
- Ph.D. Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- BicOMICs, University of Milano-Bicocca, 20900 Monza, Italy
| | - Renato Mantegazza
- Neurology 4-Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Paola Cavalcante
- Neurology 4-Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Maria Foti
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- BicOMICs, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
4
|
Ramadan YN, Kamel AM, Medhat MA, Hetta HF. MicroRNA signatures in the pathogenesis and therapy of inflammatory bowel disease. Clin Exp Med 2024; 24:217. [PMID: 39259390 PMCID: PMC11390904 DOI: 10.1007/s10238-024-01476-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
Inflammatory bowel disease (IBD) is a persistent inflammatory illness of the gastrointestinal tract (GIT) triggered by an inappropriate immune response to environmental stimuli in genetically predisposed persons. Unfortunately, IBD patients' quality of life is negatively impacted by the symptoms associated with the disease. The exact etiology of IBD pathogenesis is not fully understood, but the emerging research indicated that the microRNA (miRNA) plays an important role. miRNAs have been documented to possess a significant role in regulating pro- and anti-inflammatory pathways, in addition to their roles in several physiological processes, including cell growth, proliferation, and apoptosis. Variations in the miRNA profiles might be a helpful prognostic indicator and a valuable tool in the differential diagnosis of IBD. Most interestingly, these miRNAs have a promising therapeutic target in several pre-clinical animal studies and phase 2 clinical studies to alleviate inflammation and improve patient's quality of life. This comprehensive review discusses the current knowledge about the significant physiological role of different miRNAs in the health of the intestinal immune system and addresses the role of the most relevant differentially expressed miRNAs in IBD, identify their potential targets, and emphasize their diagnostic and therapeutic potential for future research.
Collapse
Affiliation(s)
- Yasmin N Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt.
| | - Ayat M Kamel
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt
| | - Mohammed A Medhat
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Helal F Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| |
Collapse
|
5
|
Farhan SH, Jasim SA, Bansal P, Kaur H, Abed Jawad M, Qasim MT, Jabbar AM, Deorari M, Alawadi A, Hadi A. Exosomal Non-coding RNA Derived from Mesenchymal Stem Cells (MSCs) in Autoimmune Diseases Progression and Therapy; an Updated Review. Cell Biochem Biophys 2024:10.1007/s12013-024-01432-4. [PMID: 39225902 DOI: 10.1007/s12013-024-01432-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 09/04/2024]
Abstract
Inflammation and autoimmune diseases (AD) are common outcomes of an overactive immune system. Inflammation occurs due to the immune system reacting to damaging stimuli. Exosomes are being recognized as an advanced therapeutic approach for addressing an overactive immune system, positioning them as a promising option for treating AD. Mesenchymal stem cells (MSCs) release exosomes that have strong immunomodulatory effects, influenced by their cell of origin. MSCs-exosomes, being a cell-free therapy, exhibit less toxicity and provoke a diminished immune response compared to cell-based therapies. Exosomal non-coding RNAs (ncRNA), particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are intricately linked to various biological and functional aspects of human health. Exosomal ncRNAs can lead to tissue malfunction, aging, and illnesses when they experience tissue-specific alterations as a result of various internal or external problems. In this study, we will examine current trends in exosomal ncRNA researches regarding AD. Then, therapeutic uses of MSCs-exosomal ncRNA will be outlined, with a particle focus on the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Shireen Hamid Farhan
- Biotechnology department, College of Applied Science, Fallujah University, Fallujah, Iraq
| | | | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, India
| | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq.
| | - Maytham T Qasim
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Abeer Mhussan Jabbar
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq.
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq
- College of technical engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of technical engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Hadi
- Department of medical laboratories techniques, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| |
Collapse
|
6
|
Daley AD, Bénézech C. Fat-associated lymphoid clusters: Supporting visceral adipose tissue B cell function in immunity and metabolism. Immunol Rev 2024; 324:78-94. [PMID: 38717136 DOI: 10.1111/imr.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/09/2024] [Indexed: 07/23/2024]
Abstract
It is now widely understood that visceral adipose tissue (VAT) is a highly active and dynamic organ, with many functions beyond lipid accumulation and storage. In this review, we discuss the immunological role of this tissue, underpinned by the presence of fat-associated lymphoid clusters (FALCs). FALC's distinctive structure and stromal cell composition support a very different immune cell mix to that found in classical secondary lymphoid organs, which underlies their unique functions of filtration, surveillance, innate-like immune responses, and adaptive immunity within the serous cavities. FALCs are important B cell hubs providing B1 cell-mediated frontline protection against infection and supporting B2 cell-adaptative immune responses. Beyond these beneficial immune responses orchestrated by FALCs, immune cells within VAT play important homeostatic role. Dysregulation of immune cells during obesity and aging leads to chronic pathological "metabolic inflammation", which contributes to the development of cardiometabolic diseases. Here, we examine the emerging and complex functions of B cells in VAT homeostasis and the metabolic complications of obesity, highlighting the potential role that FALCs play and emphasize the areas where further research is needed.
Collapse
Affiliation(s)
- Alexander D Daley
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Danieli MG, Casciaro M, Paladini A, Bartolucci M, Sordoni M, Shoenfeld Y, Gangemi S. Exposome: Epigenetics and autoimmune diseases. Autoimmun Rev 2024; 23:103584. [PMID: 39097180 DOI: 10.1016/j.autrev.2024.103584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
Systemic autoimmune diseases are complex conditions characterized by an immune system dysregulation and an aberrant activation against self-antigens, leading to tissue and organ damage. Even though genetic predisposition plays a role, it cannot fully explain the onset of these diseases, highlighting the significant impact of non-heritable influences such as environment, hormones and infections. The exposome represents all those factors, ranging from chemical pollutants and dietary components to psychological stressors and infectious agents. Epigenetics, which studies changes in gene expression without altering the DNA sequence, is a crucial link between exposome and the development of autoimmune diseases. Key epigenetic mechanisms include DNA methylation, histone modifications, and non-coding RNAs. These epigenetic modifications could provide a potential piece of the puzzle in understanding systemic autoimmune diseases and their connection with the exposome. In this work we have collected the most important and recent evidence in epigenetic changes linked to systemic autoimmune diseases (systemic lupus erythematosus, idiopathic inflammatory myopathies, ANCA-associated vasculitis, and rheumatoid arthritis), emphasizing the roles these changes may play in disease pathogenesis, their potential as diagnostic biomarkers and their prospective in the development of targeted therapies.
Collapse
Affiliation(s)
- Maria Giovanna Danieli
- SOS Immunologia delle Malattie Rare e dei Trapianti, AOU delle Marche & Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, via Tronto 10/A, 60126 Torrette di Ancona, Italy; Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Marco Casciaro
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| | - Alberto Paladini
- PostGraduate School of Internal Medicine, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Martina Bartolucci
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Martina Sordoni
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat Gan 52621, Israel; Reichman University, Herzelia 46101, Israel.
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
8
|
Zhang Y, Lin D, Zheng Y, Chen Y, Yu M, Cui D, Huang M, Su X, Sun Y, Chen Y, Qian Z, Carlson KS, Wen R, Wang D. MiR-9-1 controls osteoblastic regulation of lymphopoiesis. Leukemia 2023; 37:2261-2275. [PMID: 37670087 PMCID: PMC10844005 DOI: 10.1038/s41375-023-02014-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023]
Abstract
The highly conserved MicroRNA-9 (miR-9) family consists of three members. We discovered that miR-9-1 deletion reduced mature miR-9 expression, causing 43% of the mice to display smaller size and postweaning lethality. MiR-9-1-deficient mice with growth defects experienced severe lymphopenia, but other blood cells were unaffected. The lymphopenia wasn't due to defects in hematopoietic progenitors, as mutant bone marrow (BM) cells underwent normal lymphopoiesis after transplantation into wild-type recipients. Additionally, miR-9-1-deficient mice exhibited impaired osteoblastic bone formation, as mutant mesenchymal stem cells (MSCs) failed to differentiate into osteoblastic cells (OBs). RNA sequencing revealed reduced expression of master transcription factors for osteoblastic differentiation, Runt-related transcription factor 2 (Runx2) and Osterix (Osx), and genes related to collagen formation, extracellular matrix organization, and cell adhesion, in miR-9-1-deficient MSCs. Follistatin (Fst), an antagonist of bone morphogenetic proteins (BMPs), was found to be a direct target of miR-9-1. Its deficiency led to the up-regulation of Fst, inhibiting BMP signaling in MSCs, and reducing IL-7 and IGF-1. Thus, miR-9-1 controls osteoblastic regulation of lymphopoiesis by targeting the Fst/BMP/Smad signaling axis.
Collapse
Affiliation(s)
- Yongguang Zhang
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA
- Biomedical Research Center of South China, Fujian Normal University, Fujian, 350117, China
| | - Danfeng Lin
- Biomedical Research Center of South China, Fujian Normal University, Fujian, 350117, China
| | - Yongwei Zheng
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA
| | - Yuhong Chen
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA
| | - Mei Yu
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA
| | - Dongya Cui
- Biomedical Research Center of South China, Fujian Normal University, Fujian, 350117, China
| | - Miaohui Huang
- Biomedical Research Center of South China, Fujian Normal University, Fujian, 350117, China
| | - Xinlin Su
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 205006, China
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, AL, 35294, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, AL, 35294, USA
| | - Zhijian Qian
- Division of Hematology and Oncology, Department of Medicine, Department of Biochemistry and Molecular Biology, the University of Florida, Gainesville, FL, 32610, USA
| | - Karen-Sue Carlson
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Renren Wen
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA.
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI, 53213, USA.
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
9
|
Wallace L, Obeng EA. Noncoding rules of survival: epigenetic regulation of normal and malignant hematopoiesis. Front Mol Biosci 2023; 10:1273046. [PMID: 38028538 PMCID: PMC10644717 DOI: 10.3389/fmolb.2023.1273046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Hematopoiesis is an essential process for organismal development and homeostasis. Epigenetic regulation of gene expression is critical for stem cell self-renewal and differentiation in normal hematopoiesis. Increasing evidence shows that disrupting the balance between self-renewal and cell fate decisions can give rise to hematological diseases such as bone marrow failure and leukemia. Consequently, next-generation sequencing studies have identified various aberrations in histone modifications, DNA methylation, RNA splicing, and RNA modifications in hematologic diseases. Favorable outcomes after targeting epigenetic regulators during disease states have further emphasized their importance in hematological malignancy. However, these targeted therapies are only effective in some patients, suggesting that further research is needed to decipher the complexity of epigenetic regulation during hematopoiesis. In this review, an update on the impact of the epigenome on normal hematopoiesis, disease initiation and progression, and current therapeutic advancements will be discussed.
Collapse
Affiliation(s)
| | - Esther A. Obeng
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
10
|
Nassiri SM, Ahmadi Afshar N, Almasi P. Insight into microRNAs' involvement in hematopoiesis: current standing point of findings. Stem Cell Res Ther 2023; 14:282. [PMID: 37794439 PMCID: PMC10552299 DOI: 10.1186/s13287-023-03504-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
Hematopoiesis is a complex process in which hematopoietic stem cells are differentiated into all mature blood cells (red blood cells, white blood cells, and platelets). Different microRNAs (miRNAs) involve in several steps of this process. Indeed, miRNAs are small single-stranded non-coding RNA molecules, which control gene expression by translational inhibition and mRNA destabilization. Previous studies have revealed that increased or decreased expression of some of these miRNAs by targeting several proto-oncogenes could inhibit or stimulate the myeloid and erythroid lineage commitment, proliferation, and differentiation. During the last decades, the development of molecular and bioinformatics techniques has led to a comprehensive understanding of the role of various miRNAs in hematopoiesis. The critical roles of miRNAs in cell processes such as the cell cycle, apoptosis, and differentiation have been confirmed as well. However, the main contribution of some miRNAs is still unclear. Therefore, it seems undeniable that future studies are required to focus on miRNA activities during various hematopoietic stages and hematological malignancy.
Collapse
Affiliation(s)
- Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Qarib St., Azadi Ave, Tehran, Iran.
| | - Neda Ahmadi Afshar
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Qarib St., Azadi Ave, Tehran, Iran
| | - Parsa Almasi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Qarib St., Azadi Ave, Tehran, Iran
| |
Collapse
|
11
|
Wu Y, Hao S, Xu X, Dong G, Ouyang W, Liu C, Sun HX. A novel computational method enables RNA editome profiling during human hematopoiesis from scRNA-seq data. Sci Rep 2023; 13:10335. [PMID: 37365211 DOI: 10.1038/s41598-023-37325-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
RNA editing is a post-transcriptional modification with a cell-specific manner and important biological implications. Although single-cell RNA-seq (scRNA-seq) is an effective method for studying cellular heterogeneity, it is difficult to detect and study RNA editing events from scRNA-seq data because of the low sequencing coverage. To overcome this, we develop a computational method to systematically identify RNA editing sites of cell types from scRNA-seq data. To demonstrate its effectiveness, we apply it to scRNA-seq data of human hematopoietic stem/progenitor cells (HSPCs) with an annotated lineage differentiation relationship according to previous research and study the impacts of RNA editing on hematopoiesis. The dynamic editing patterns reveal the relevance of RNA editing on different HSPCs. For example, four microRNA (miRNA) target sites on 3' UTR of EIF2AK2 are edited across all HSPC populations, which may abolish the miRNA-mediated inhibition of EIF2AK2. Elevated EIF2AK2 may thus activate the integrated stress response (ISR) pathway to initiate global translational attenuation as a protective mechanism to maintain cellular homeostasis during HSPCs' differentiation. Besides, our findings also indicate that RNA editing plays an essential role in the coordination of lineage commitment and self-renewal of hematopoietic stem cells (HSCs). Taken together, we demonstrate the capacity of scRNA-seq data to exploit RNA editing events of cell types, and find that RNA editing may exert multiple modules of regulation in hematopoietic processes.
Collapse
Affiliation(s)
- Yan Wu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI-Shenzhen, Shenzhen, 518083, China
- BGI-Beijing, Beijing, 102601, China
| | - Shijie Hao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Xiaojing Xu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI-Shenzhen, Shenzhen, 518083, China
- BGI-Beijing, Beijing, 102601, China
| | - Guoyi Dong
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI-Shenzhen, Shenzhen, 518083, China
| | | | - Chao Liu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Hai-Xi Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- BGI-Shenzhen, Shenzhen, 518083, China.
- BGI-Beijing, Beijing, 102601, China.
| |
Collapse
|
12
|
Soares MF, Costa SF, de Freitas JH, Rebech GT, Dos Santos MO, de Lima VMF. MiR-150 regulates the Leishmania infantum parasitic load and granzyme B levels in peripheral blood mononuclear cells of dogs with canine visceral leishmaniosis. Vet Parasitol 2023; 320:109958. [PMID: 37269731 DOI: 10.1016/j.vetpar.2023.109958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023]
Abstract
Leishmania infantum causes visceral leishmaniosis, a neglected tropical disease that can modulate the host immune response by altering the expression of small non-coding RNAs called microRNAs (miRNAs). Some miRNAs are differentially expressed in peripheral blood mononuclear cells (PBMCs) of dogs with canine visceral leishmaniosis (CanL), like the down-regulated miR-150. Even though miR-150 is negatively correlated with L. infantum parasitic load, it is unclear if miR-150 directly affects L. infantum parasitic load and (if so) how this miRNA would contribute to infection. Here, we isolated PBMCs from 14 naturally infected dogs (CanL group) and six healthy dogs (Control group) and treated them in vitro with miR-150 mimic or inhibitor. We measured L. infantum parasitic load using qPCR and compared treatments. We also measured miR-150 in silico predicted target protein levels (STAT1, TNF-α, HDAC8, and GZMB) using flow cytometry or enzyme-linked immunosorbent assays. Increasing miR-150 activity diminished L. infantum parasitic load in CanL PBMCs. We also found that inhibition of miR-150 reduced GZMB (granzyme B) levels. These findings demonstrate that miR-150 plays an important role in L. infantum infection in canine PBMCs, and they merit further studies aiming at drug development.
Collapse
Affiliation(s)
- Matheus Fujimura Soares
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Sidnei Ferro Costa
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Jéssica Henrique de Freitas
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Gabriela Torres Rebech
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Marilene Oliveira Dos Santos
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil.
| |
Collapse
|
13
|
Cucher MA, Mariconti M, Manciulli T, Vola A, Rosenzvit MC, Brehm K, Kamenetzky L, Brunetti E. Circulating Small RNA Profiling of Patients with Alveolar and Cystic Echinococcosis. BIOLOGY 2023; 12:biology12050715. [PMID: 37237528 DOI: 10.3390/biology12050715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/19/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Alveolar (AE) and cystic (CE) echinococcosis are two parasitic diseases caused by the tapeworms Echinococcus multilocularis and E. granulosus sensu lato (s. l.), respectively. Currently, AE and CE are mainly diagnosed by means of imaging techniques, serology, and clinical and epidemiological data. However, no viability markers that indicate parasite state during infection are available. Extracellular small RNAs (sRNAs) are short non-coding RNAs that can be secreted by cells through association with extracellular vesicles, proteins, or lipoproteins. Circulating sRNAs can show altered expression in pathological states; hence, they are intensively studied as biomarkers for several diseases. Here, we profiled the sRNA transcriptomes of AE and CE patients to identify novel biomarkers to aid in medical decisions when current diagnostic procedures are inconclusive. For this, endogenous and parasitic sRNAs were analyzed by sRNA sequencing in serum from disease negative, positive, and treated patients and patients harboring a non-parasitic lesion. Consequently, 20 differentially expressed sRNAs associated with AE, CE, and/or non-parasitic lesion were identified. Our results represent an in-depth characterization of the effect E. multilocularis and E. granulosus s. l. exert on the extracellular sRNA landscape in human infections and provide a set of novel candidate biomarkers for both AE and CE detection.
Collapse
Affiliation(s)
- Marcela A Cucher
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121ABG, Argentina
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Mara Mariconti
- Unit of Infectious and Tropical Diseases, San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Tommaso Manciulli
- Unit of Infectious and Tropical Diseases, San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Ambra Vola
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Mara C Rosenzvit
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121ABG, Argentina
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Klaus Brehm
- Institute for Hygiene and Microbiology, University of Würzburg, 97080 Würzburg, Germany
| | - Laura Kamenetzky
- Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Enrico Brunetti
- Immunology and Infectious Diseases, San Matteo Hospital Foundation, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
14
|
MicroRNA: Crucial modulator in purinergic signalling involved diseases. Purinergic Signal 2023; 19:329-341. [PMID: 35106737 PMCID: PMC9984628 DOI: 10.1007/s11302-022-09840-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022] Open
Abstract
Both microRNAs (miRNAs) and purinergic signalling are widely and respectively expressed in various tissues of different organisms and play vital roles in a variety of physiological and pathological processes. Here, we reviewed the current publications contributed to the relationship of miRNAs and purinergic signalling in cardiovascular diseases, gastrointestinal diseases, neurological diseases, and ophthalmic diseases. We tried to decode the miRNAs-purinergic signalling network of purinergic signalling involved diseases. The evidence indicated that more than 30 miRNAs (miR-22, miR-30, miR-146, miR-150, miR-155, miR-187, etc.) directly or indirectly modulate P1 receptors (A1, A2A, A2B, A3), P2 receptors (P2X1, P2X3, P2X4, P2X7, P2Y2, P2Y6, P2Y12), and ecto-enzymes (CD39, CD73, ADA2); P2X7 and CD73 could be modulated by multiple miRNAs (P2X7: miR-21, miR-22, miR-30, miR-135a, miR-150, miR-186, miR-187, miR-216b; CD73: miR-141, miR-101, miR-193b, miR-340, miR-187, miR-30, miR-422a); miR-187 would be the common miRNA to modulate P2X7 and CD73.
Collapse
|
15
|
Urata S, Yamaguchi S, Nambu A, Sudo K, Nakae S, Yasuda J. The roles of BST-2 in murine B cell development and on virus propagation. Microbiol Immunol 2023; 67:105-113. [PMID: 36604771 DOI: 10.1111/1348-0421.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
The bone marrow (BM) stromal cell antigen-2 (BST-2), also known as tetherin, CD317, PDCA-1, or HM1.24, is a membrane protein overexpressed in several types of tumors and may act as a promising target for cancer treatment via antibody-dependent cellular cytotoxicity. BST-2 is also expressed in human BM stromal cells (BMSC), which support B cell development. While the activity of BST-2 as an antiviral factor has been demonstrated, the expression patterns and the role of BST-2 on B-cell development and activation have not been investigated, especially in vivo. In this study, Bst2 knockout (Bst2-/- ) mice were generated to assess the role of BST-2 on B cell development and activation. It was observed that BST-2 was not expressed in BMSC or all B cell progenitors even in wild-type mice and does not play a significant role in B cell development. In addition, the loss of BST-2 had no effect on B cell activation. Furthermore and in contrast to the well-known antiviral role of BST-2, infection of vesicular stomatitis Indiana virus to the BM cells collected from the Bst2-/- mice produced less infectious virus compared with that from the WT mice. These results suggest that murine BST-2 is different from human BST-2 in the expression pattern, physiological function, in vivo, and might possess positive role on VSV replication.
Collapse
Affiliation(s)
- Shuzo Urata
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan
| | - Sachiko Yamaguchi
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Aya Nambu
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Katsuko Sudo
- Pre-clinical Research Center, Tokyo Medical University, Tokyo, Japan
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan.,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
16
|
Crosstalk of Transcriptional Regulators of Adaptive Immune System and microRNAs: An Insight into Differentiation and Development. Cells 2023; 12:cells12040635. [PMID: 36831302 PMCID: PMC9953855 DOI: 10.3390/cells12040635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/27/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
MicroRNAs (miRNAs), as small regulatory RNA molecules, are involved in gene expression at the post-transcriptional level. Hence, miRNAs contribute to gene regulation of various steps of different cell subsets' differentiation, maturation, and activation. The adaptive immune system arm, which exhibits the most specific immune responses, is also modulated by miRNAs. The generation and maturation of various T-cell subsets concomitant with B-cells is under precise regulation of miRNAs which function directly on the hallmark genes of each cell subset or indirectly through regulation of signaling pathway mediators and/or transcription factors involved in this maturation journey. In this review, we first discussed the origination process of common lymphocyte progenitors from hematopoietic stem cells, which further differentiate into various T-cell subsets under strict regulation of miRNAs and transcription factors. Subsequently, the differentiation of B-cells from common lymphocyte progenitors in bone marrow and periphery were discussed in association with a network of miRNAs and transcription factors.
Collapse
|
17
|
Ishihara S, Sato M, Miyazaki H, Saito H, Sato T, Fujikado N, Sawai S, Kotani A, Katagiri K. Deletion of miR-150 Prevents Spontaneous T Cell Proliferation and the Development of Colitis. GASTRO HEP ADVANCES 2023; 2:487-496. [PMID: 39132043 PMCID: PMC11308117 DOI: 10.1016/j.gastha.2023.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/31/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims To examine the roles of microRNAs in the development of colitis, we conducted the RNA-sequencing studies using RNA derived from normal and colitogenic CD4+ T cells. Colitogenic CD4+ T cells demonstrated the increased expression of miR-150. We focused on the involvement of miR-150 in the colitis. Methods We crossed miR-150 knockout mice and T-cell-specific Rap1KO mice, which is colitis model mice and spontaneously develop the colitis with tubular adenomas in microbiota-dependent manner. Results MiR-150 silencing completely inhibited the expansion of pathogenic Th17 cells and the development of colitis. Conclusion MiR-150 is a potential therapeutic target of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Sayaka Ishihara
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Innovative Medical Science, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| | - Masashi Sato
- Department of Immunology, School of Medicine, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Haruka Miyazaki
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Haruka Saito
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Tsuyoshi Sato
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Noriyuki Fujikado
- Discovery Immunology, Ferring Research Institute, Ferring Pharmaceuticals, San Diego, California
| | - Satoshi Sawai
- Department of Basic Science, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| | - Ai Kotani
- Department of Innovative Medical Science, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Koko Katagiri
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Innovative Medical Science, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
- Department of Basic Science, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Ménoret A, Agliano F, Karginov TA, Karlinsey KS, Zhou B, Vella AT. Antigen-specific downregulation of miR-150 in CD4 T cells promotes cell survival. Front Immunol 2023; 14:1102403. [PMID: 36817480 PMCID: PMC9936563 DOI: 10.3389/fimmu.2023.1102403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
MicroRNA-150 (miR-150) has been shown to play a general role in the immune system, but very little is known about its role on CD4+ T cell responses. During T cell responses against superantigen Staphylococcal Enterotoxin A, miR-150 expression was down-regulated in antigen-specific CD4+ T cells but up-regulated in CD8+ T cells. CD4+ and CD8+ T cell clonal expansion was greater in miR-150-KO mice than in WT mice, but miR-150 selectively repressed IL-2 production in CD4+ T cells. Transcriptome analysis of CD4+ T cells demonstrated that apoptosis and mTOR pathways were highly enriched in the absence of miR-150. Mechanistic studies confirmed that miR-150 promoted apoptosis specifically in antigen-specific CD4+ T cells, but not in bystander CD4+ nor in CD8+ T cells. Furthermore, inhibition of mTOR-linked mitochondrial superoxidedismutase-2 increased apoptosis in miR-150-/- antigen-specific CD4+ T. Thus, miR-150 impacts CD4+ T cell helper activity by attenuating IL-2 production along with clonal expansion, and suppresses superoxidedismutase to promote apoptosis.
Collapse
Affiliation(s)
- Antoine Ménoret
- Department of Immunology, UConn Health, Farmington, CT, United States
| | | | | | | | | | - Anthony T. Vella
- Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
19
|
Hu YZ, Li Q, Wang PF, Li XP, Hu ZL. Multiple functions and regulatory network of miR-150 in B lymphocyte-related diseases. Front Oncol 2023; 13:1140813. [PMID: 37182123 PMCID: PMC10172652 DOI: 10.3389/fonc.2023.1140813] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/06/2023] [Indexed: 05/16/2023] Open
Abstract
MicroRNAs (miRNAs) play vital roles in the post-transcriptional regulation of gene expression. Previous studies have shown that miR-150 is a crucial regulator of B cell proliferation, differentiation, metabolism, and apoptosis. miR-150 regulates the immune homeostasis during the development of obesity and is aberrantly expressed in multiple B-cell-related malignant tumors. Additionally, the altered expression of MIR-150 is a diagnostic biomarker of various autoimmune diseases. Furthermore, exosome-derived miR-150 is considered as prognostic tool in B cell lymphoma, autoimmune diseases and immune-mediated disorders, suggesting miR-150 plays a vital role in disease onset and progression. In this review, we summarized the miR-150-dependent regulation of B cell function in B cell-related immune diseases.
Collapse
Affiliation(s)
- Yue-Zi Hu
- Clinical Laboratory, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Qiao Li
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Fei Wang
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Ping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhao-Lan Hu,
| |
Collapse
|
20
|
Kumar D, Sahoo SS, Chauss D, Kazemian M, Afzali B. Non-coding RNAs in immunoregulation and autoimmunity: Technological advances and critical limitations. J Autoimmun 2023; 134:102982. [PMID: 36592512 PMCID: PMC9908861 DOI: 10.1016/j.jaut.2022.102982] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 01/02/2023]
Abstract
Immune cell function is critically dependent on precise control over transcriptional output from the genome. In this respect, integration of environmental signals that regulate gene expression, specifically by transcription factors, enhancer DNA elements, genome topography and non-coding RNAs (ncRNAs), are key components. The first three have been extensively investigated. Even though non-coding RNAs represent the vast majority of cellular RNA species, this class of RNA remains historically understudied. This is partly because of a lag in technological and bioinformatic innovations specifically capable of identifying and accurately measuring their expression. Nevertheless, recent progress in this domain has enabled a profusion of publications identifying novel sub-types of ncRNAs and studies directly addressing the function of ncRNAs in human health and disease. Many ncRNAs, including circular and enhancer RNAs, have now been demonstrated to play key functions in the regulation of immune cells and to show associations with immune-mediated diseases. Some ncRNAs may function as biomarkers of disease, aiding in diagnostics and in estimating response to treatment, while others may play a direct role in the pathogenesis of disease. Importantly, some are relatively stable and are amenable to therapeutic targeting, for example through gene therapy. Here, we provide an overview of ncRNAs and review technological advances that enable their study and hold substantial promise for the future. We provide context-specific examples by examining the associations of ncRNAs with four prototypical human autoimmune diseases, specifically rheumatoid arthritis, psoriasis, inflammatory bowel disease and multiple sclerosis. We anticipate that the utility and mechanistic roles of these ncRNAs in autoimmunity will be further elucidated in the near future.
Collapse
Affiliation(s)
- Dhaneshwar Kumar
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Subhransu Sekhar Sahoo
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| |
Collapse
|
21
|
Sex-Specific Alterations in Inflammatory MicroRNAs in Mouse Brain and Bone Marrow CD11b+ Cells Following Traumatic Brain Injury. Cell Mol Neurobiol 2023; 43:423-429. [PMID: 34761332 DOI: 10.1007/s10571-021-01164-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/01/2021] [Indexed: 01/12/2023]
Abstract
Sex is a key biological variable in traumatic brain injury (TBI) and plays a significant role in neuroinflammatory responses. However, the molecular mechanisms contributing to this sexually dimorphic neuroinflammatory response remain elusive. Here we describe a significant and previously unreported tissue enrichment and sex-specific alteration of a set of inflammatory microRNAs (miRNAs) in CD11b+ cells of brain and bone marrow isolated from naïve mice as well as mice subjected to TBI. Our data from naïve mice demonstrated that expression levels of miR-146a-5p and miR-150-5p were relatively higher in brain CD11b+ cells, and that miR-155-5p and miR-223-3p were highly enriched in bone marrow CD11b+ cells. Furthermore, while miR-150-5p and miR-155-5p levels were higher in male brain CD11b+ cells, no significant sexual difference was observed for miR-146a-5p and miR-223-3p. However, TBI resulted in sex-specific differential responses of these miRNAs in brain CD11b+ cells. Specifically, miR-223-3p levels in brain CD11b+ cells were markedly elevated in both sexes in response to TBI at 3 and 24 h, with levels in females being significantly higher than males at 24 h. We then focused on analyzing several miR-223-3p targets and inflammation-related marker genes following injury. Corresponding to the greater elevation of miR-223-3p in females, the miR-223-3p targets, TRAF6 and FBXW7 were significantly reduced in females compared to males. Interestingly, anti-inflammatory genes ARG1 and IL4 were higher in females after TBI than in males. These observations suggest miR-223-3p and other inflammatory responsive miRNAs may play a key role in sex-specific neuroinflammatory response following TBI.
Collapse
|
22
|
MicroRNA-150 (miR-150) and Diabetic Retinopathy: Is miR-150 Only a Biomarker or Does It Contribute to Disease Progression? Int J Mol Sci 2022; 23:ijms232012099. [PMID: 36292956 PMCID: PMC9603433 DOI: 10.3390/ijms232012099] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetic retinopathy (DR) is a chronic disease associated with diabetes mellitus and is a leading cause of visual impairment among the working population in the US. Clinically, DR has been diagnosed and treated as a vascular complication, but it adversely impacts both neural retina and retinal vasculature. Degeneration of retinal neurons and microvasculature manifests in the diabetic retina and early stages of DR. Retinal photoreceptors undergo apoptosis shortly after the onset of diabetes, which contributes to the retinal dysfunction and microvascular complications leading to vision impairment. Chronic inflammation is a hallmark of diabetes and a contributor to cell apoptosis, and retinal photoreceptors are a major source of intraocular inflammation that contributes to vascular abnormalities in diabetes. As the levels of microRNAs (miRs) are changed in the plasma and vitreous of diabetic patients, miRs have been suggested as biomarkers to determine the progression of diabetic ocular diseases, including DR. However, few miRs have been thoroughly investigated as contributors to the pathogenesis of DR. Among these miRs, miR-150 is downregulated in diabetic patients and is an endogenous suppressor of inflammation, apoptosis, and pathological angiogenesis. In this review, how miR-150 and its downstream targets contribute to diabetes-associated retinal degeneration and pathological angiogenesis in DR are discussed. Currently, there is no effective treatment to stop or reverse diabetes-caused neural and vascular degeneration in the retina. Understanding the molecular mechanism of the pathogenesis of DR may shed light for the future development of more effective treatments for DR and other diabetes-associated ocular diseases.
Collapse
|
23
|
Souza OF, Popi AF. Role of microRNAs in B-Cell Compartment: Development, Proliferation and Hematological Diseases. Biomedicines 2022; 10:biomedicines10082004. [PMID: 36009551 PMCID: PMC9405569 DOI: 10.3390/biomedicines10082004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
B-cell development is a very orchestrated pathway that involves several molecules, such as transcription factors, cytokines, microRNAs, and also different cells. All these components maintain the ideal microenvironment and control B-cell differentiation. MicroRNAs are small non-coding RNAs that bind to target mRNA to control gene expression. These molecules could circulate in the body in a free form, protein-bounded, or encapsulated into extracellular vesicles, such as exosomes. The comprehension of the role of microRNAs in the B-cell development was possible based on microRNA profile of each B-cell stage and functional studies. Herein, we report the knowledge about microRNAs in the B-cell the differentiation, proliferation, and also in hematological malignancies.
Collapse
|
24
|
D’Agostino DM, Raimondi V, Silic-Benussi M, Ciminale V. MiR-150 in HTLV-1-infection and T-cell transformation. Front Immunol 2022; 13:974088. [PMID: 36072598 PMCID: PMC9442802 DOI: 10.3389/fimmu.2022.974088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Human T-cell leukemia virus-1 (HTLV-1) is a retrovirus that persistently infects CD4+ T-cells, and is the causative agent of adult T-cell leukemia/lymphoma (ATLL), tropical spastic paraparesis/HTLV-1-associated myelopathy (TSP/HAM) and several inflammatory diseases. T-cell transformation by HTLV-1 is driven by multiple interactions between viral regulatory proteins and host cell pathways that govern cell proliferation and survival. Studies performed over the last decade have revealed alterations in the expression of many microRNAs in HTLV-1-infected cells and ATLL cells, and have identified several microRNA targets with roles in the viral life cycle and host cell turnover. This review centers on miR-150-5p, a microRNA whose expression is temporally regulated during lymphocyte development and altered in several hematological malignancies. The levels of miR-150-5p are reduced in many HTLV-1-transformed- and ATLL-derived cell lines. Experiments in these cell lines showed that downregulation of miR-150-5p results in activation of the transcription factor STAT1, which is a direct target of the miRNA. However, data on miR-150-5p levels in freshly isolated ATLL samples are suggestive of its upregulation compared to controls. These apparently puzzling findings highlight the need for more in-depth studies of the role of miR-150-5p in HTLV-1 infection and pathogenesis based on knowledge of miR-150-5p-target mRNA interactions and mechanisms regulating its function in normal leukocytes and hematologic neoplasms.
Collapse
Affiliation(s)
- Donna M. D’Agostino
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Istituto Oncologico Veneto (IOV)- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padova, Italy
- *Correspondence: Donna M. D’Agostino, ; Vincenzo Ciminale,
| | - Vittoria Raimondi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Micol Silic-Benussi
- Istituto Oncologico Veneto (IOV)- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padova, Italy
| | - Vincenzo Ciminale
- Istituto Oncologico Veneto (IOV)- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- *Correspondence: Donna M. D’Agostino, ; Vincenzo Ciminale,
| |
Collapse
|
25
|
Hao X, Luan J, Jiao C, Ma C, Feng Z, Zhu L, Zhang Y, Fu J, Lai E, Zhang B, Wang Y, Kopp JB, Pi J, Zhou H. LNA-anti-miR-150 alleviates renal interstitial fibrosis by reducing pro-inflammatory M1/M2 macrophage polarization. Front Immunol 2022; 13:913007. [PMID: 35990680 PMCID: PMC9389080 DOI: 10.3389/fimmu.2022.913007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022] Open
Abstract
Renal interstitial fibrosis (RIF) is a common pathological feature contributing to chronic injury and maladaptive repair following acute kidney injury. Currently, there is no effective therapy for RIF. We have reported that locked nuclear acid (LNA)-anti-miR-150 antagonizes pro-fibrotic pathways in human renal tubular cells by regulating the suppressor of cytokine signal 1 (SOCS1)/Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. In the present study, we aimed to clarify whether LNA-anti-miR-150 attenuates folic acid-induced RIF mice by regulating this pathway and by reducing pro-inflammatory M1/M2 macrophage polarization. We found that renal miR-150 was upregulated in folic acid-induced RIF mice at day 30 after injection. LNA-anti-miR-150 alleviated the degree of RIF, as shown by periodic acid–Schiff and Masson staining and by the expression of pro-fibrotic proteins, including alpha-smooth muscle actin and fibronectin. In RIF mice, SOCS1 was downregulated, and p-JAK1 and p-STAT1 were upregulated. LNA-anti-miR-150 reversed the changes in renal SOCS1, p-JAK1, and p-STAT1 expression. In addition, renal infiltration of total macrophages, pro-inflammatory M1 and M2 macrophages as well as their secreted cytokines were increased in RIF mice compared to control mice. Importantly, in folic acid-induced RIF mice, LNA-anti-miR-150 attenuated the renal infiltration of total macrophages and pro-inflammatory subsets, including M1 macrophages expressing CD11c and M2 macrophages expressing CD206. We conclude that the anti-renal fibrotic role of LNA-anti-miR-150 in folic acid-induced RIF mice may be mediated by reducing pro-inflammatory M1 and M2 macrophage polarization via the SOCS1/JAK1/STAT1 pathway.
Collapse
Affiliation(s)
- Xiangnan Hao
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junjun Luan
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Congcong Jiao
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cong Ma
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zixuan Feng
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lingzi Zhu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yixiao Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Enyin Lai
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Beiru Zhang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanqiu Wang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jeffrey B. Kopp
- Kidney Disease Section, NIDDK/NIH, Bethesda, MD, United States
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Hua Zhou,
| |
Collapse
|
26
|
Su Y, Li Z, Rang X, Wang Y, Fu J. Integrated Analysis and Identification of CSF-Derived Risk miRNAs and Pivotal Genes in Multiple Sclerosis. J Mol Neurosci 2022; 72:1916-1928. [PMID: 35819635 DOI: 10.1007/s12031-022-02007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/25/2022] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is a common chronic autoimmune disorder of the central nervous system that predominantly affects young adults. Mounting evidence indicates that deregulation of microRNAs (miRNAs) in cerebrospinal fluid (CSF) has been implicated in MS as a potential biomarker. However, comprehensive assessments of CSF miRNAs and their target genes are lacking. Here, aberrantly expressed CSF miRNAs of MS patients were obtained from numerous studies by manual search. With detailed information on these miRNAs, we utilized online databases to screen out immune-related target genes and further performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. To identify MS high-risk pathways and pivotal genes, pathway crosstalk and pathway-gene networks were constructed, followed by the establishment of a protein-protein interaction (PPI) network. The datasets collected from ArrayExpress were used to assess pivotal genes. Overall, 21 MS-related CSF miRNAs were included in this study. Subsequently, we identified 469 MS-related genes and 14 high-risk pathways. In the pathway-gene network, 27 critical MS-related genes participated in at least half of the high-risk pathways, and these genes were used to identify pivotal genes. Finally, miR-150, miR-328, and miR-34c-5p were determined to be risk miRNAs via the regulation of the pivotal risk genes MAPK1, AKT1, and VEGFA. Among them, VEGFA was validated to be significantly decreased in the CSF cells of MS patients by transcriptomic datasets. These findings may provide potential biomarkers or therapeutic targets and help elucidate the molecular mechanisms underlying the pathogenesis of MS.
Collapse
Affiliation(s)
- Yingchao Su
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Xinming Rang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Yifei Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Jin Fu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China.
| |
Collapse
|
27
|
Gramantieri L, Fornari F, Giovannini C, Trerè D. MicroRNAs at the Crossroad between Immunoediting and Oncogenic Drivers in Hepatocellular Carcinoma. Biomolecules 2022; 12:biom12070930. [PMID: 35883486 PMCID: PMC9313100 DOI: 10.3390/biom12070930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary In recent years, treatments enhancing the antitumor immune response have revealed a new promising approach for advanced hepatocellular carcinoma (HCC). Beside favorable results in about one third of patients, much still remains to be done to face primary nonresponse, early, and late disease reactivation. Understanding the mechanisms underneath immune system modulation by immune checkpoint inhibitors in HCC might give additional opportunities for patient selection and combined approaches. MicroRNAs have emerged as relevant modulators of cancer cell hallmarks, including aberrant proliferation, invasion and migration capabilities, epithelial-to-mesenchymal transition, and glycolytic metabolism. At the same time, they contribute to the immune system development, response, and programs activation, with particular regard towards regulatory functions. Thus, miRNAs are relevant not only in cancer cells’ biology, but also in the immune response and interplay between cancer, microenvironment, and immune system. Abstract Treatments aimed to reverse the tumor-induced immune tolerance represent a promising approach for advanced hepatocellular carcinoma (HCC). Notwithstanding, primary nonresponse, early, and late disease reactivation still represent major clinical challenges. Here, we focused on microRNAs (miRNAs) acting both as modulators of cancer cell hallmarks and immune system response. We outlined the bidirectional function that some oncogenic miRNAs play in the differentiation and program activation of the immune system development and, at the same time, in the progression of HCC. Indeed, the multifaceted spectrum of miRNA targets allows the modulation of both immune-associated factors and oncogenic or tumor suppressor drivers at the same time. Understanding the molecular changes contributing to disease onset, progression, and resistance to treatments might help to identify possible novel biomarkers for selecting patient subgroups, and to design combined tailored treatments to potentiate antitumor approaches. Preliminary findings seem to argue in favor of a bidirectional function of some miRNAs, which enact an effective modulation of molecular pathways driving oncogenic and immune-skipping phenotypes associated with cancer aggressiveness. The identification of these miRNAs and the characterization of their ‘dual’ role might help to unravel novel biomarkers identifying those patients more likely to respond to immune checkpoint inhibitors and to identify possible therapeutic targets with both antitumor and immunomodulatory functions. In the present review, we will focus on the restricted panel of miRNAs playing a bidirectional role in HCC, influencing oncogenic and immune-related pathways at once. Even though this field is still poorly investigated in HCC, it might represent a source of candidate molecules acting as both biomarkers and therapeutic targets in the setting of immune-based treatments.
Collapse
Affiliation(s)
- Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Fornari
- Department for Life Quality Studies (QuVi), University of Bologna, 47921 Rimini, Italy
- Centre for Applied Biomedical Research-CRBA, University of Bologna, IRCCS St. Orsola Hospital, 40138 Bologna, Italy
| | - Catia Giovannini
- Centre for Applied Biomedical Research-CRBA, University of Bologna, IRCCS St. Orsola Hospital, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy
| | - Davide Trerè
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy
- Departmental Program in Laboratory Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
28
|
Small noncoding RNAs play superior roles in maintaining hematopoietic stem cell homeostasis. BLOOD SCIENCE 2022; 4:125-132. [DOI: 10.1097/bs9.0000000000000123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022] Open
|
29
|
Abstract
Obesity is a prevalent health risk by inducing chronic, low-grade inflammation and insulin resistance, in part from adipose tissue inflammation perpetuated by activated B cells and other resident immune cells. However, regulatory mechanisms controlling B-cell actions in adipose tissue remain poorly understood, limiting therapeutic innovations. MicroRNAs are potent regulators of immune cell dynamics through fine-tuning a network of downstream genes in multiple signaling pathways. In particular, miR-150 is crucial to B-cell development and suppresses obesity-associated inflammation via regulating adipose tissue B-cell function. Herein, we review the effect of microRNAs on B-cell development, activation, and function and highlight miR-150-regulated B-cell actions during obesity which modulate systemic inflammation and insulin resistance. In this way, we hope to promote translational discoveries that mitigate obesity-induced health risks by targeting microRNA-regulated B-cell actions.
Collapse
|
30
|
Plantone D, Pardini M, Locci S, Nobili F, De Stefano N. B Lymphocytes in Alzheimer's Disease-A Comprehensive Review. J Alzheimers Dis 2022; 88:1241-1262. [PMID: 35754274 DOI: 10.3233/jad-220261] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) represents the most common type of neurodegenerative dementia and is characterized by extracellular amyloid-β (Aβ) deposition, pathologic intracellular tau protein tangles, and neuronal loss. Increasing evidence has been accumulating over the past years, supporting a pivotal role of inflammation in the pathogenesis of AD. Microglia, monocytes, astrocytes, and neurons have been shown to play a major role in AD-associated inflammation. However recent studies showed that the role of both T and B lymphocytes may be important. In particular, B lymphocytes are the cornerstone of humoral immunity, they constitute a heterogenous population of immune cells, being their mature subsets significantly impacted by the inflammatory milieu. The role of B lymphocytes on AD pathogenesis is gaining interest for several reasons. Indeed, the majority of elderly people develop the process of "inflammaging", which is characterized by increased blood levels of proinflammatory molecules associated with an elevated susceptibility to chronic diseases. Epitope-specific alteration pattern of naturally occurring antibodies targeting the amino-terminus and the mid-domain of Aβ in both plasma and cerebrospinal fluid has been described in AD patients. Moreover, a possible therapeutic role of B lymphocytes depletion was recently demonstrated in murine AD models. Interestingly, active immunization against Aβ and tau, one of the main therapeutic strategies under investigation, depend on B lymphocytes. Finally. several molecules being tested in AD clinical trials can modify the homeostasis of B cells. This review summarizes the evidence supporting the role of B lymphocytes in AD from the pathogenesis to the possible therapeutic implications.
Collapse
Affiliation(s)
- Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy.,Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Sara Locci
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Flavio Nobili
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy.,Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
31
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
32
|
Comprehensive Characterization of Platelet-Enriched MicroRNAs as Biomarkers of Platelet Activation. Cells 2022; 11:cells11081254. [PMID: 35455934 PMCID: PMC9030873 DOI: 10.3390/cells11081254] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
Dysregulation of platelet function is causally connected to thrombus formation and cardiovascular diseases. Therefore, assessing platelet reactivity is crucial. However, current platelet function tests come with pitfalls, limiting clinical use. Plasma miRNA signatures have been suggested as novel biomarkers for predicting/diagnosing cardiovascular diseases and monitoring antiplatelet therapy. Here, we provide results from a comprehensive study on the feasibility of using circulatory platelet miRNAs as surrogate markers of platelet activation. We performed small RNA-Seq on different blood cell types to confirm known and identify novel platelet-enriched miRNAs and validated a panel of 16 miRNAs using RT-qPCR. To identify the main carrier of these blood-based platelet miRNAs, we enriched and analyzed distinct microvesicle populations. Platelets were stimulated with GPVI and P2Y12 agonists in vitro to monitor the release of the selected miRNAs following activation. Finally, the miRNA panel was also measured in plasma from mice undergoing the Folts intervention (recurrent thrombus formation in the carotid artery). Applying an unbiased bioinformatics-supported workflow to our NGS data, we were able to confirm a panel of previously established miRNA biomarker candidates and identify three new candidates (i.e., miR-199a-3p, miR-151a-5p, and miR-148b-3p). Basal levels of platelet-derived miRNAs in plasma were mainly complexed with proteins, not extracellular vesicles. We show that changes in miRNA levels due to platelet activation are detectable using RT-qPCR. In addition, we highlight limitations of studying the in vitro release of miRNAs from platelets. In vivo thrombosis resulted in significant elevations of platelet-derived miRNA levels in mice. In conclusion, we provide in-depth evidence that activated platelets release miRNAs, resulting in measurable changes in circulatory miRNA levels, rendering them promising biomarker candidates.
Collapse
|
33
|
Perdaens O, van Pesch V. Molecular Mechanisms of Immunosenescene and Inflammaging: Relevance to the Immunopathogenesis and Treatment of Multiple Sclerosis. Front Neurol 2022; 12:811518. [PMID: 35281989 PMCID: PMC8913495 DOI: 10.3389/fneur.2021.811518] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/27/2021] [Indexed: 12/18/2022] Open
Abstract
Aging is characterized, amongst other features, by a complex process of cellular senescence involving both innate and adaptive immunity, called immunosenescence and associated to inflammaging, a low-grade chronic inflammation. Both processes fuel each other and partially explain increasing incidence of cancers, infections, age-related autoimmunity, and vascular disease as well as a reduced response to vaccination. Multiple sclerosis (MS) is a lifelong disease, for which considerable progress in disease-modifying therapies (DMTs) and management has improved long-term survival. However, disability progression, increasing with age and disease duration, remains. Neurologists are now involved in caring for elderly MS patients, with increasing comorbidities. Aging of the immune system therefore has relevant implications for MS pathogenesis, response to DMTs and the risks mediated by these treatments. We propose to review current evidence regarding markers and molecular mechanisms of immunosenescence and their relevance to understanding MS pathogenesis. We will focus on age-related changes in the innate and adaptive immune system in MS and other auto-immune diseases, such as systemic lupus erythematosus and rheumatoid arthritis. The consequences of these immune changes on MS pathology, in interaction with the intrinsic aging process of central nervous system resident cells will be discussed. Finally, the impact of immunosenescence on disease evolution and on the safety and efficacy of current DMTs will be presented.
Collapse
Affiliation(s)
- Océane Perdaens
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent van Pesch
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- *Correspondence: Vincent van Pesch
| |
Collapse
|
34
|
Del Gaizo M, Sergio I, Lazzari S, Cialfi S, Pelullo M, Screpanti I, Felli MP. MicroRNAs as Modulators of the Immune Response in T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2022; 23:829. [PMID: 35055013 PMCID: PMC8776227 DOI: 10.3390/ijms23020829] [Citation(s) in RCA: 4] [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: 12/03/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 02/05/2023] Open
Abstract
Acute lymphoblastic leukaemia (ALL) is an aggressive haematological tumour driven by the malignant transformation and expansion of B-cell (B-ALL) or T-cell (T-ALL) progenitors. The evolution of T-ALL pathogenesis encompasses different master developmental pathways, including the main role played by Notch in cell fate choices during tissue differentiation. Recently, a growing body of evidence has highlighted epigenetic changes, particularly the altered expression of microRNAs (miRNAs), as a critical molecular mechanism to sustain T-ALL. The immune response is emerging as key factor in the complex multistep process of cancer but the role of miRNAs in anti-leukaemia response remains elusive. In this review we analyse the available literature on miRNAs as tuners of the immune response in T-ALL, focusing on their role in Natural Killer, T, T-regulatory and Myeloid-derived suppressor cells. A better understanding of this molecular crosstalk may provide the basis for the development of potential immunotherapeutic strategies in the leukemia field.
Collapse
Affiliation(s)
- Martina Del Gaizo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Roma, Italy; (M.D.G.); (S.L.); (S.C.)
| | - Ilaria Sergio
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Roma, Italy;
| | - Sara Lazzari
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Roma, Italy; (M.D.G.); (S.L.); (S.C.)
| | - Samantha Cialfi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Roma, Italy; (M.D.G.); (S.L.); (S.C.)
| | - Maria Pelullo
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161 Rome, Italy;
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Roma, Italy; (M.D.G.); (S.L.); (S.C.)
| | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Roma, Italy;
| |
Collapse
|
35
|
Zhou X, Dai H, Jiang H, Rui H, Liu W, Dong Z, Zhang N, Zhao Q, Feng Z, Hu Y, Hou F, Zheng Y, Liu B. MicroRNAs: Potential mediators between particulate matter 2.5 and Th17/Treg immune disorder in primary membranous nephropathy. Front Pharmacol 2022; 13:968256. [PMID: 36210816 PMCID: PMC9532747 DOI: 10.3389/fphar.2022.968256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Primary membranous nephropathy (PMN), is an autoimmune glomerular disease and the main reason of nephrotic syndrome in adults. Studies have confirmed that the incidence of PMN increases yearly and is related to fine air pollutants particulate matter 2.5 (PM2.5) exposure. These imply that PM2.5 may be associated with exposure to PMN-specific autoantigens, such as the M-type receptor for secretory phospholipase A2 (PLA2R1). Emerging evidence indicates that Th17/Treg turns to imbalance under PM2.5 exposure, but the molecular mechanism of this process in PMN has not been elucidated. As an important indicator of immune activity in multiple diseases, Th17/Treg immune balance is sensitive to antigens and cellular microenvironment changes. These immune pathways play an essential role in the disease progression of PMN. Also, microRNAs (miRNAs) are susceptible to external environmental stimulation and play link role between the environment and immunity. The contribution of PM2.5 to PMN may induce Th17/Treg imbalance through miRNAs and then produce epigenetic affection. We summarize the pathways by which PM2.5 interferes with Th17/Treg immune balance and attempt to explore the intermediary roles of miRNAs, with a particular focus on the changes in PMN. Meanwhile, the mechanism of PM2.5 promoting PLA2R1 exposure is discussed. This review aims to clarify the potential mechanism of PM2.5 on the pathogenesis and progression of PMN and provide new insights for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Xiaoshan Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Haoran Dai
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Hanxue Jiang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Hongliang Rui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Institute of Chinese Medicine, Beijing, China
| | - Wenbin Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhaocheng Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Na Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zhendong Feng
- Pinggu Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Yuehong Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Fanyu Hou
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Zheng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
36
|
Ahmad Z, Somanath PR. AKT Isoforms in the Immune Response in Cancer. Curr Top Microbiol Immunol 2022; 436:349-366. [PMID: 36243852 DOI: 10.1007/978-3-031-06566-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
AKT is a protein kinase that exists in three isoforms: AKT1, AKT2, and AKT3. Though similar in structure, these isoforms display different effects. AKT is activated downstream of PI3K, and together, this signaling pathway helps regulate cellular processes including cell growth, proliferation, metabolism, survival, and apoptosis. Disruption in these pathways has been associated with disorders including cardiovascular diseases, developmental disorders, inflammatory responses, autoimmune diseases, neurologic disorders, type 2 diabetes, and several cancers. In cancer, deregulation in the PI3K/AKT pathway can be manifested as tumorigenesis, pathological angiogenesis, and metastasis. Increased activity has been correlated with tumor progression and resistance to cancer treatments. Recent studies have suggested that inhibition of the PI3K/AKT pathway plays a significant role in the development, expansion, and proliferation of cells of the immune system. Additionally, AKT has been found to play an important role in differentiating regulatory T cells, activating B cells, and augmenting tumor immunosurveillance. This emphasizes AKT as a potential target for inhibition in cancer therapy. This chapter reviews AKT structure and regulation, its different isoforms, its role in immune cells, and its modulation in oncotherapy.
Collapse
Affiliation(s)
- Zayd Ahmad
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Georgia Cancer Center, Vascular Biology Center and Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
37
|
Fernandes M, Marques H, Teixeira AL, Medeiros R. Competitive Endogenous RNA Network Involving miRNA and lncRNA in Non-Hodgkin Lymphoma: Current Advances and Clinical Perspectives. Biomedicines 2021; 9:1934. [PMID: 34944752 PMCID: PMC8698845 DOI: 10.3390/biomedicines9121934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
Non-Hodgkin lymphoma (NHL) is a heterogeneous malignancy with variable patient outcomes. There is still a lack of understanding about the different players involved in lymphomagenesis, and the identification of new diagnostic and prognostic biomarkers is urgent. MicroRNAs and long non-coding RNAs emerged as master regulators of B-cell development, and their deregulation has been associated with the initiation and progression of lymphomagenesis. They can function by acting alone or, as recently proposed, by creating competing endogenous RNA (ceRNA) networks. Most studies have focused on individual miRNAs/lncRNAs function in lymphoma, and there is still limited data regarding their interactions in lymphoma progression. The study of miRNAs' and lncRNAs' deregulation in NHL, either alone or as ceRNAs networks, offers new insights into the molecular mechanisms underlying lymphoma pathogenesis and opens a window of opportunity to identify potential diagnostic and prognostic biomarkers. In this review, we summarized the current knowledge regarding the role of miRNAs and lncRNAs in B-cell lymphoma, including their interactions and regulatory networks. Finally, we summarized the studies investigating the potential of miRNAs and lncRNAs as clinical biomarkers, with a special focus on the circulating profiles, to be applied as a non-invasive, easy-to-obtain, and reproducible liquid biopsy for dynamic management of NHL patients.
Collapse
Affiliation(s)
- Mara Fernandes
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (A.L.T.)
- Research Department of the Portuguese League against Cancer Regional Nucleus of the North (LPCC-NRN), 4200-177 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Herlander Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s–PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
- Department of Oncology, Hospital de Braga, 4710-243 Braga, Portugal
- CINTESIS, Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (A.L.T.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-513 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (A.L.T.)
- Research Department of the Portuguese League against Cancer Regional Nucleus of the North (LPCC-NRN), 4200-177 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-513 Porto, Portugal
- Biomedical Research Center (CEBIMED), Faculty of Health Sciences of Fernando Pessoa University (UFP), 4249-004 Porto, Portugal
| |
Collapse
|
38
|
Bolouri H, Ries R, Pardo L, Hylkema T, Zhou W, Smith JL, Leonti A, Loken M, Farrar JE, Triche TJ, Meshinchi S. A B-cell developmental gene regulatory network is activated in infant AML. PLoS One 2021; 16:e0259197. [PMID: 34793513 PMCID: PMC8601427 DOI: 10.1371/journal.pone.0259197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022] Open
Abstract
Infant Acute Myeloid Leukemia (AML) is a poorly-addressed, heterogeneous malignancy distinguished by surprisingly few mutations per patient but accompanied by myriad age-specific translocations. These characteristics make treatment of infant AML challenging. While infant AML is a relatively rare disease, it has enormous impact on families, and in terms of life-years-lost and life limiting morbidities. To better understand the mechanisms that drive infant AML, we performed integrative analyses of genome-wide mRNA, miRNA, and DNA-methylation data in diagnosis-stage patient samples. Here, we report the activation of an onco-fetal B-cell developmental gene regulatory network in infant AML. AML in infants is genomically distinct from AML in older children/adults in that it has more structural genomic aberrations and fewer mutations. Differential expression analysis of ~1500 pediatric AML samples revealed a large number of infant-specific genes, many of which are associated with B cell development and function. 18 of these genes form a well-studied B-cell gene regulatory network that includes the epigenetic regulators BRD4 and POU2AF1, and their onco-fetal targets LIN28B and IGF2BP3. All four genes are hypo-methylated in infant AML. Moreover, micro-RNA Let7a-2 is expressed in a mutually exclusive manner with its target and regulator LIN28B. These findings suggest infant AML may respond to bromodomain inhibitors and immune therapies targeting CD19, CD20, CD22, and CD79A.
Collapse
Affiliation(s)
- Hamid Bolouri
- Center for Systems Immunology, Benaroya Research Institute, Seattle, WA, United States of America
- * E-mail: (HB); (SM)
| | - Rhonda Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Laura Pardo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Hematologics Inc., Seattle, WA, United States of America
| | - Tiffany Hylkema
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Wanding Zhou
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | - Jenny L. Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Amanda Leonti
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Michael Loken
- Hematologics Inc., Seattle, WA, United States of America
| | - Jason E. Farrar
- Arkansas Children’s Research Institute and University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Timothy J. Triche
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- * E-mail: (HB); (SM)
| |
Collapse
|
39
|
Liang Y, Wang L. Inflamma-MicroRNAs in Alzheimer's Disease: From Disease Pathogenesis to Therapeutic Potentials. Front Cell Neurosci 2021; 15:785433. [PMID: 34776873 PMCID: PMC8581643 DOI: 10.3389/fncel.2021.785433] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 01/16/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of senile dementia. Although AD research has made important breakthroughs, the pathogenesis of this disease remains unclear, and specific AD diagnostic biomarkers and therapeutic strategies are still lacking. Recent studies have demonstrated that neuroinflammation is involved in AD pathogenesis and is closely related to other health effects. MicroRNAs (miRNAs) are a class of endogenous short sequence non-coding RNAs that indirectly inhibit translation or directly degrade messenger RNA (mRNA) by specifically binding to its 3′ untranslated region (UTR). Several broadly expressed miRNAs including miR-21, miR-146a, and miR-155, have now been shown to regulate microglia/astrocytes activation. Other miRNAs, including miR-126 and miR-132, show a progressive link to the neuroinflammatory signaling. Therefore, further studies on these inflamma-miRNAs may shed light on the pathological mechanisms of AD. The differential expression of inflamma-miRNAs (such as miR-29a, miR-125b, and miR-126-5p) in the peripheral circulation may respond to AD progression, similar to inflammation, and therefore may become potential diagnostic biomarkers for AD. Moreover, inflamma-miRNAs could also be promising therapeutic targets for AD treatment. This review provides insights into the role of inflamma-miRNAs in AD, as well as an overview of general inflamma-miRNA biology, their implications in pathophysiology, and their potential roles as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yuanyuan Liang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Payet M, Dargai F, Gasque P, Guillot X. Epigenetic Regulation (Including Micro-RNAs, DNA Methylation and Histone Modifications) of Rheumatoid Arthritis: A Systematic Review. Int J Mol Sci 2021; 22:ijms222212170. [PMID: 34830057 PMCID: PMC8625518 DOI: 10.3390/ijms222212170] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 11/16/2022] Open
Abstract
The inflammatory reaction in rheumatoid arthritis (RA) is controlled by major epigenetic modifications that modulate the phenotype of synovial and immune cells. The aim of this work was to perform a systematic review focusing on miR expression, DNA methylation and histone modifications in RA. We demonstrated that, in human samples, the expressions of miR-155, miR-146a and miR-150 were significantly decreased while the expression of miR-410-3p was significantly increased in the RA group. Moreover, miR-146a significantly decreased pro-autoimmune IL-17 cytokine expression in RA. In a murine model, miR-34a inhibition can ameliorate the arthritis score. However, this evidence remain critically insufficient to support current therapeutic applications in RA patients.
Collapse
Affiliation(s)
- Melissa Payet
- Research Unit ‘Etudes en Pharmaco-Immunologie’ UR EPI, Université de la Réunion, 97400 Réunion, France; (P.G.); (X.G.)
- Correspondence:
| | - Farouk Dargai
- Orthopedic Clinical Department, CHU Bellepierre, Reunion University Hospital, 97400 Réunion, France;
| | - Philippe Gasque
- Research Unit ‘Etudes en Pharmaco-Immunologie’ UR EPI, Université de la Réunion, 97400 Réunion, France; (P.G.); (X.G.)
- Immunology Laboratory (LICE-OI), CHU Bellepierre, Reunion University Hospital, 97400 Réunion, France
| | - Xavier Guillot
- Research Unit ‘Etudes en Pharmaco-Immunologie’ UR EPI, Université de la Réunion, 97400 Réunion, France; (P.G.); (X.G.)
- Rheumatology Clinical Department, CHU Bellepierre, Reunion University Hospital, 97400 Réunion, France
| |
Collapse
|
41
|
Yadav M, Liu J, Song F, Mo X, Jacob NR, Xu-Welliver M, Chakravarti A, Jacob NK. Utility of circulating microRNA-150 for rapid evaluation of bone marrow depletion after radiation, and efficiency of bone marrow reconstitution. Int J Radiat Oncol Biol Phys 2021; 112:964-974. [PMID: 34767935 DOI: 10.1016/j.ijrobp.2021.10.150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Total body irradiation (TBI) is a common myeloablative preparative regimen used in acute myeloid and lymphoblastic leukemia patients prior to allogenic hematopoietic stem cell transplantation (HSCT). The inefficient clearance of tumor cells and radiation-induced toxicity to normal tissues is attributed to relapse and morbidity in a significant fraction of patients. Developing biomarkers that provide an individual's physiological response to radiation will allow personalized treatment and follow-up. We investigated the utility of circulating microRNA150-5p (miR150) for evaluation of radiation dose response. MATERIALS AND METHODS Age-, gender-, and strain-matched wild type and miR150 null (knock out, KO) mice were subjected to TBI and evaluated for the impact of circulating miR150 expression on survival and hematological endpoints. Dose- and time-dependent changes of the miR150 level in bone marrow were assessed using flow cytometry. The functional roles of miR150 in cellular response to radiation were evaluated using apoptosis assay. miR150 expression in leukemic cell lines and in blood collected from leukemia patients with diverse outcomes were evaluated by quantitative RT-PCR. RESULTS Absence of miR150 in mice conferred resistance to radiation injury and resulted in accelerated recovery of lymphoid and myeloid cells after ablative or partially ablative TBI in mice. Overexpression of miR150 resulted in a higher percentage of cells at G2/M phases of cell cycle which is associated with increased sensitivity and susceptibility to apoptotic cell death after radiation. Levels of circulating miR150 were found to be decreased after radiation in leukemia patients and exhibited an inverse correlation with recurrence. CONCLUSION Current study demonstrates the utility of a miR150-based blood test for rapid evaluation of the efficiency of marrow ablation and recovery following radiation and HSCT. The internally controlled blood test will potentially provide near real-time evaluation of functional marrow that will allow optimal dosing based on an individual's physiological response to radiation.
Collapse
Affiliation(s)
- Marshleen Yadav
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Joseph Liu
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Feifei Song
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Xiaokui Mo
- Center for Biostatistics, Ohio State University, Columbus, Ohio
| | - Nitya R Jacob
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Meng Xu-Welliver
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Department of Radiation Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Arnab Chakravarti
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Department of Radiation Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Naduparambil K Jacob
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Department of Radiation Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
42
|
Attaway M, Chwat-Edelstein T, Vuong BQ. Regulatory Non-Coding RNAs Modulate Transcriptional Activation During B Cell Development. Front Genet 2021; 12:678084. [PMID: 34721515 PMCID: PMC8551670 DOI: 10.3389/fgene.2021.678084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/29/2021] [Indexed: 01/07/2023] Open
Abstract
B cells play a significant role in the adaptive immune response by secreting immunoglobulins that can recognize and neutralize foreign antigens. They develop from hematopoietic stem cells, which also give rise to other types of blood cells, such as monocytes, neutrophils, and T cells, wherein specific transcriptional programs define the commitment and subsequent development of these different cell lineages. A number of transcription factors, such as PU.1, E2A, Pax5, and FOXO1, drive B cell development. Mounting evidence demonstrates that non-coding RNAs, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), modulate the expression of these transcription factors directly by binding to the mRNA coding for the transcription factor or indirectly by modifying cellular pathways that promote expression of the transcription factor. Conversely, these transcription factors upregulate expression of some miRNAs and lncRNAs to determine cell fate decisions. These studies underscore the complex gene regulatory networks that control B cell development during hematopoiesis and identify new regulatory RNAs that require additional investigation. In this review, we highlight miRNAs and lncRNAs that modulate the expression and activity of transcriptional regulators of B lymphopoiesis and how they mediate this regulation.
Collapse
Affiliation(s)
- Mary Attaway
- Department of Biology, The City College of New York, New York, NY, United States
| | - Tzippora Chwat-Edelstein
- Department of Biology, The City College of New York, New York, NY, United States.,Macaulay Honors College, New York, NY, United States
| | - Bao Q Vuong
- Department of Biology, The City College of New York, New York, NY, United States.,The Graduate Center, The City University of New York, New York, NY, United States
| |
Collapse
|
43
|
Borbet TC, Hines MJ, Koralov SB. MicroRNA regulation of B cell receptor signaling. Immunol Rev 2021; 304:111-125. [PMID: 34523719 PMCID: PMC8616848 DOI: 10.1111/imr.13024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022]
Abstract
B lymphocytes play a central role in host immune defense. B cell receptor (BCR) signaling regulates survival, proliferation, and differentiation of B lymphocytes. Signaling through the BCR signalosome is a multi-component cascade that is tightly regulated and is important in the coordination of B cell differentiation and function. At different stages of development, B cells that have BCRs recognizing self are eliminated to prevent autoimmunity. microRNAs (miRNAs) are small single-stranded non-coding RNAs that contribute to post-transcriptional regulation of gene expression and have been shown to orchestrate cell fate decisions through the regulation of lineage-specific transcriptional profiles. Studies have identified miRNAs to be crucial for B cell development in the bone marrow and their subsequent population of the peripheral immune system. In this review, we focus on the role of miRNAs in the regulation of BCR signaling as it pertains to B lymphocyte development and function. In particular, we discuss the most recent studies describing the role of miRNAs in the regulation of both early B cell development and peripheral B cell responses and examine the ways by which miRNAs regulate signal downstream of B cell antigen receptor to prevent aberrant activation and autoimmunity.
Collapse
Affiliation(s)
- Timothy C. Borbet
- New York University School of Medicine, Department of Pathology, New York, NY 10016
| | - Marcus J. Hines
- New York University School of Medicine, Department of Pathology, New York, NY 10016
| | - Sergei B. Koralov
- New York University School of Medicine, Department of Pathology, New York, NY 10016
| |
Collapse
|
44
|
Cai W, Li C, Li J, Song J, Zhang S. Integrated Small RNA Sequencing, Transcriptome and GWAS Data Reveal microRNA Regulation in Response to Milk Protein Traits in Chinese Holstein Cattle. Front Genet 2021; 12:726706. [PMID: 34712266 PMCID: PMC8546187 DOI: 10.3389/fgene.2021.726706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/21/2021] [Indexed: 01/04/2023] Open
Abstract
Milk protein is one of the most important economic traits in the dairy industry. Yet, the regulatory network of miRNAs for the synthesis of milk protein in mammary is poorly understood. Samples from 12 Chinese Holstein cows with three high ( ≥ 3.5%) and three low ( ≤ 3.0%) phenotypic values for milk protein percentage in lactation and non-lactation were examined through deep small RNA sequencing. We characterized 388 known and 212 novel miRNAs in the mammary gland. Differentially expressed analysis detected 28 miRNAs in lactation and 52 miRNAs in the non-lactating period with a highly significant correlation with milk protein concentration. Target prediction and correlation analysis identified some key miRNAs and their targets potentially involved in the synthesis of milk protein. We analyzed for enrichments of GWAS signals in miRNAs and their correlated targets. Our results demonstrated that genomic regions harboring DE miRNA genes in lactation were significantly enriched with GWAS signals for milk protein percentage traits and that enrichments within DE miRNA targets were significantly higher than in random gene sets for the majority of milk production traits. This integrated study on the transcriptome and posttranscriptional regulatory profiles between significantly differential phenotypes of milk protein concentration provides new insights into the mechanism of milk protein synthesis, which should reveal the regulatory mechanisms of milk secretion.
Collapse
Affiliation(s)
- Wentao Cai
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Department of Animal and Avian Science, University of Maryland, College Park, MD, United States
| | - Cong Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junya Li
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiuzhou Song
- Department of Animal and Avian Science, University of Maryland, College Park, MD, United States
| | - Shengli Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
45
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Ghani E, Barazesh A, Karimazar M, Nguewa P, Carrera Silva EA. Highlighting the interplay of microRNAs from Leishmania parasites and infected-host cells. Parasitology 2021; 148:1434-1446. [PMID: 34218829 PMCID: PMC11010138 DOI: 10.1017/s0031182021001177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/10/2021] [Accepted: 06/27/2021] [Indexed: 02/05/2023]
Abstract
Leishmania parasites, the causative agents of leishmaniasis, are protozoan parasites with the ability to modify the signalling pathway and cell responses of their infected host cells. These parasite strategies alter the host cell environment and conditions favouring their replication, survival and pathogenesis. Since microRNAs (miRNAs) are able to post-transcriptionally regulate gene expression processes, these biomolecules can exert critical roles in controlling Leishmania-host cell interplay. Therefore, the identification of relevant miRNAs differentially expressed in Leishmania parasites as well as in infected cells, which affect the host fitness, could be critical to understand the infection biology, pathogenicity and immune response against these parasites. Accordingly, the current review aims to address the differentially expressed miRNAs in both, the parasite and infected host cells and how these biomolecules change cell signalling and host immune responses during infection. A deep understanding of these processes could provide novel guidelines and therapeutic strategies for managing and treating leishmaniasis.
Collapse
Affiliation(s)
- Sajad Rashidi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Esmaeel Ghani
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Barazesh
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Karimazar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paul Nguewa
- University of Navarra, ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), c/Irunlarrea 1, 31008Pamplona, Spain
| | | |
Collapse
|
46
|
Kersy O, Salmon-Divon M, Shpilberg O, Hershkovitz-Rokah O. Non-Coding RNAs in Normal B-Cell Development and in Mantle Cell Lymphoma: From Molecular Mechanism to Biomarker and Therapeutic Agent Potential. Int J Mol Sci 2021; 22:ijms22179490. [PMID: 34502399 PMCID: PMC8430640 DOI: 10.3390/ijms22179490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/23/2021] [Accepted: 08/29/2021] [Indexed: 12/27/2022] Open
Abstract
B-lymphocytes are essential for an efficient immune response against a variety of pathogens. A large fraction of hematologic malignancies are of B-cell origin, suggesting that the development and activation of B cells must be tightly regulated. In recent years, differentially expressed non-coding RNAs have been identified in mantle cell lymphoma (MCL) tumor samples as opposed to their naive, normal B-cell compartment. These aberrantly expressed molecules, specifically microRNAs (miRNAs), circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs), have a role in cellular growth and survival pathways in various biological models. Here, we provide an overview of current knowledge on the role of non-coding RNAs and their relevant targets in B-cell development, activation and malignant transformation, summarizing the current understanding of the role of aberrant expression of non-coding RNAs in MCL pathobiology with perspectives for clinical use.
Collapse
Affiliation(s)
- Olga Kersy
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel; (O.K.); (M.S.-D.)
- Translational Research Lab, Assuta Medical Centers, Tel-Aviv 6971028, Israel;
| | - Mali Salmon-Divon
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel; (O.K.); (M.S.-D.)
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Ofer Shpilberg
- Translational Research Lab, Assuta Medical Centers, Tel-Aviv 6971028, Israel;
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel
- Institute of Hematology, Assuta Medical Centers, Tel-Aviv 6971028, Israel
| | - Oshrat Hershkovitz-Rokah
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel; (O.K.); (M.S.-D.)
- Translational Research Lab, Assuta Medical Centers, Tel-Aviv 6971028, Israel;
- Correspondence: ; Tel.: +972-3-764-4094
| |
Collapse
|
47
|
Bhatnagar B, Garzon R. Clinical Applications of MicroRNAs in Acute Myeloid Leukemia: A Mini-Review. Front Oncol 2021; 11:679022. [PMID: 34458136 PMCID: PMC8385666 DOI: 10.3389/fonc.2021.679022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/13/2021] [Indexed: 01/19/2023] Open
Abstract
MicroRNAs (miRs) are short non-coding RNAs, typically 18-25 nucleotides in length, that are critically important, through their direct effects on target mRNAs, in a variety of cellular processes including cell differentiation, proliferation and survival. Dysregulated miR expression has been identified in numerous cancer types including acute myeloid leukemia (AML). From a clinical standpoint, several miRs have been shown to associate with prognosis in AML patients. Furthermore, they also carry the potential to be used as biomarkers and to inform medical decision making. In addition, several preclinical studies have provided strong rationale to develop novel therapeutic strategies to target miRs in AML. This review will focus on potential clinical applications of miRs in adult AML and will discuss unique miR signatures in specific AML subtypes, their role in prognostication and response to therapy, as well as miRs that are promising therapeutic targets and ongoing clinical trials directed towards targeting clinically relevant miRs in AML that could allow for improvements in current treatment strategies.
Collapse
Affiliation(s)
- Bhavana Bhatnagar
- Division of Hematology and Medical Oncology, West Virginia University Cancer Institute, Schiffler Cancer Center, Wheeling, WV, United States
| | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.,The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
48
|
The Role of microRNAs in Pulp Inflammation. Cells 2021; 10:cells10082142. [PMID: 34440911 PMCID: PMC8391605 DOI: 10.3390/cells10082142] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
The dental pulp can be affected by thermal, physical, chemical, and bacterial phenomena that stimulate the inflammatory response. The pulp tissue produces an immunological, cellular, and vascular reaction in an attempt to defend itself and resolve the affected tissue. The expression of different microRNAs during pulp inflammation has been previously documented. MicroRNAs (miRNAs) are endogenous small molecules involved in the transcription of genes that regulate the immune system and the inflammatory response. They are present in cellular and physiological functions, as well as in the pathogenesis of human diseases, becoming potential biomarkers for diagnosis, prognosis, monitoring, and safety. Previous studies have evidenced the different roles played by miRNAs in proinflammatory, anti-inflammatory, and immunological phenomena in the dental pulp, highlighting specific key functions of pulp pathology. This systematized review aims to provide an understanding of the role of the different microRNAs detected in the pulp and their effects on the expression of the different target genes that are involved during pulp inflammation.
Collapse
|
49
|
The Role of microRNAs in NK Cell Development and Function. Cells 2021; 10:cells10082020. [PMID: 34440789 PMCID: PMC8391642 DOI: 10.3390/cells10082020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
The clinical use of natural killer (NK) cells is at the forefront of cellular therapy. NK cells possess exceptional antitumor cytotoxic potentials and can generate significant levels of proinflammatory cytokines. Multiple genetic manipulations are being tested to augment the anti-tumor functions of NK cells. One such method involves identifying and altering microRNAs (miRNAs) that play essential roles in the development and effector functions of NK cells. Unique miRNAs can bind and inactivate mRNAs that code for cytotoxic proteins. MicroRNAs, such as the members of the Mirc11 cistron, downmodulate ubiquitin ligases that are central to the activation of the obligatory transcription factors responsible for the production of inflammatory cytokines. These studies reveal potential opportunities to post-translationally enhance the effector functions of human NK cells while reducing unwanted outcomes. Here, we summarize the recent advances made on miRNAs in murine and human NK cells and their relevance to NK cell development and functions.
Collapse
|
50
|
Mi QS, Wang J, Liu Q, Wu X, Zhou L. microRNA dynamic expression regulates invariant NKT cells. Cell Mol Life Sci 2021; 78:6003-6015. [PMID: 34236444 PMCID: PMC11073247 DOI: 10.1007/s00018-021-03895-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Invariant natural killer T cells (iNKT) are a prevalent population of innate-like T cells in mice, but quite rare in humans that are critical for regulation of the innate and adaptive immune responses during antimicrobial immunity, tumor rejection, and inflammatory diseases. Multiple transcription factors and signaling molecules that contribute to iNKT cell selection and functional differentiation have been identified. However, the full molecular network responsible for regulating and maintaining iNKT populations remains unclear. MicroRNAs (miRNAs) are an abundant class of evolutionarily conserved, small, non-coding RNAs that regulate gene expression post-transcriptionally. Previous reports uncovered the important roles of miRNAs in iNKT cell development and function using Dicer mutant mice. In this review, we discuss the emerging roles of individual miRNAs in iNKT cells reported by our group and other groups, including miR-150, miR-155, miR-181, let-7, miR-17 ~ 92 cluster, and miR-183-96-182 cluster. It is likely that iNKT cell development, differentiation, homeostasis, and functions are orchestrated through a multilayered network comprising interactions among master transcription factors, signaling molecules, and dynamically expressed miRNAs. We provide a comprehensive view of the molecular mechanisms underlying iNKT cell differentiation and function controlled by dynamically expressed miRNAs.
Collapse
Affiliation(s)
- Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, USA.
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA.
- Department of Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.
| | - Jie Wang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA
| | - Queping Liu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA
| | - Xiaojun Wu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, 1 Ford Place, Detroit, MI, USA.
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA.
- Department of Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.
| |
Collapse
|