1
|
Barbagallo D, Ponti D, Bassani B, Bruno A, Pulze L, Akkihal SA, George-William JN, Gundamaraju R, Campomenosi P. MiR-223-3p in Cancer Development and Cancer Drug Resistance: Same Coin, Different Faces. Int J Mol Sci 2024; 25:8191. [PMID: 39125761 PMCID: PMC11311375 DOI: 10.3390/ijms25158191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
MicroRNAs (miRNAs) are mighty post-transcriptional regulators in cell physiology and pathophysiology. In this review, we focus on the role of miR-223-3p (henceforth miR-223) in various cancer types. MiR-223 has established roles in hematopoiesis, inflammation, and most cancers, where it can act as either an oncogenic or oncosuppressive miRNA, depending on specific molecular landscapes. MiR-223 has also been linked to either the sensitivity or resistance of cancer cells to treatments in a context-dependent way. Through this detailed review, we highlight that for some cancers (i.e., breast, non-small cell lung carcinoma, and glioblastoma), the oncosuppressive role of miR-223 is consistently reported in the literature, while for others (i.e., colorectal, ovarian, and pancreatic cancers, and acute lymphocytic leukemia), an oncogenic role prevails. In prostate cancer and other hematological malignancies, although an oncosuppressive role is frequently described, there is less of a consensus. Intriguingly, NLRP3 and FBXW7 are consistently identified as miR-223 targets when the miRNA acts as an oncosuppressor or an oncogene, respectively, in different cancers. Our review also describes that miR-223 was increased in biological fluids or their extracellular vesicles in most of the cancers analyzed, as compared to healthy or lower-risk conditions, confirming the potential application of this miRNA as a diagnostic and prognostic biomarker in the clinic.
Collapse
Affiliation(s)
- Davide Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics “Giovanni Sichel”, University of Catania, Via Santa Sofia 89, 95123 Catania, Italy
- Interdisciplinary Research Centre on the Diagnosis and Therapy of Brain Tumors, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Donatella Ponti
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Via Fantoli 16/15, 20138 Milano, Italy; (B.B.); (A.B.)
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Via Fantoli 16/15, 20138 Milano, Italy; (B.B.); (A.B.)
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| | - Laura Pulze
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| | - Shreya A. Akkihal
- Independent Researcher, 35004 SE Swenson St, Snoqualmie, WA 98065, USA;
| | - Jonahunnatha N. George-William
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi, 93, 20054 Segrate, Italy;
| | - Rohit Gundamaraju
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA;
- ER Stress and Mucosal Immunology Team, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia
| | - Paola Campomenosi
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| |
Collapse
|
2
|
Ghosh S, Thamotharan S, Fong J, Lei MYY, Janzen C, Devaskar SU. Circulating extracellular vesicular microRNA signatures in early gestation show an association with subsequent clinical features of pre-eclampsia. Sci Rep 2024; 14:16770. [PMID: 39039088 PMCID: PMC11263608 DOI: 10.1038/s41598-024-64057-w] [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: 01/12/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024] Open
Abstract
In a prospective cohort of subjects who subsequently developed preeclampsia (PE, n = 14) versus remaining healthy (NORM, n = 12), early gestation circulating extracellular vesicles (EVs) containing a panel of microRNA signatures were characterized and their biological networks of targets deciphered. Multiple microRNAs of which some arose from the placenta (19MC and 14MC) demonstrated changes in association with advancing gestation, while others expressed were pathognomonic of the subsequent development of characteristic clinical features of PE which set in as a late-onset subtype. This panel of miRNAs demonstrated a predictability with an area under the curve of 0.96 using leave-one-out cross-validation training in a logistic regression model with elastic-net regularization and precautions against overfitting. In addition, this panel of miRNAs, some of which were previously detected in either placental tissue or as maternal cell-free non-coding transcripts, lent further validation to our EV studies and the observed association with PE. Further, the identified biological networks of targets of these detected miRNAs revealed biological functions related to vascular remodeling, cellular proliferation, growth, VEGF, EGF and the PIP3/Akt signaling pathways, all mediating key cellular functions. We conclude that we have demonstrated a proof-of-principle by detecting a panel of EV packaged miRNAs in the maternal circulation early in gestation with possibilities of biological function in the placenta and other maternal tissues, along with the probability of predicting the subsequent clinical appearance of PE, particularly the late-onset subtype.
Collapse
Affiliation(s)
- Shubhamoy Ghosh
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833, Le Conte Avenue, MDCC-22-412, Los Angeles, CA, 90095, USA
| | - Shanthie Thamotharan
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833, Le Conte Avenue, MDCC-22-412, Los Angeles, CA, 90095, USA
| | - Jeanette Fong
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833, Le Conte Avenue, MDCC-22-412, Los Angeles, CA, 90095, USA
| | - Margarida Y Y Lei
- Department of Obstetrics & Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Carla Janzen
- Department of Obstetrics & Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Sherin U Devaskar
- Department of Pediatrics, David Geffen School of Medicine, University of California, 10833, Le Conte Avenue, MDCC-22-412, Los Angeles, CA, 90095, USA.
| |
Collapse
|
3
|
Aboismaiel MG, Amin MN, Eissa LA. Renoprotective effect of a novel combination of 6-gingerol and metformin in high-fat diet/streptozotocin-induced diabetic nephropathy in rats via targeting miRNA-146a, miRNA-223, TLR4/TRAF6/NLRP3 inflammasome pathway and HIF-1α. Biol Res 2024; 57:47. [PMID: 39033184 PMCID: PMC11265012 DOI: 10.1186/s40659-024-00527-9] [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: 10/14/2023] [Accepted: 06/29/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND MiRNA-146a and miRNA-223 are key epigenetic regulators of toll-like receptor 4 (TLR4)/tumor necrosis factor-receptor-associated factor 6 (TRAF6)/NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome pathway, which is involved in diabetic nephropathy (DN) pathogenesis. The currently available oral anti-diabetic treatments have been insufficient to halt DN development and progression. Therefore, this work aimed to assess the renoprotective effect of the natural compound 6-gingerol (GR) either alone or in combination with metformin (MET) in high-fat diet/streptozotocin-induced DN in rats. The proposed molecular mechanisms were also investigated. METHODS Oral gavage of 6-gingerol (100 mg/kg) and metformin (300 mg/kg) were administered to rats daily for eight weeks. MiRNA-146a, miRNA-223, TLR4, TRAF6, nuclear factor-kappa B (NF-κB) (p65), NLRP3, caspase-1, and hypoxia-inducible factor-1 alpha (HIF-1α) mRNA expressions were measured using real-time PCR. ELISA was used to measure TLR4, TRAF6, NLRP3, caspase-1, tumor necrosis factor-alpha (TNF-α), and interleukin-1-beta (IL-1β) renal tissue levels. Renal tissue histopathology and immunohistochemical examination of fibronectin and NF-κB (p65) were performed. RESULTS 6-Gingerol treatment significantly reduced kidney tissue damage and fibrosis. 6-Gingerol up-regulated miRNA-146a and miRNA-223 and reduced TLR4, TRAF6, NF-κB (p65), NLRP3, caspase-1, TNF-α, IL-1β, HIF-1α and fibronectin renal expressions. 6-Gingerol improved lipid profile and renal functions, attenuated renal hypertrophy, increased reduced glutathione, and decreased blood glucose and malondialdehyde levels. 6-Gingerol and metformin combination showed superior renoprotective effects than either alone. CONCLUSION 6-Gingerol demonstrated a key protective role in DN by induction of miRNA-146a and miRNA-223 expression and inhibition of TLR4/TRAF6/NLRP3 inflammasome signaling. 6-Gingerol, a safe, affordable, and abundant natural compound, holds promise for use as an adjuvant therapy with metformin in diabetic patients to attenuate renal damage and stop the progression of DN.
Collapse
Affiliation(s)
- Merna G Aboismaiel
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mohamed N Amin
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Laila A Eissa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
4
|
Selvakumar B, Sekar P, Samsudin AR. Intestinal macrophages in pathogenesis and treatment of gut leakage: current strategies and future perspectives. J Leukoc Biol 2024; 115:607-619. [PMID: 38198217 DOI: 10.1093/jleuko/qiad165] [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: 07/02/2023] [Revised: 11/13/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Macrophages play key roles in tissue homeostasis, defense, disease, and repair. Macrophages are highly plastic and exhibit distinct functional phenotypes based on micro-environmental stimuli. In spite of several advancements in understanding macrophage biology and their different functional phenotypes in various physiological and pathological conditions, currently available treatment strategies targeting macrophages are limited. Macrophages' high plasticity and diverse functional roles-including tissue injury and wound healing mechanisms-mark them as potential targets to mine for efficient therapeutics to treat diseases. Despite mounting evidence on association of gut leakage with several extraintestinal diseases, there is no targeted standard therapy to treat gut leakage. Therefore, there is an urgent need to develop therapeutic strategies to treat this condition. Macrophages are the cells that play the largest role in interacting with the gut microbiota in the intestinal compartment and exert their intended functions in injury and repair mechanisms. In this review, we have summarized the current knowledge on the origins and phenotypes of macrophages. The specific role of macrophages in intestinal barrier function, their role in tissue repair mechanisms, and their association with gut microbiota are discussed. In addition, currently available therapies and the putative tissue repair mediators of macrophages for treating microbiota dysbiosis induced gut leakage are also discussed. The overall aim of this review is to convey the intense need to screen for microbiota induced macrophage-released prorepair mediators, which could lead to the identification of potential candidates that could be developed for treating the leaky gut and associated diseases.
Collapse
Affiliation(s)
- Balachandar Selvakumar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Priyadharshini Sekar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - A Rani Samsudin
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
5
|
Rodriguez NM, Loren P, Paez I, Martínez C, Chaparro A, Salazar LA. MicroRNAs: The Missing Link between Hypertension and Periodontitis? Int J Mol Sci 2024; 25:1992. [PMID: 38396672 PMCID: PMC10889313 DOI: 10.3390/ijms25041992] [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: 01/01/2024] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, and arterial hypertension is a recognized cardiovascular risk factor that is responsible for high morbidity and mortality. Arterial hypertension is the result of an inflammatory process that results in the remodeling and thickening of the vascular walls, which is associated with an immunological response. Previous studies have attempted to demonstrate the relationship between oral disease, inflammation, and the development of systemic diseases. Currently, the existence of an association between periodontitis and hypertension is a controversial issue because the underlying pathophysiological processes and inflammatory mechanisms common to both diseases are unknown. This is due to the fact that periodontitis is a chronic inflammatory disease that affects the interface of teeth and surrounding tissues. However, the most likely explanation for understanding this association is related to low-grade chronic inflammation. An initial path in the study of the relationship between the mentioned pathologies is the possibility of an epigenetic influence, mediated by noncoding RNAs as microRNAs. Thus, in the present review we describe the role of microRNAs related to arterial hypertension and/or periodontitis. In addition, we identified 13 common microRNAs between periodontitis and hypertension. According to the predictions of the DIANA-mirPath program, they can regulate genes involved in 52 signaling pathways.
Collapse
Affiliation(s)
- Nelia M Rodriguez
- Doctoral Program in Sciences, Major in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
- Center for Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Universidad de La Frontera, Temuco 4811230, Chile
| | - Pía Loren
- Center for Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Universidad de La Frontera, Temuco 4811230, Chile
| | - Isis Paez
- Doctoral Program in Sciences, Major in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
- Center for Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Universidad de La Frontera, Temuco 4811230, Chile
| | - Constanza Martínez
- Department of Oral Pathology and Conservative Dentistry, Periodontics, Faculty of Dentistry, Universidad de Los Andes, Santiago 7620001, Chile
| | - Alejandra Chaparro
- Department of Oral Pathology and Conservative Dentistry, Periodontics, Faculty of Dentistry, Universidad de Los Andes, Santiago 7620001, Chile
- Center for Biomedical Research and Innovation (CIIB), Universidad de Los Andes, Santiago 7620001, Chile
| | - Luis A Salazar
- Center for Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Universidad de La Frontera, Temuco 4811230, Chile
| |
Collapse
|
6
|
Wang J, Tian F, Cao L, Du R, Tong J, Ding X, Yuan Y, Wang C. Macrophage polarization in spinal cord injury repair and the possible role of microRNAs: A review. Heliyon 2023; 9:e22914. [PMID: 38125535 PMCID: PMC10731087 DOI: 10.1016/j.heliyon.2023.e22914] [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: 03/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The prevention, treatment, and rehabilitation of spinal cord injury (SCI) have always posed significant medical challenges. After mechanical injury, disturbances in microcirculation, edema formation, and the generation of free radicals lead to additional damage, impeding effective repair processes and potentially exacerbating further dysfunction. In this context, inflammatory responses, especially the activation of macrophages, play a pivotal role. Different phenotypes of macrophages have distinct effects on inflammation. Activation of classical macrophage cells (M1) promotes inflammation, while activation of alternative macrophage cells (M2) inhibits inflammation. The polarization of macrophages is crucial for disease healing. A non-coding RNA, known as microRNA (miRNA), governs the polarization of macrophages, thereby reducing inflammation following SCI and facilitating functional recovery. This study elucidates the inflammatory response to SCI, focusing on the infiltration of immune cells, specifically macrophages. It examines their phenotype and provides an explanation of their polarization mechanisms. Finally, this paper introduces several well-known miRNAs that contribute to macrophage polarization following SCI, including miR-155, miR-130a, and miR-27 for M1 polarization, as well as miR-22, miR-146a, miR-21, miR-124, miR-223, miR-93, miR-132, and miR-34a for M2 polarization. The emphasis is placed on their potential therapeutic role in SCI by modulating macrophage polarization, as well as the present developments and obstacles of miRNA clinical therapy.
Collapse
Affiliation(s)
- Jiawei Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Feng Tian
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Lili Cao
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Ruochen Du
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Jiahui Tong
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Xueting Ding
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Yitong Yuan
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Chunfang Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| |
Collapse
|
7
|
Mancuso R, Citterio LA, Agostini S, Marventano I, La Rosa F, Re F, Seneci P, Saresella M, Clerici M. Glibenclamide-Loaded Nanoparticles Reduce NLRP3 Inflammasome Activation and Modulate miR-223-3p/miR-7-1-5p Expression in THP-1 Cells. Pharmaceuticals (Basel) 2023; 16:1590. [PMID: 38004455 PMCID: PMC10675475 DOI: 10.3390/ph16111590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
The anti-hyperglycemic drug glibenclamide (Glb) might represent an interesting therapeutic option in human neurodegenerative diseases because of its anti-inflammatory activity and its ability to downregulate activation of the NLRP3 inflammasome. Bi-functionalized liposomes that can cross the blood-brain barrier (BBB) may be used to release Glb into the central nervous system (CNS), overcoming its poor solubility and bioavailability. Here, we analyzed in vitro the effect of Glb-loaded nanovectors (GNVs) and Glb itself on NLRP3 inflammasome activation using a lipopolysaccharide- and nigericine-activated THP-1 cell model. Apoptosis-associated speck-like protein containing a CARD (ASC) aggregation and NLRP3-related cytokine (IL-1β, caspase 1, and IL-18) production and gene expression, as well as the concentration of miR-223-3p and miR-7-1-5p, known to modulate the NLRP3 inflammasome, were evaluated in all conditions. Results showed that both GNVs and Glb reduced significantly ASC-speck oligomerization, transcription and translation of NLRP3, as well as the secretion of caspase 1 and IL-1β (p < 0.05 for all). Unexpectedly, GNVs/Glb significantly suppressed miR-223-3p and upregulated miR-7-1-5p expression (p < 0.01). These preliminary results thus suggest that GNVs, similarly to Glb, are able to dampen NLRP3 inflammasome activation, inflammatory cytokine release, and modulate miR-223-3p/miR-7-1-5p. Although the mechanisms underlying the complex relation among these elements remain to be further investigated, these results can open new roads to the use of GNVs as a novel strategy to reduce inflammasome activation in disease and rehabilitation.
Collapse
Affiliation(s)
- Roberta Mancuso
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
| | - Lorenzo Agostino Citterio
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
| | - Simone Agostini
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
| | - Ivana Marventano
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
| | - Francesca La Rosa
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
| | - Francesca Re
- School of Medicine and Surgery, University of Milano-Bicocca, 20854 Milan, Italy;
| | | | - Marina Saresella
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
| | - Mario Clerici
- IRCCS Fondazione Don Gnocchi—ONLUS, 20148 Milan, Italy; (R.M.); (L.A.C.); (I.M.); (F.L.R.); (M.S.); (M.C.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| |
Collapse
|
8
|
Ortiz GGR, Zaidi NH, Saini RS, Ramirez Coronel AA, Alsandook T, Hadi Lafta M, Arias-Gonzáles JL, Amin AH, Maaliw Iii RR. The developing role of extracellular vesicles in autoimmune diseases: special attention to mesenchymal stem cell-derived extracellular vesicles. Int Immunopharmacol 2023; 122:110531. [PMID: 37437434 DOI: 10.1016/j.intimp.2023.110531] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/10/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
Autoimmune diseases are complex, chronic inflammatory conditions initiated by the loss of immunological tolerance to self-antigens. Nowadays, there is no effective and useful therapy for autoimmune diseases, and the existing medications have some limitations due to their nonspecific targets and side effects. During the last few decades, it has been established that mesenchymal stem cells (MSCs) have immunomodulatory functions. It is proposed that MSCs can exert an important therapeutic effect on autoimmune disorders. In parallel with these findings, several investigations have shown that MSCs alleviate autoimmune diseases. Intriguingly, the results of studies have demonstrated that the effective roles of MSCs in autoimmune diseases do not depend on direct intercellular communication but on their ability to release a wide spectrum of paracrine mediators such as growth factors, cytokines and extracellular vehicles (EVs). EVs that range from 50 to 5,000 nm were produced by almost any cell type, and these nanoparticles participate in homeostasis and intercellular communication via the transfer of a broad range of biomolecules such as modulatory proteins, nucleic acids (DNA and RNA), lipids, cytokines, and metabolites. EVs derived from MSCs display the exact properties of MSCs and can be safer and more beneficial than their parent cells. In this review, we will discuss the features of MSCs and their EVs, EVs biogenesis, and their cargos, and then we will highlight the existing discoveries on the impacts of EVs from MSCs on autoimmune diseases such as multiple sclerosis, arthritis rheumatic, inflammatory bowel disease, Type 1 diabetes mellitus, systemic lupus erythematosus, autoimmune liver diseases, Sjögren syndrome, and osteoarthritis, suggesting a potential alternative for autoimmune conditions therapy.
Collapse
Affiliation(s)
- Geovanny Genaro Reivan Ortiz
- Laboratory of Basic Psychology, Behavioral Analysis and Programmatic Development (PAD-LAB), Catholic University of Cuenca, Cuenca, Ecuador
| | - Neelam Hazoor Zaidi
- Umanand Prasad School of Medicine and Health Science, The University of Fiji, Saweni Campus, Lautoka, Fiji
| | | | | | - Tahani Alsandook
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | | | | | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Renato R Maaliw Iii
- College of Engineering, Southern Luzon State University, Lucban, Quezon, Philippines.
| |
Collapse
|
9
|
Chen J, Ma S, Luo B, Hao H, Li Y, Yang H, Zhu F, Zhang P, Niu R, Pan P. Human umbilical cord mesenchymal stromal cell small extracellular vesicle transfer of microRNA-223-3p to lung epithelial cells attenuates inflammation in acute lung injury in mice. J Nanobiotechnology 2023; 21:295. [PMID: 37626408 PMCID: PMC10464265 DOI: 10.1186/s12951-023-02038-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI), manifested as strong pulmonary inflammation and alveolar epithelial damage, is a life-threatening disease with high morbidity and mortality. Small extracellular vesicles (sEVs), secreted by multiple types of cells, are critical cellular communication mediators and can inhibit inflammation by transferring bioactive molecules, such as microRNAs (miRNAs). Thus, we hypothesized that sEVs derived from mesenchymal stromal cells (MSC sEVs) could transfer miRNAs to attenuate inflammation of lung epithelial cells during ALI. METHODS C57BL/6 male mice were intratracheally administered LPS (10 mg/kg). Six hours later, the mice were randomly administered with MSC sEVs (40 µg per mouse in 150 µl of saline), which were collected by ultracentrifugation. Control group received saline administration. After 48 h, the mice were sacrificed to evaluate pulmonary microvascular permeability and inflammatory responses. In vitro, A549 cells and primary human small airway epithelial cells (SAECs) were stimulated with LPS with or without MSC sEVs treatment. RESULTS In vitro, MSC sEVs could also inhibit the inflammation induced by LPS in A549 cells and SAECs (reducing TNF-α, IL-1β, IL-6 and MCP-1). Moreover, MSC sEV treatment improved the survival rate, alleviated pulmonary microvascular permeability, and inhibited proinflammatory responses (reducing TNF-α, IL-1β, IL-6 and JE-1) in ALI mice. Notably, miR-223-3p was found to be served as a critical mediator in MSC sEV-induced regulatory effects through inhibition of poly (adenosine diphosphate-ribose) polymerase-1 (PARP-1) in lung epithelial cells. CONCLUSIONS Overall, these findings suggest that MSC sEVs may offer a novel promising strategy for ALI.
Collapse
Affiliation(s)
- Jie Chen
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Shiyang Ma
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Baihua Luo
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Haojie Hao
- Institute of Basic Medicine Science, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Medical College, Beijing, China
| | - Yanqin Li
- Center of Pulmonary & Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical College, Beijing, China
| | - Hang Yang
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Fei Zhu
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Peipei Zhang
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Ruichao Niu
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China.
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China.
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Pinhua Pan
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China.
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China.
| |
Collapse
|
10
|
Wu M, Li G, Wang W, Ren H. Emerging roles of microRNAs in septic cardiomyopathy. Front Pharmacol 2023; 14:1181372. [PMID: 37475718 PMCID: PMC10354437 DOI: 10.3389/fphar.2023.1181372] [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: 03/07/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
As one of the serious complications of sepsis, septic cardiomyopathy has gained more and more attention, because of its high morbidity and mortality. With the in-depth study of septic cardiomyopathy, several methods have been adopted clinically but have poor therapeutic effects due to failure to find precise therapeutic targets. In recent years, microRNAs have been found to be related to the pathogenesis, diagnosis, and treatment of septic cardiomyopathy via regulating immunity and programmed cell death. This paper reviews the role of microRNAs in septic cardiomyopathy, aiming to provide new targets for the diagnosis and treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
| | | | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongsheng Ren
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
11
|
Shi M, Lu Q, Zhao Y, Ding Z, Yu S, Li J, Ji M, Fan H, Hou S. miR-223: a key regulator of pulmonary inflammation. Front Med (Lausanne) 2023; 10:1187557. [PMID: 37465640 PMCID: PMC10350674 DOI: 10.3389/fmed.2023.1187557] [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: 03/16/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Small noncoding RNAs, known as microRNAs (miRNAs), are vital for the regulation of diverse biological processes. miR-223, an evolutionarily conserved anti-inflammatory miRNA expressed in cells of the myeloid lineage, has been implicated in the regulation of monocyte-macrophage differentiation, proinflammatory responses, and the recruitment of neutrophils. The biological functions of this gene are regulated by its expression levels in cells or tissues. In this review, we first outline the regulatory role of miR-223 in granulocytes, macrophages, endothelial cells, epithelial cells and dendritic cells (DCs). Then, we summarize the possible role of miR-223 in chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), coronavirus disease 2019 (COVID-19) and other pulmonary inflammatory diseases to better understand the molecular regulatory networks in pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Mingyu Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Ziling Ding
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sifan Yu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Junfeng Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Mengjun Ji
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| |
Collapse
|
12
|
Bhati T, Ray A, Arora R, Siraj F, Parvez S, Rastogi S. Immunomodulation of cytokine signalling at feto-maternal interface by microRNA-223 and -150-5p in infection-associated spontaneous preterm birth. Mol Immunol 2023; 160:1-11. [PMID: 37285685 DOI: 10.1016/j.molimm.2023.05.009] [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: 02/10/2023] [Revised: 04/24/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Spontaneous preterm birth (sPTB) is a global health concern and it is the most prevalent cause of infant mortality and morbidity with occurrence rate of 5 - 18% worldwide. Studies suggest infection and infection-driven activation of inflammatory responses are the potential risk factors for sPTB. MicroRNAs (miRNAs) are thought to control the expression of several immune genes, making them crucial components of the intricate immune regulatory network and the dysregulation of miRNAs in placenta has been associated to several pregnancy-related complications. However, studies on possible role of miRNAs in immunomodulation of cytokine signalling in infection-associated sPTB are scarce. Present study aimed to investigate expression/ correlation of a few circulating miRNAs (miR-223, -150-5p, -185-5p, -191-5p), miRNA target genes and associated cytokines in sPTB women found infected with Chlamydia trachomatis/ Mycoplasma hominis/ Ureaplasma urealyticum. Non-heparinized blood and placental sample were collected from 140 sPTB and 140 term women visiting Safdarjung hospital, New Delhi (India) for conducting PCR and RT-PCR for pathogen detection and miRNA/ target gene/ cytokine expression, respectively. Common target genes of differentially expressed miRNAs were obtained from databases. The correlation between select target genes/ cytokines and serum miRNAs was determined by Spearman's rank correlation. 43 sPTB were infected with either pathogen and a significant upregulation of serum miRNAs was observed. However, miR-223 and 150-5p showed maximum fold-change (4.78 and 5.58, respectively) in PTB versus control group. IL-6ST, TGF-β R3 and MMP-14 were important target genes among 454 common targets, whereas, IL-6 and TGF-β were associated cytokines. miR-223 and 150-5p showed significant negative correlation with IL-6ST/ IL-6/ MMP-14 and positive correlation with TGF-β R3/ TGF-β. A significant positive correlation was found between IL-6ST and IL-6, TGF-β R3 and TGF-β. However, miR-185-5p and 191-5p were not significantly correlated. Although post-transcriptional validation is required, yet on the basis of mRNA findings, the study concludes that miR-223 and 150-5p are apparently of clinical importance in regulation of inflammatory processes during infection-associated sPTB.
Collapse
Affiliation(s)
- Tanu Bhati
- Molecular Microbiology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | - Ankita Ray
- Molecular Microbiology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | - Renu Arora
- Department of Obstetrics and Gynecology, Vardhman Mahavir Medical College (VMMC) and Safdarjung Hospital, New Delhi 110029, India
| | - Fouzia Siraj
- Pathology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | - Sangita Rastogi
- Molecular Microbiology laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box no. 4909, New Delhi 110029, India.
| |
Collapse
|
13
|
Li YX, Jiao P, Wang XP, Wang JP, Feng F, Bao BW, Dong YW, Luoreng ZM, Wei DW. RNA-seq reveals the role of miR-223 in alleviating inflammation of bovine mammary epithelial cells. Res Vet Sci 2023; 159:257-266. [PMID: 37192556 DOI: 10.1016/j.rvsc.2023.04.012] [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: 02/22/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/18/2023]
Abstract
Bovine mammary epithelial cells (bMECs) are involved in the early defense against the invasion of intramammary pathogens and are essential for the health of bovine mammary gland. MicroRNA (MiRNA) is a key factor that regulates cell state and physiological function. In the present study, the transcriptome profiles of miR-223 inhibitor transfection group (miR-223_Inhibitor) and negative control inhibitor transfection group (NC_Inhibitor) within bMECs were detected via the RNA sequencing (RNA-seq) platform. Based on these experiments, the differentially expressed mRNAs (DE-mRNAs) of the miR-223_Inhibitor transfection group were screened, and the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional analyses of DE-mRNAs were performed. The results revealed that compared with the NC_Inhibitor, 224 differentially expressed genes (DEGs) were identified in the miR-223_Inhibitor, including 184 upregulated and 40 downregulated genes. The functional annotation of the above DEGs indicated that some of these genes are involved in the immune response generated by extracellular substance stimulation, regulation of the activity of cytokines and chemokines, and the immune signaling pathways of NF-κB and TNF. Meanwhile, miR-223_inhibitor upregulated the immune key genes IRF1 and NFκBIA, cytokines IL-6 and IL-24, as well as chemokines CXCL3, CXCL5, and CCR6, triggering a signaling cascade response that exacerbated inflammation in bMECs. These results suggested that miR-223 plays an important role in inhibiting the inflammatory response and maintaining the stability of bMECs, and is a potential target for treating mastitis in dairy cows.
Collapse
Affiliation(s)
- Yan-Xia Li
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Peng Jiao
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Xing-Ping Wang
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Jin-Peng Wang
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Fen Feng
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Bin-Wu Bao
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yi-Wen Dong
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Zhuo-Ma Luoreng
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China.
| | - Da-Wei Wei
- School of Agriculture, Ningxia University, Yinchuan, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| |
Collapse
|
14
|
Singh SK, Dwivedi SD, Yadav K, Shah K, Chauhan NS, Pradhan M, Singh MR, Singh D. Novel Biotherapeutics Targeting Biomolecular and Cellular Approaches in Diabetic Wound Healing. Biomedicines 2023; 11:biomedicines11020613. [PMID: 36831151 PMCID: PMC9952895 DOI: 10.3390/biomedicines11020613] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Wound healing responses play a major role in chronic inflammation, which affects millions of people around the world. One of the daunting tasks of creating a wound-healing drug is finding equilibrium in the inflammatory cascade. In this study, the molecular and cellular mechanisms to regulate wound healing are explained, and recent research is addressed that demonstrates the molecular and cellular events during diabetic wound healing. Moreover, a range of factors or agents that facilitate wound healing have also been investigated as possible targets for successful treatment. It also summarises the various advances in research findings that have revealed promising molecular targets in the fields of therapy and diagnosis of cellular physiology and pathology of wound healing, such as neuropeptides, substance P, T cell immune response cDNA 7, miRNA, and treprostinil growth factors such as fibroblast growth factor, including thymosin beta 4, and immunomodulators as major therapeutic targets.
Collapse
Affiliation(s)
- Suraj Kumar Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
| | - Shradha Devi Dwivedi
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
| | - Krishna Yadav
- Raipur Institute of Pharmaceutical Educations and Research, Sarona, Raipur 492010, Chhattisgarh, India
| | - Kamal Shah
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, Uttar Pradesh, India
| | | | - Madhulika Pradhan
- Gracious College of Pharmacy Abhanpur Raipur, Village-Belbhata, Taluka, Abhanpur 493661, Chhattisgarh, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
- Correspondence:
| |
Collapse
|
15
|
Sprenkle NT, Serezani CH, Pua HH. MicroRNAs in Macrophages: Regulators of Activation and Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:359-368. [PMID: 36724439 PMCID: PMC10316964 DOI: 10.4049/jimmunol.2200467] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/13/2022] [Indexed: 02/03/2023]
Abstract
Macrophages are sentinels of the innate immune system that maintain tissue homeostasis and contribute to inflammatory responses. Their broad scope of action depends on both functional heterogeneity and plasticity. Small noncoding RNAs called microRNAs (miRNAs) contribute to macrophage function as post-transcriptional inhibitors of target gene networks. Genetic and pharmacologic studies have uncovered genes regulated by miRNAs that control macrophage cellular programming and macrophage-driven pathology. miRNAs control proinflammatory M1-like activation, immunoregulatory M2-like macrophage activation, and emerging macrophage functions in metabolic disease and innate immune memory. Understanding the gene networks regulated by individual miRNAs enhances our understanding of the spectrum of macrophage function at steady state and during responses to injury or pathogen invasion, with the potential to develop miRNA-based therapies. This review aims to consolidate past and current studies investigating the complexity of the miRNA interactome to provide the reader with a mechanistic view of how miRNAs shape macrophage behavior.
Collapse
Affiliation(s)
| | - C Henrique Serezani
- Department of Pathology, Microbiology, and Immunology
- Department of Medicine, Division of Infectious Diseases
- Vanderbilt Center for Immunobiology, Nashville, Tennessee 37232, USA
- Vandebilt Institute of Infection, Immunology and Inflammation; Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology
- Vanderbilt Center for Immunobiology, Nashville, Tennessee 37232, USA
- Vandebilt Institute of Infection, Immunology and Inflammation; Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
16
|
Peng X, Wang Q, Li W, Ge G, Peng J, Xu Y, Yang H, Bai J, Geng D. Comprehensive overview of microRNA function in rheumatoid arthritis. Bone Res 2023; 11:8. [PMID: 36690624 PMCID: PMC9870909 DOI: 10.1038/s41413-023-00244-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/15/2022] [Accepted: 12/04/2022] [Indexed: 01/25/2023] Open
Abstract
MicroRNAs (miRNAs), a class of endogenous single-stranded short noncoding RNAs, have emerged as vital epigenetic regulators of both pathological and physiological processes in animals. They direct fundamental cellular pathways and processes by fine-tuning the expression of multiple genes at the posttranscriptional level. Growing evidence suggests that miRNAs are implicated in the onset and development of rheumatoid arthritis (RA). RA is a chronic inflammatory disease that mainly affects synovial joints. This common autoimmune disorder is characterized by a complex and multifaceted pathogenesis, and its morbidity, disability and mortality rates remain consistently high. More in-depth insights into the underlying mechanisms of RA are required to address unmet clinical needs and optimize treatment. Herein, we comprehensively review the deregulated miRNAs and impaired cellular functions in RA to shed light on several aspects of RA pathogenesis, with a focus on excessive inflammation, synovial hyperplasia and progressive joint damage. This review also provides promising targets for innovative therapies of RA. In addition, we discuss the regulatory roles and clinical potential of extracellular miRNAs in RA, highlighting their prospective applications as diagnostic and predictive biomarkers.
Collapse
Affiliation(s)
- Xiaole Peng
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Qing Wang
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Wenming Li
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Gaoran Ge
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Jiachen Peng
- grid.413390.c0000 0004 1757 6938Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, 563000 Zunyi, P. R. China
| | - Yaozeng Xu
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Huilin Yang
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Jiaxiang Bai
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| | - Dechun Geng
- grid.429222.d0000 0004 1798 0228Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006 Jiangsu P. R. China
| |
Collapse
|
17
|
Millozzi F, Papait A, Bouché M, Parolini O, Palacios D. Nano-Immunomodulation: A New Strategy for Skeletal Muscle Diseases and Aging? Int J Mol Sci 2023; 24:1175. [PMID: 36674691 PMCID: PMC9862642 DOI: 10.3390/ijms24021175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
The skeletal muscle has a very remarkable ability to regenerate upon injury under physiological conditions; however, this regenerative capacity is strongly diminished in physio-pathological conditions, such as those present in diseased or aged muscles. Many muscular dystrophies (MDs) are characterized by aberrant inflammation due to the deregulation of both the lymphoid and myeloid cell populations and the production of pro-inflammatory cytokines. Pathological inflammation is also observed in old muscles due to a systemic change in the immune system, known as "inflammaging". Immunomodulation represents, therefore, a promising therapeutic opportunity for different skeletal muscle conditions. However, the use of immunomodulatory drugs in the clinics presents several caveats, including their low stability in vivo, the need for high doses to obtain therapeutically relevant effects, and the presence of strong side effects. Within this context, the emerging field of nanomedicine provides the powerful tools needed to control the immune response. Nano-scale materials are currently being explored as biocarriers to release immunomodulatory agents in the damaged tissues, allowing therapeutic doses with limited off-target effects. In addition, the intrinsic immunomodulatory properties of some nanomaterials offer further opportunities for intervention that still need to be systematically explored. Here we exhaustively review the state-of-the-art regarding the use of nano-sized materials to modulate the aberrant immune response that characterizes some physio-pathological muscle conditions, such as MDs or sarcopenia (the age-dependent loss of muscle mass). Based on our learnings from cancer and immune tolerance induction, we also discuss further opportunities, challenges, and limitations of the emerging field of nano-immunomodulation.
Collapse
Affiliation(s)
- Francesco Millozzi
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
- IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Andrea Papait
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Marina Bouché
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| |
Collapse
|
18
|
Fu SP, Chen SY, Pang QM, Zhang M, Wu XC, Wan X, Wan WH, Ao J, Zhang T. Advances in the research of the role of macrophage/microglia polarization-mediated inflammatory response in spinal cord injury. Front Immunol 2022; 13:1014013. [PMID: 36532022 PMCID: PMC9751019 DOI: 10.3389/fimmu.2022.1014013] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
It is often difficult to regain neurological function following spinal cord injury (SCI). Neuroinflammation is thought to be responsible for this failure. Regulating the inflammatory response post-SCI may contribute to the recovery of neurological function. Over the past few decades, studies have found that macrophages/microglia are one of the primary effector cells in the inflammatory response following SCI. Growing evidence has documented that macrophages/microglia are plastic cells that can polarize in response to microenvironmental signals into M1 and M2 macrophages/microglia. M1 produces pro-inflammatory cytokines to induce inflammation and worsen tissue damage, while M2 has anti-inflammatory activities in wound healing and tissue regeneration. Recent studies have indicated that the transition from the M1 to the M2 phenotype of macrophage/microglia supports the regression of inflammation and tissue repair. Here, we will review the role of the inflammatory response and macrophages/microglia in SCI and repair. In addition, we will discuss potential molecular mechanisms that induce macrophage/microglia polarization, with emphasis on neuroprotective therapies that modulate macrophage/microglia polarization, which will provide new insights into therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ming Pang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China,The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,*Correspondence: Tao Zhang,
| |
Collapse
|
19
|
Zhang S, Meng Y, Zhou L, Qiu L, Wang H, Su D, Zhang B, Chan K, Han J. Targeting epigenetic regulators for inflammation: Mechanisms and intervention therapy. MedComm (Beijing) 2022; 3:e173. [PMID: 36176733 PMCID: PMC9477794 DOI: 10.1002/mco2.173] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/28/2022] [Accepted: 08/05/2022] [Indexed: 11/11/2022] Open
Abstract
Emerging evidence indicates that resolution of inflammation is a critical and dynamic endogenous process for host tissues defending against external invasive pathogens or internal tissue injury. It has long been known that autoimmune diseases and chronic inflammatory disorders are characterized by dysregulated immune responses, leading to excessive and uncontrol tissue inflammation. The dysregulation of epigenetic alterations including DNA methylation, posttranslational modifications to histone proteins, and noncoding RNA expression has been implicated in a host of inflammatory disorders and the immune system. The inflammatory response is considered as a critical trigger of epigenetic alterations that in turn intercede inflammatory actions. Thus, understanding the molecular mechanism that dictates the outcome of targeting epigenetic regulators for inflammatory disease is required for inflammation resolution. In this article, we elucidate the critical role of the nuclear factor-κB signaling pathway, JAK/STAT signaling pathway, and the NLRP3 inflammasome in chronic inflammatory diseases. And we formulate the relationship between inflammation, coronavirus disease 2019, and human cancers. Additionally, we review the mechanism of epigenetic modifications involved in inflammation and innate immune cells. All that matters is that we propose and discuss the rejuvenation potential of interventions that target epigenetic regulators and regulatory mechanisms for chronic inflammation-associated diseases to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Su Zhang
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yang Meng
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Lian Zhou
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Lei Qiu
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Heping Wang
- Department of NeurosurgeryTongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan Su
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Bo Zhang
- Laboratory of Cancer Epigenetics and GenomicsDepartment of Gastrointestinal SurgeryFrontiers Science Center for Disease‐Related Molecular NetworkWest China HospitalSichuan UniversityChengduChina
| | - Kui‐Ming Chan
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
| | - Junhong Han
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
20
|
López‐Cuevas P, Xu C, Severn CE, Oates TCL, Cross SJ, Toye AM, Mann S, Martin P. Macrophage Reprogramming with Anti-miR223-Loaded Artificial Protocells Enhances In Vivo Cancer Therapeutic Potential. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202717. [PMID: 36314048 PMCID: PMC9762313 DOI: 10.1002/advs.202202717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Several immune cell-expressed miRNAs (miRs) are associated with altered prognostic outcome in cancer patients, suggesting that they may be potential targets for development of cancer therapies. Here, translucent zebrafish (Danio rerio) is utilized to demonstrate that genetic knockout or knockdown of one such miR, microRNA-223 (miR223), globally or specifically in leukocytes, does indeed lead to reduced cancer progression. As a first step toward potential translation to a clinical therapy, a novel strategy is described for reprogramming neutrophils and macrophages utilizing miniature artificial protocells (PCs) to deliver anti-miRs against the anti-inflammatory miR223. Using genetic and live imaging approaches, it is shown that phagocytic uptake of anti-miR223-loaded PCs by leukocytes in zebrafish (and by human macrophages in vitro) effectively prolongs their pro-inflammatory state by blocking the suppression of pro-inflammatory cytokines, which, in turn, drives altered immune cell-cancer cell interactions and ultimately leads to a reduced cancer burden by driving reduced proliferation and increased cell death of tumor cells. This PC cargo delivery strategy for reprogramming leukocytes toward beneficial phenotypes has implications also for treating other systemic or local immune-mediated pathologies.
Collapse
Affiliation(s)
- Paco López‐Cuevas
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
| | - Can Xu
- Centre for Protolife ResearchSchool of ChemistryUniversity of BristolBristolBS8 1TSUK
| | - Charlotte E. Severn
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolBS34 7QHUK
| | - Tiah C. L. Oates
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolBS34 7QHUK
| | - Stephen J. Cross
- Wolfson Bioimaging FacilityBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
| | - Ashley M. Toye
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolBS34 7QHUK
| | - Stephen Mann
- Centre for Protolife ResearchSchool of ChemistryUniversity of BristolBristolBS8 1TSUK
- Max Planck Bristol Centre for Minimal BiologySchool of ChemistryUniversity of BristolBristolBS8 1TSUK
- School of Materials Science and EngineeringShanghai Jiao Tong UniversityShanghai200240P. R. China
| | - Paul Martin
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
| |
Collapse
|
21
|
Sharma S, Tyagi T, Antoniak S. Platelet in thrombo-inflammation: Unraveling new therapeutic targets. Front Immunol 2022; 13:1039843. [PMID: 36451834 PMCID: PMC9702553 DOI: 10.3389/fimmu.2022.1039843] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
In the broad range of human diseases, thrombo-inflammation appears as a clinical manifestation. Clinically, it is well characterized in context of superficial thrombophlebitis that is recognized as thrombosis and inflammation of superficial veins. However, it is more hazardous when developed in the microvasculature of injured/inflamed/infected tissues and organs. Several diseases like sepsis and ischemia-reperfusion can cause formation of microvascular thrombosis subsequently leading to thrombo-inflammation. Thrombo-inflammation can also occur in cases of antiphospholipid syndrome, preeclampsia, sickle cell disease, bacterial and viral infection. One of the major contributors to thrombo-inflammation is the loss of normal anti-thrombotic and anti-inflammatory potential of the endothelial cells of vasculature. This manifest itself in the form of dysregulation of the coagulation pathway and complement system, pathologic platelet activation, and increased recruitment of leukocyte within the microvasculature. The role of platelets in hemostasis and formation of thrombi under pathologic and non-pathologic conditions is well established. Platelets are anucleate cells known for their essential role in primary hemostasis and the coagulation pathway. In recent years, studies provide strong evidence for the critical involvement of platelets in inflammatory processes like acute ischemic stroke, and viral infections like Coronavirus disease 2019 (COVID-19). This has encouraged the researchers to investigate the contribution of platelets in the pathology of various thrombo-inflammatory diseases. The inhibition of platelet surface receptors or their intracellular signaling which mediate initial platelet activation and adhesion might prove to be suitable targets in thrombo-inflammatory disorders. Thus, the present review summarizes the concept and mechanism of platelet signaling and briefly discuss their role in sterile and non-sterile thrombo-inflammation, with the emphasis on role of platelets in COVID-19 induced thrombo-inflammation. The aim of this review is to summarize the recent developments in deciphering the role of the platelets in thrombo-inflammation and discuss their potential as pharmaceutical targets.
Collapse
Affiliation(s)
- Swati Sharma
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Silvio Antoniak
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
22
|
Pottash AE, Levy D, Jeyaram A, Kuo L, Kronstadt SM, Chao W, Jay SM. Combinatorial microRNA Loading into Extracellular Vesicles for Increased Anti-Inflammatory Efficacy. Noncoding RNA 2022; 8:71. [PMID: 36287123 PMCID: PMC9611452 DOI: 10.3390/ncrna8050071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as promising therapeutic entities in part due to their potential to regulate multiple signaling pathways in target cells. This potential is derived from the broad array of constituent and/or cargo molecules associated with EVs. Among these, microRNAs (miRNAs) are commonly implicated as important and have been associated with a wide variety of EV-induced biological phenomena. While controlled loading of single miRNAs is a well-documented approach for enhancing EV bioactivity, loading of multiple miRNAs has not been fully leveraged to maximize the potential of EV-based therapies. Here, an established approach to extrinsic nucleic acid loading of EVs, sonication, was utilized to load multiple miRNAs in HEK293T EVs. Combinations of miRNAs were compared to single miRNAs with respect to anti-inflammatory outcomes in assays of increasing stringency, with the combination of miR-146a, miR-155, and miR-223 found to have the most potential amongst the tested groups.
Collapse
Affiliation(s)
- Alex Eli Pottash
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Daniel Levy
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Anjana Jeyaram
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Leo Kuo
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Stephanie M. Kronstadt
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, 660 West Redwood Street, Baltimore, MD 21201, USA
| | - Steven M. Jay
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
- Program in Molecular and Cell Biology, University of Maryland, 4062 Campus Drive, College Park, MD 20742, USA
| |
Collapse
|
23
|
Deny M, Arroba Nuñez LA, Romano M, Denis O, Casimir G, Chamekh M. Sex difference in innate inflammatory response and macrophage polarization in Streptococcus agalactiae-induced pneumonia and potential role of microRNA-223-3p. Sci Rep 2022; 12:17126. [PMID: 36224333 PMCID: PMC9555696 DOI: 10.1038/s41598-022-21587-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/29/2022] [Indexed: 01/04/2023] Open
Abstract
While number of studies have shown that biological sex is a risk factor in the incidence and severity of infection-induced inflammatory diseases, the underlying mechanisms are still poorly understood. In this study, we compared the innate inflammatory response in male and female mice with group B streptococcal (GBS)-induced pneumoniae. Although male and female mice displayed similar bacterial burdens, males exhibited more innate inflammatory cytokines and chemokines and a higher proportion of infiltrating monocytes/macrophages. The analysis of the distribution of macrophage subtypes M1 (pro-inflammatory) versus M2 (anti-inflammatory) yielded a higher M1/M2 ratio in infected males compared with females. Given the importance of the chromosome X-linked microRNA-223-3p (miR-223-3p) in modulating the inflammatory process and macrophage polarization, we investigated its potential contribution in sex bias of GBS-induced innate inflammatory response. Knock-down of miR-223-3p with specific antagomiR resulted in increased inflammatory response and higher M1/M2 ratio following GBS infection. Notably, compared to male mice, we detected higher amount of miR-223-3p in macrophages from females that correlated negatively with M1 phenotype. These results suggest that differential expression of miR-233-3p may impact macrophage polarization, thereby contributing to fine-tune sex differences in inflammatory response.
Collapse
Affiliation(s)
- Maud Deny
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Brussels, Belgium
| | - Luis Alexis Arroba Nuñez
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Brussels, Belgium
| | - Marta Romano
- Immune Response Service, Sciensano, Brussels, Belgium
| | - Olivier Denis
- Immune Response Service, Sciensano, Brussels, Belgium
| | - Georges Casimir
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Queen Fabiola University Children's Hospital, Brussels, Belgium
| | - Mustapha Chamekh
- Inflammation Unit, Laboratory of Pediatric Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.
- ULB Center for Research in Immunology (U-CRI), Brussels, Belgium.
| |
Collapse
|
24
|
Wu X, Xu X, Xiang Y, Fan D, An Q, Yue G, Jin Z, Ding J, Hu Y, Du Q, Xu J, Xie R. Exosome-mediated effects and applications in inflammatory diseases of the digestive system. Eur J Med Res 2022; 27:163. [PMID: 36045437 PMCID: PMC9429695 DOI: 10.1186/s40001-022-00792-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Exosomes are membranous vesicles containing RNA and proteins that are specifically secreted in vivo. Exosomes have many functions, such as material transport and signal transduction between cells. Many studies have proven that exosomes can not only be used as biomarkers for disease diagnosis but also as carriers to transmit information between cells. Exosomes participate in a variety of physiological and pathological processes, including the immune response, antigen presentation, cell migration, cell differentiation, and tumour development. Differences in exosome functions depend on cell type. In recent years, exosome origin, cargo composition, and precise regulatory mechanisms have been the focus of research. Although exosomes have been extensively reported in digestive tumours, few articles have reviewed their roles in inflammatory diseases of the digestive system, especially inflammatory-related diseases (such as reflux oesophagitis, gastritis, inflammatory bowel disease, hepatitis, and pancreatitis). This paper briefly summarizes the roles of exosomes in inflammatory diseases of the digestive system to provide a basis for research on the mechanism of inflammatory diseases of the digestive system targeted by exosomes.
Collapse
Affiliation(s)
- Xianli Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Xiaolin Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Yiwei Xiang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Dongdong Fan
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Qiming An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Gengyu Yue
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Zhe Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Jianhong Ding
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Yanxia Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Qian Du
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Jingyu Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China. .,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| | - Rui Xie
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China. .,The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
25
|
Antonakos N, Gilbert C, Théroude C, Schrijver IT, Roger T. Modes of action and diagnostic value of miRNAs in sepsis. Front Immunol 2022; 13:951798. [PMID: 35990654 PMCID: PMC9389448 DOI: 10.3389/fimmu.2022.951798] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a clinical syndrome defined as a dysregulated host response to infection resulting in life-threatening organ dysfunction. Sepsis is a major public health concern associated with one in five deaths worldwide. Sepsis is characterized by unbalanced inflammation and profound and sustained immunosuppression, increasing patient susceptibility to secondary infections and mortality. microRNAs (miRNAs) play a central role in the control of many biological processes, and deregulation of their expression has been linked to the development of oncological, cardiovascular, neurodegenerative and metabolic diseases. In this review, we discuss the role of miRNAs in sepsis pathophysiology. Overall, miRNAs are seen as promising biomarkers, and it has been proposed to develop miRNA-based therapies for sepsis. Yet, the picture is not so straightforward because of the versatile and dynamic features of miRNAs. Clearly, more research is needed to clarify the expression and role of miRNAs in sepsis, and to promote the use of miRNAs for sepsis management.
Collapse
Affiliation(s)
| | | | | | | | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
26
|
Hussein S, Abdelazem AS, Abdelmoneem S, Abdelnabi ALSM, Khamis T, Obaya AA, Ebian HF. Evaluation of miRNA 223/125a and COBLL1 Expressions and ROR-1 Levels as Reliable Markers in B- chronic Lymphocytic Leukemia. Asian Pac J Cancer Prev 2022; 23:2735-2742. [PMID: 36037128 PMCID: PMC9741902 DOI: 10.31557/apjcp.2022.23.8.2735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND miRNA 223 /125a and Cordon-bleu Protein Like 1 (COBLL1) are novel biomarkers that can predict prognosis and guide treatment decisions in patients with chronic lymphocytic leukemia (CLL). Also, there is a growing interest in CLL monitoring based on flow cytometry of receptor tyrosine kinase-like orphan receptor-1 (ROR-1). Objective: This study aimed to evaluate the relationship between miRNA 223 /125a and COBLL1 expressions and ROR-1 expression in patients with CLL. Also, the study evaluated the relationship between the expression of these biomarkers with tumor staging and cancer progression. METHODS Our study included 40 patients newly diagnosed with B-CLL. In peripheral blood (PB), miRNA 223/125a and COBLL1 expressions were detected by real-time polymerase chain reaction (real-time PCR) and ROR-1 percentage was detected by flow cytometry before and after treatment. Results: High level of COBLL1 expression was statistically significantly associated with high ROR-1 percentage expression (P= 0.03). However, a high level of miRNA 223/125a expression was statistically significantly associated with low ROR-1 percentage expression (P=0.002). The sensitivity and specificity of ROR-1 as a predictor of high WBCs count after treatment were 96.6 and 81.1%, respectively. There was a statistically significant reduction of ROR-1 percentage after treatment compared to before treatment (P <0.001). CONCLUSION ROR-1 percentage expression can be considered a possible prognostic predictor in CLL along with miRNA 223/125a and COBLL1 expressions. This can be explained by the significant correlation between ROR-1 and the studied molecular biomarkers; miRNA 223/125a and COBLL1. In addition, there was a significantly higher ROR-1 percentage in patients with higher WBC counts. Moreover, there was a significant reduction in ROR-1 percentage after treatment.
Collapse
Affiliation(s)
- Samia Hussein
- Medical Biochemistry& Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt. ,Ibn Sina University for Medical Sciences, Jordan. ,For Correspondence:
| | | | - Shimaa Abdelmoneem
- Hematologyl unit, Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | | | - Tarek Khamis
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Ahmed Ali Obaya
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Huda F Ebian
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
27
|
Gu J, Xu H, Chen Y, Li N, Hou X. MiR-223 as a Regulator and Therapeutic Target in Liver Diseases. Front Immunol 2022; 13:860661. [PMID: 35371024 PMCID: PMC8965842 DOI: 10.3389/fimmu.2022.860661] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/23/2022] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous non-coding single-stranded small molecule RNAs consisting of 20–24 nucleotides that are highly conserved in species evolution. Expression of miRNAs is strictly tissue-specific, and it is chronological in fungi and plants, as well as in animals. MiR-223 has been shown to play a key role in innate immunity, and dysregulation of its expression contributes to the pathogenesis of multiple inflammatory diseases, and cancers. In this article the biosynthesis and functions of miR-223 in innate immunity are reviewed, and the role of miR-223 in liver physiopathology and therapeutic prospects are highlighted.
Collapse
Affiliation(s)
- Jiarong Gu
- School of Medicine, Ningbo University, Ningbo, China
| | - Hao Xu
- School of Medicine, Ningbo University, Ningbo, China
| | - Yandong Chen
- School of Medicine, Ningbo University, Ningbo, China
| | - Na Li
- School of Medicine, Ningbo University, Ningbo, China
| | - Xin Hou
- School of Medicine, Ningbo University, Ningbo, China.,The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
28
|
Ginckels P, Holvoet P. Oxidative Stress and Inflammation in Cardiovascular Diseases and Cancer: Role of Non-coding RNAs. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:129-152. [PMID: 35370493 PMCID: PMC8961704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
High oxidative stress, Th1/Th17 immune response, M1 macrophage inflammation, and cell death are associated with cardiovascular diseases. Controlled oxidative stress, Th2/Treg anti-tumor immune response, M2 macrophage inflammation, and survival are associated with cancer. MiR-21 protects against cardiovascular diseases but may induce tumor growth by retaining the anti-inflammatory M2 macrophage and Treg phenotypes and inhibiting apoptosis. Down-regulation of let-7, miR-1, miR-9, miR-16, miR-20a, miR-22a, miR-23a, miR-24a, miR-26a, miR-29, miR-30a, miR-34a, miR-124, miR-128, miR-130a, miR-133, miR-140, miR-143-145, miR-150, miR-153, miR-181a, miR-378, and miR-383 may aid cancer cells to escape from stresses. Upregulation of miR-146 and miR-223 may reduce anti-tumor immune response together with miR-21 that also protects against apoptosis. MiR-155 and silencing of let-7e, miR-125, and miR-126 increase anti-tumor immune response. MiR expression depends on oxidative stress, cytokines, MYC, and TGF-β, and expression of silencing lncRNAs and circ-RNAs. However, one lncRNA or circ-RNA may have opposite effects by targeting several miRs. For example, PVT1 induces apoptosis by targeting miR-16a and miR-30a but inhibits apoptosis by silencing miR-17. In addition, levels of a non-coding RNA in a cell type depend not only on expression in that cell type but also on an exchange of microvesicles between cell types and tumors. Although we got more insight into the function of a growing number of individual non-coding RNAs, overall, we do not know enough how several of them interact in functional networks and how their expression changes at different stages of disease progression.
Collapse
Affiliation(s)
- Pieterjan Ginckels
- Department of Architecture, Brussels and Gent, KU Leuven, Leuven, Belgium
| | - Paul Holvoet
- Experimental Cardiology, KU Leuven, Leuven, Belgium,To whom all correspondence should be addressed: Paul Holvoet, Experimental
Cardiology, KU Leuven, Belgium; ; ORCID iD:
https://orcid.org/0000-0001-9201-0772
| |
Collapse
|
29
|
Tian Y, Wang TS, Bu H, Shao G, Zhang W, Zhang L. Role of Exosomal miR-223 in Chronic Skeletal Muscle Inflammation. Orthop Surg 2022; 14:644-651. [PMID: 35293669 PMCID: PMC9002075 DOI: 10.1111/os.13232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 12/30/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
As skeletal muscle is one of the largest organs in the body, its damage can directly reflect a decline in somatic function, thus, further affecting daily life and health. Inflammation is a prerequisite for the repair of injured skeletal muscles. Chronic inflammation induced by inadequate repair in skeletal muscle aggravates tissue injury. Exosomes regulate inflammatory responses to facilitate the repair of skeletal muscle injury. Moreover, exosomal miR‐223 with high specificity is the most abundant miRNA in peripheral blood and regarded as biomarkers for inflammation post skeletal muscle injury, which warrants further investigation. Available studies have demonstrated that exosomal miR‐223 negatively correlates with TNF‐α levels in serum and regulates the canonical inflammatory NF‐κB signaling pathway. miR‐223 is a negative feedback regulator with great potential for adjusting inflammatory imbalance and promoting skeletal muscle repair. The research on the regulation of negative feedback factors in the inflammatory signaling pathway is essential in biology and medicine. Therefore, this review mainly elaborates the formation, heterogeneity and markers of exosomes and points out exosomal miR‐223 as a beneficial role in chronic skeletal muscle inflammation and can be expected to be a potential therapeutic target for skeletal muscle damage.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.,Department of Acupuncture-Moxibustion and Tuina, The Second Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Tie-Shan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - He Bu
- Department of Acupuncture-Moxibustion and Tuina, The Second Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Guo Shao
- Center for Translational Medicine and Department of Laboratory Medicine, the Third People's Hospital of Longgang District, Shenzhen, China
| | - Wei Zhang
- Department of Pathology, the First Affiliated Hospital of Baotou Medical College, Baotou, Inner Mongolia, China
| | - Li Zhang
- Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Millrine D, Jenkins RH, Hughes STO, Jones SA. Making sense of IL-6 signalling cues in pathophysiology. FEBS Lett 2022; 596:567-588. [PMID: 34618359 PMCID: PMC9673051 DOI: 10.1002/1873-3468.14201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022]
Abstract
Unravelling the molecular mechanisms that account for functional pleiotropy is a major challenge for researchers in cytokine biology. Cytokine-receptor cross-reactivity and shared signalling pathways are considered primary drivers of cytokine pleiotropy. However, reports epitomized by studies of Jak-STAT cytokine signalling identify interesting biochemical and epigenetic determinants of transcription factor regulation that affect the delivery of signal-dependent cytokine responses. Here, a regulatory interplay between STAT transcription factors and their convergence to specific genomic enhancers support the fine-tuning of cytokine responses controlling host immunity, functional identity, and tissue homeostasis and repair. In this review, we provide an overview of the signalling networks that shape the way cells sense and interpret cytokine cues. With an emphasis on the biology of interleukin-6, we highlight the importance of these mechanisms to both physiological processes and pathophysiological outcomes.
Collapse
Affiliation(s)
- David Millrine
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
- Present address:
Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSir James Black CentreSchool of Life SciencesUniversity of Dundee3rd FloorDundeeUK
| | - Robert H. Jenkins
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Stuart T. O. Hughes
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Simon A. Jones
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| |
Collapse
|
31
|
Buffolo F, Monticone S, Camussi G, Aikawa E. Role of Extracellular Vesicles in the Pathogenesis of Vascular Damage. Hypertension 2022; 79:863-873. [PMID: 35144490 PMCID: PMC9010370 DOI: 10.1161/hypertensionaha.121.17957] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracellular vesicles (EVs) are nanosized membrane-bound structures released by cells that are able to transfer nucleic acids, protein cargos, and metabolites to specific recipient cells, allowing cell-to-cell communications in an endocrine and paracrine manner. Endothelial, leukocyte, and platelet-derived EVs have emerged both as biomarkers and key effectors in the development and progression of different stages of vascular damage, from earliest alteration of endothelial function, to advanced atherosclerotic lesions and cardiovascular calcification. Under pathological conditions, circulating EVs promote endothelial dysfunction by impairing vasorelaxation and instigate vascular inflammation by increasing levels of adhesion molecules, reactive oxygen species, and proinflammatory cytokines. Platelets, endothelial cells, macrophages, and foam cells secrete EVs that regulate macrophage polarization and contribute to atherosclerotic plaque progression. Finally, under pathological stimuli, smooth muscle cells and macrophages secrete EVs that aggregate between collagen fibers and serve as nucleation sites for ectopic mineralization in the vessel wall, leading to formation of micro- and macrocalcification. In this review, we summarize the emerging evidence of the pathological role of EVs in vascular damage, highlighting the major findings from the most recent studies and discussing future perspectives in this research field.
Collapse
Affiliation(s)
- Fabrizio Buffolo
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy. (F.B., S.M.).,Center for Interdisciplinary Cardiovascular Sciences, Department of Cardiovascular Medicine (F.B, E.A.)
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy. (F.B., S.M.)
| | - Giovanni Camussi
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Italy. (G.C.)
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Department of Cardiovascular Medicine (F.B, E.A.).,Center for Excellence in Vascular Biology, Department of Cardiovascular Medicine (E.A.)
| |
Collapse
|
32
|
Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity 2022; 55:31-55. [PMID: 35021057 PMCID: PMC8773457 DOI: 10.1016/j.immuni.2021.12.013] [Citation(s) in RCA: 627] [Impact Index Per Article: 313.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023]
Abstract
Obesity leads to chronic, systemic inflammation and can lead to insulin resistance (IR), β-cell dysfunction, and ultimately type 2 diabetes (T2D). This chronic inflammatory state contributes to long-term complications of diabetes, including non-alcoholic fatty liver disease (NAFLD), retinopathy, cardiovascular disease, and nephropathy, and may underlie the association of type 2 diabetes with other conditions such as Alzheimer's disease, polycystic ovarian syndrome, gout, and rheumatoid arthritis. Here, we review the current understanding of the mechanisms underlying inflammation in obesity, T2D, and related disorders. We discuss how chronic tissue inflammation results in IR, impaired insulin secretion, glucose intolerance, and T2D and review the effect of inflammation on diabetic complications and on the relationship between T2D and other pathologies. In this context, we discuss current therapeutic options for the treatment of metabolic disease, advances in the clinic and the potential of immune-modulatory approaches.
Collapse
Affiliation(s)
- Theresa V. Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel T. Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jerrold M. Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland.,Correspondence:
| |
Collapse
|
33
|
Alwani A, Andreasik A, Szatanek R, Siedlar M, Baj-Krzyworzeka M. The Role of miRNA in Regulating the Fate of Monocytes in Health and Cancer. Biomolecules 2022; 12:100. [PMID: 35053248 PMCID: PMC8773712 DOI: 10.3390/biom12010100] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 12/23/2022] Open
Abstract
Monocytes represent a heterogeneous population of blood cells that provide a link between innate and adaptive immunity. The unique potential of monocytes as both precursors (e.g., of macrophages) and effector cells (as phagocytes or cytotoxic cells) makes them an interesting research and therapeutic target. At the site of a tumor, monocytes/macrophages constitute a major population of infiltrating leukocytes and, depending on the type of tumor, may play a dual role as either a bad or good indicator for cancer recovery. The functional activity of monocytes and macrophages derived from them is tightly regulated at the transcriptional and post-transcriptional level. This review summarizes the current understanding of the role of small regulatory miRNA in monocyte formation, maturation and function in health and cancer development. Additionally, signatures of miRNA-based monocyte subsets and the influence of exogenous miRNA generated in the tumor environment on the function of monocytes are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland; (A.A.); (A.A.); (R.S.); (M.S.)
| |
Collapse
|
34
|
Shan Y, Chen Y, Brkić J, Fournier L, Ma H, Peng C. miR-218-5p Induces Interleukin-1β and Endovascular Trophoblast Differentiation by Targeting the Transforming Growth Factor β-SMAD2 Pathway. Front Endocrinol (Lausanne) 2022; 13:842587. [PMID: 35299960 PMCID: PMC8920978 DOI: 10.3389/fendo.2022.842587] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/02/2022] [Indexed: 01/10/2023] Open
Abstract
The acquisition of an endovascular trophoblast (enEVT) phenotype is essential for normal placental development and healthy pregnancy. MicroRNAs (miRNAs) are small noncoding RNAs that play critical roles in regulating gene expression. We have recently reported that miR-218-5p promotes enEVT differentiation and spiral artery remodeling in part by targeting transforming growth factor β2 (TGFβ2). We also identified IL1B, which encodes interleukin 1β (IL1β), as one of the most highly upregulated genes by miR-218-5p. In this study, we investigated how miR-218-5p regulates IL1B expression and IL1β secretion and the potential role of IL1β in enEVT differentiation. Using two cell lines derived from extravillous trophoblasts (EVTs), HTR-8/SVneo and Swan 71, we found that stable overexpression of miR-218-5p precursor, mir-218-1, or transient transfection of miR-218-5p mimic, significantly increased IL1B mRNA and IL1β protein levels in cells and conditioned media. We also showed that miR-218-5p directly interacted with SMAD2 3'UTR and reduced SMAD2 at mRNA and protein levels. Knockdown of SMAD2 induced IL1B expression and attenuated the inhibitory effect of TGFβ2 on IL1B expression. On the other hand, overexpression of SMAD2 reduced IL1β levels and blocked the stimulatory effects of miR-218-5p on IL1B expression, trophoblast migration and endothelial-like network formation. In addition, treatment of trophoblasts with IL1β induced the formation of endothelial-like networks and the expression of enEVT markers in a dose-dependent manner. These results suggest that miR-218-5p inhibits the TGFβ/SMAD2 pathway to induce IL1β and enEVT differentiation. Finally, low doses of IL1β also inhibited the expression of miR-218-5p, suggesting the existence of a negative feedback regulatory loop. Taken together, our findings suggest a novel interactive miR-218-5p/TGFβ/SMAD2/IL1β signaling nexus that regulates enEVT differentiation.
Collapse
Affiliation(s)
- Yanan Shan
- Department of Biology, York University, Toronto, ON, Canada
| | - Yan Chen
- Department of Biology, York University, Toronto, ON, Canada
| | - Jelena Brkić
- Department of Biology, York University, Toronto, ON, Canada
| | | | - Haiying Ma
- Department of Biology, York University, Toronto, ON, Canada
| | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, ON, Canada
- *Correspondence: Chun Peng,
| |
Collapse
|
35
|
Yuan S, Wu Q, Wang Z, Che Y, Zheng S, Chen Y, Zhong X, Shi F. miR-223: An Immune Regulator in Infectious Disorders. Front Immunol 2021; 12:781815. [PMID: 34956210 PMCID: PMC8702553 DOI: 10.3389/fimmu.2021.781815] [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: 09/23/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are diminutive noncoding RNAs that can influence disease development and progression by post-transcriptionally regulating gene expression. The anti-inflammatory miRNA, miR-223, was first identified as a regulator of myelopoietic differentiation in 2003. This miR-223 exhibits multiple regulatory functions in the immune response, and abnormal expression of miR-223 is shown to be associated with multiple infectious diseases, including viral hepatitis, human immunodeficiency virus type 1 (HIV-1), and tuberculosis (TB) by influencing neutrophil infiltration, macrophage function, dendritic cell (DC) maturation and inflammasome activation. This review summarizes the current understanding of miR-223 physiopathology and highlights the molecular mechanism by which miR-223 regulates immune responses to infectious diseases and how it may be targeted for diagnosis and treatment.
Collapse
Affiliation(s)
- Shun Yuan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qi Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanjia Che
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Sihao Zheng
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyang Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohan Zhong
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Shi
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Song F, Li JZ, Wu Y, Wu WY, Wang Y, Li G. Ubiquitinated ligation protein NEDD4L participates in MiR-30a-5p attenuated atherosclerosis by regulating macrophage polarization and lipid metabolism. MOLECULAR THERAPY - NUCLEIC ACIDS 2021; 26:1303-1317. [PMID: 34853729 PMCID: PMC8609110 DOI: 10.1016/j.omtn.2021.10.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/22/2021] [Accepted: 10/28/2021] [Indexed: 11/04/2022]
Abstract
MiR-30a-5p plays an important role in various cardiovascular diseases, but its effect in atherosclerosis has not been reported. Apolipoprotein E-deficient (Apo E−/−) mice were used to investigate the role of miR-30a-5p in atherosclerosis, and the underlying mechanism was investigated in vivo and in vitro. The fluorescence in situ hybridization test revealed that miR-30a-5p was expressed in Apo E−/− mice lesions. Nevertheless, in RAW264.7 macrophages, the expression of miR-30a-5p was reduced by lipopolysaccharide (LPS) or oxidized low-density lipoprotein. MiR-30a-5p-ago-treated Apo E−/− mice significantly reduced lesion areas in the aorta and aortic root, reduced levels of lipoprotein and pro-inflammatory cytokines, and increased levels of anti-inflammatory cytokines. The ratio of M1/M2 macrophages was decreased in miR-30a-5p-ago-treated Apo E−/− mice and LPS-treated RAW264.7 macrophages by the regulation of Smad-1/2 phosphorylation. MiR-30a-5p reduced lipid uptake in oxidized low-density lipoprotein-treated macrophages by regulating the expression of PPAR-γ, ABCA1, ABCG1, LDLR, and PCSK9. Ubiquitinated ligase NEDD4L was identified as a target of miR-30a-5p. Interestingly, knockdown of NEDD4L decreased the M1/M2 ratio and oxidized low-density lipoprotein uptake in macrophages by inhibiting the ubiquitination of PPAR-γ and phosphorylation of Smad-1/2 and regulating ABCA1, ABCG1, LDLR, and PCSK9. We demonstrated a novel effect and mechanism of miR-30a-5p in atherosclerosis.
Collapse
|
37
|
Deng Y, Tong J, Shi W, Tian Z, Yu B, Tang J. Thromboangiitis obliterans plasma-derived exosomal miR-223-5p inhibits cell viability and promotes cell apoptosis of human vascular smooth muscle cells by targeting VCAM1. Ann Med 2021; 53:1129-1141. [PMID: 34259105 PMCID: PMC8281010 DOI: 10.1080/07853890.2021.1949487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Exosomes-encapsulated microRNAs (miRNAs) have been established to be implicated in the pathogenesis of different diseases. Nevertheless, circulating exosomal miRNAs of thromboangiitis obliterans (TAO) remains poorly understood. This study aimed to explore the effects of exosomal miRNAs associated with TAO on human vascular smooth muscle cells (HVSMCs).Methods: The exosomes were isolated from the plasma of TAO patients and normal controls and then were sent for small RNA sequencing. Differentially expressed miRNAs (DE-miRNAs) were identified by bioinformatics analysis and were confirmed by RT-qPCR. After that, PKH67 staining was used to label exosomes and co-cultured with HVSMCs. Cell viability and apoptosis were, respectively, tested by CCK-8 assay and flow cytometry. Finally, dual-luciferase reporter assay was used to confirm the downstream targets of miR-223-5p.Results: A total of 39 DE-miRNAs were identified between TAO patients and normal controls, of which, miR-223-5p was one of the most significantly up-regulated miRNAs. TAO plasma-derived exosomes or miR-223-5p mimics inhibited cell viability of HVSMCs and promoted cell apoptosis. The pro-apoptotic effect of TAO plasma-derived exosomes was alleviated by miR-223-5p inhibitor. Additionally, the expressions of VCAM1 and IGF1R were down-regulated by exosomes and miR-223-5p mimics, and were abrogated by miR-223-5p inhibitor. Dual-luciferase report showed that VCAM1 was the target of miR-223-5p.Conclusions: Our findings imply that circulating exosomal miR-223-5p may play an essential role in the pathogenesis of TAO, and provide a basis for miR-6515-5p/VCAM1 as novel therapeutic targets and pathways for TAO treatment.
Collapse
Affiliation(s)
- Ying Deng
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jindong Tong
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Weijun Shi
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhongyi Tian
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jingdong Tang
- Department of Vascular Surgery, Shanghai Pudong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
38
|
Circulating miRNAs as Potential Biomarkers Distinguishing Relapsing-Remitting from Secondary Progressive Multiple Sclerosis. A Review. Int J Mol Sci 2021; 22:ijms222111887. [PMID: 34769314 PMCID: PMC8584709 DOI: 10.3390/ijms222111887] [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: 09/21/2021] [Revised: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating neurodegenerative, highly heterogeneous disease with a variable course. The most common MS subtype is relapsing–remitting (RR), having interchanging periods of worsening and relative stabilization. After a decade, in most RR patients, it alters into the secondary progressive (SP) phase, the most debilitating one with no clear remissions, leading to progressive disability deterioration. Among the greatest challenges for clinicians is understanding disease progression molecular mechanisms, since RR is mainly characterized by inflammatory processes, while in SP, the neurodegeneration prevails. This is especially important because distinguishing RR from the SP subtype early will enable faster implementation of appropriate treatment. Currently, the MS course is not well-correlated with the biomarkers routinely used in clinical practice. Despite many studies, there are still no reliable indicators correlating with the disease stage and its activity degree. Circulating microRNAs (miRNAs) may be considered valuable molecules for the MS diagnosis and, presumably, helpful in predicting disease subtype. MiRNA expression dysregulation is commonly observed in the MS course. Moreover, knowledge of diverse miRNA panel expression between RRMS and SPMS may allow for deterring disability progression through successful treatment. Therefore, in this review, we address the current state of research on differences in miRNA panel expression between the phases.
Collapse
|
39
|
Amini-Farsani Z, Yadollahi-Farsani M, Arab S, Forouzanfar F, Yadollahi M, Asgharzade S. Prediction and analysis of microRNAs involved in COVID-19 inflammatory processes associated with the NF-kB and JAK/STAT signaling pathways. Int Immunopharmacol 2021; 100:108071. [PMID: 34482267 PMCID: PMC8378592 DOI: 10.1016/j.intimp.2021.108071] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
COVID-19 is the cause of a pandemic associated with substantial morbidity and mortality. As yet, there is no available approved drug to eradicate the virus. In this review article, we present an alternative study area that may contribute to the development of therapeutic targets for COVID-19. Growing evidence is revealing further pathophysiological mechanisms of COVID-19 related to the disregulation of inflammation pathways that seem to play a critical role toward COVID-19 complications. The NF-kB and JAK/STAT signaling pathways are highly activated in acute inflammation, and the excessive activity of these pathways in COVID-19 patients likely exacerbates the inflammatory responses of the host. A group of non-coding RNAs (miRNAs) manage certain features of the inflammatory process. In this study, we discuss recent advances in our understanding of miRNAs and their connection to inflammatory responses. Additionally, we consider the link between perturbations in miRNA levels and the onset of COVID-19 disease. Furthermore, previous studies published in the online databases, namely web of science, MEDLINE (PubMed), and Scopus, were reviewed for the potential role of miRNAs in the inflammatory manifestations of COVID-19. Moreover, we disclosed the interactions of inflammatory genes using STRING DB and designed interactions between miRNAs and target genes using Cityscape software. Several miRNAs, particularly miR-9, miR-98, miR-223, and miR-214, play crucial roles in the regulation of NF-kB and JAK-STAT signaling pathways as inflammatory regulators. Therefore, this group of miRNAs that mitigate inflammatory pathways can be further regarded as potential targets for far-reaching-therapeutic strategies in COVID-19 diseases.
Collapse
Affiliation(s)
- Zeinab Amini-Farsani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran
| | - Mahtab Yadollahi-Farsani
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samaneh Arab
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mitra Yadollahi
- Department of Operative Dentistry, School of Dentistry, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
40
|
Li Y, Tan J, Miao Y, Zhang Q. MicroRNA in extracellular vesicles regulates inflammation through macrophages under hypoxia. Cell Death Dis 2021; 7:285. [PMID: 34635652 PMCID: PMC8505641 DOI: 10.1038/s41420-021-00670-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
Extracellular vesicle (EV), critical mediators of cell-cell communication, allow cells to exchange proteins, lipids, and genetic material and therefore profoundly affect the general homeostasis. A hypoxic environment can affect the biogenesis and secrete of EVs, and the cargoes carried can participate in a variety of physiological and pathological processes. In hypoxia-induced inflammation, microRNA(miRNA) in EV participates in transcriptional regulation through various pathways to promote or reduce the inflammatory response. Meanwhile, as an important factor of immune response, the polarization of macrophages is closely linked to miRNAs, which will eventually affect the inflammatory state. In this review, we outline the possible molecular mechanism of EV changes under hypoxia, focusing on the signaling pathways of several microRNAs involved in inflammation regulation and describing the process and mechanism of EV-miRNAs regulating macrophage polarization in hypoxic diseases.
Collapse
Affiliation(s)
- Ye Li
- grid.412645.00000 0004 1757 9434Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| | - Jin Tan
- grid.412645.00000 0004 1757 9434Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| | - Yuyang Miao
- grid.265021.20000 0000 9792 1228Tianjin Medical University, 300052 Tianjin, China
| | - Qiang Zhang
- grid.412645.00000 0004 1757 9434Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, 300052 Tianjin, China
| |
Collapse
|
41
|
Ahmad A. Epigenetic regulation of immunosuppressive tumor-associated macrophages through dysregulated microRNAs. Semin Cell Dev Biol 2021; 124:26-33. [PMID: 34556420 DOI: 10.1016/j.semcdb.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/02/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023]
Abstract
Macrophages are immune cells that play different roles under different physiological conditions. They are present in all tissues where they primarily protect from bacteria and pathogens in addition to assisting in tissue repair. During tumor progression, macrophages can exert contrasting effects based on the M1 vs. M2 polarization. The M2 macrophages support tumor growth through mechanisms that help suppress immune responses and/or circumvent immune-surveillance. A number of such mechanisms such as production of IL-10 and arginase, and expression of PD-L1, V-domain Ig suppressor of T cell activation and B7 family molecule B7-H4 are now believed central to the immunosuppressive effects of tumor-associated macrophages (TAMs). Emerging data has identified epigenetic regulation of these immunosuppressive mechanisms by small non-coding RNAs, the microRNAs (miRNAs). This review discusses the available literature on the subject, including the exosomes mediated transfer of miRNAs between cancer cells and the macrophages within the tumor microenvironment. A number of miRNAs are now believed to be involved in TAMs' production of IL-10 and expression of PD-L1 while the information on such regulation of other immunosuppressive mechanisms is slowly emerging. A better understanding of epigenetic regulation of macrophages-mediated immunosuppressive effect can help identify novel targets for therapy and aid the design of future studies aimed at sensitizing tumors to immune responses.
Collapse
Affiliation(s)
- Aamir Ahmad
- Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
42
|
The Role of miRNA in the Pathophysiology of Neuroendocrine Tumors. Int J Mol Sci 2021; 22:ijms22168569. [PMID: 34445276 PMCID: PMC8395312 DOI: 10.3390/ijms22168569] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/16/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022] Open
Abstract
Neuroendocrine tumors (NETs) represent a tumor group that is both rare and heterogeneous. Prognosis is largely determined by the tumor grading and the site of the primary tumor and metastases. Despite intensive research efforts, only modest advances in diagnostic and therapeutic approaches have been achieved in recent years. For patients with non-respectable tumor stages, prognosis is poor. In this context, the development of novel diagnostic tools for early detection of NETs and prediction of tumor response to therapy as well as estimation of the overall prognosis would greatly improve the clinical management of NETs. However, identification of novel diagnostic molecules is hampered by an inadequate understanding of the pathophysiology of neuroendocrine malignancies. It has recently been demonstrated that microRNA (miRNA), a family of small RNA molecules with an established role in the pathophysiology of quite different cancer entities, may also play a role as a biomarker. Here, we summarize the available knowledge on the role of miRNAs in the development of NET and highlight their potential use as serum-based biomarkers in the context of this disease. We discuss important challenges currently preventing their use in clinical routine and give an outlook on future directions of miRNA research in NET.
Collapse
|
43
|
Liu Y, Wan X, Yuan Y, Huang J, Jiang Y, Zhao K, Wang Y, Liu Y, Wang Q, Jin H. Opposite effects of miR-155 in the initial and later stages of lipopolysaccharide (LPS)-induced inflammatory response. J Zhejiang Univ Sci B 2021; 22:590-598. [PMID: 34269011 DOI: 10.1631/jzus.b2000826] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although microRNA-155 (miR-155) is considered a pro-inflammatory mediator, cumulative evidence indicates that it also has anti-inflammatory effects in macrophages and dendritic cells. In this study, we identified the dramatic expression changes of more than half of potential miR-155-targeted genes upon lipopolysaccharide (LPS) stimulation; 223 genes were down-regulated and 85 genes were up-regulated, including suppressor of cytokine signaling 1 (SOCS1) and transforming growth factor-β-activated kinase 1-binding protein 2 (TAB2), two well-known genes involved in miR-155-mediated regulation of the Toll-like receptor 4 (TLR4) signaling pathway. We also found that miR-155 acted as an anti-inflammatory mediator in the initial stage of LPS-induced inflammatory response mainly through repressing TAB2 protein translation, and as a pro-inflammatory mediator by down-regulating SOCS1 in the later stage. Meanwhile, overexpression of TAB2 3' untranslated region (UTR) in macrophages promoted the development of endotoxin tolerance by competing for binding with miR-155, which resulted in an elevated expression level of SOCS1 protein. These findings provide new insights for understanding the regulatory mechanisms in fine-tuning of LPS-induced innate immune response.
Collapse
Affiliation(s)
- Yuhua Liu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou 310016, China.,Department of Clinical Laboratory, the Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Xiaopeng Wan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuan Yuan
- Department of Clinical Laboratory, the Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Jingjing Huang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yijia Jiang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kaiyue Zhao
- Department of Radiology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Yan Wang
- Department of Clinical Laboratory, the Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Yang Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
44
|
Wu CY, Yang HY, Huang JL, Lai JH. Signals and Mechanisms Regulating Monocyte and Macrophage Activation in the Pathogenesis of Juvenile Idiopathic Arthritis. Int J Mol Sci 2021; 22:ijms22157960. [PMID: 34360720 PMCID: PMC8347893 DOI: 10.3390/ijms22157960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Monocytes (Mos) and macrophages (Mφs) are key players in the innate immune system and are critical in coordinating the initiation, expansion, and regression of many autoimmune diseases. In addition, they display immunoregulatory effects that impact inflammation and are essential in tissue repair and regeneration. Juvenile idiopathic arthritis (JIA) is an umbrella term describing inflammatory joint diseases in children. Accumulated evidence suggests a link between Mo and Mφ activation and JIA pathogenesis. Accordingly, topics regarding the signals and mechanisms regulating Mo and Mφ activation leading to pathologies in patients with JIA are of great interest. In this review, we critically summarize recent advances in the understanding of how Mo and Mφ activation is involved in JIA pathogenesis and focus on the signaling pathways and mechanisms participating in the related cell activation processes.
Collapse
Affiliation(s)
- Chao-Yi Wu
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-Y.W.); (J.-L.H.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Huang-Yu Yang
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Jing-Long Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-Y.W.); (J.-L.H.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei City 236, Taiwan
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan
- National Defense Medical Center, Graduate Institute of Medical Science, Taipei 114, Taiwan
- Correspondence: ; Tel./Fax: +886-2-8791-8382
| |
Collapse
|
45
|
Wang X, Zhang H, Guo R, Li X, Liu H, Wang Z, Du Q, Tong D, Huang Y. MicroRNA-223 modulates the IL-4-medicated macrophage M2-type polarization to control the progress of sepsis. Int Immunopharmacol 2021; 96:107783. [PMID: 34162147 DOI: 10.1016/j.intimp.2021.107783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
MicroRNAs play a variety of roles in the progress of inflammation. Herein, we investigated the roles of miR-223 in governing macrophage polarization balance in the progress of sepsis. We firstly observed that miR-223 was down-regulated at the early phase and up-regulated at the late phase of sepsis in macrophages; the levels of miR-223 were positively correlated to the ratio of M2 macrophages during sepsis. In miR-223 knockout mice, we observed that miR-223 was dispensable for efficient pro-inflammatory responses, but was required for efficient M2-associated phenotype and function. miR-223 deletion increased clinical scores of sepsis, leading to increased mortality in septic mice. Furthermore, we found that miR-223 expression in M2-type macrophages was controlled by interleukin (IL)-4, but not IL-10; IL-4 antibodies were able to downregulate the levels of miR-223, increased the expression of targeted genes Nfat5 and Rasa1, reduced the ratio of M2 macrophages, resulting in a decreased survival rate in septic mice. Meanwhile, miR-223 deficient macrophages appeared a markedly decreased M2-type polarization when induced by IL-4, but did not affect macrophages skew to M2 phenotype induced by IL-10. Taken together, our results demonstrate that miR-223 acts as an important regulator to modulate IL-4-meditated M2-type polarization of macrophages via targeting to Nfat5 and Rasa1 to control the progress of sepsis.
Collapse
Affiliation(s)
- Xiaoya Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Hai Zhang
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, No. 169 Changle West Road, Xi'an 710032, China; Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Rui Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xiaomin Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Haixin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zhenyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
46
|
Inoue K, Ng C, Xia Y, Zhao B. Regulation of Osteoclastogenesis and Bone Resorption by miRNAs. Front Cell Dev Biol 2021; 9:651161. [PMID: 34222229 PMCID: PMC8249944 DOI: 10.3389/fcell.2021.651161] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/12/2021] [Indexed: 01/12/2023] Open
Abstract
Osteoclasts are specialized bone-resorbing cells that contribute to physiological bone development and remodeling in bone metabolism throughout life. Abnormal production and activation of osteoclasts lead to excessive bone resorption in pathological conditions, such as in osteoporosis and in arthritic diseases with bone destruction. Recent epigenetic studies have shed novel insight into the dogma of the regulation of gene expression. microRNAs belong to a category of epigenetic regulators, which post-transcriptionally regulate and silence target gene expression, and thereby control a variety of biological events. In this review, we discuss miRNA biogenesis, the mechanisms utilized by miRNAs, several miRNAs that play important roles in osteoclast differentiation, function, survival and osteoblast-to-osteoclast communication, and their translational potential and challenges in bone biology and skeletal diseases.
Collapse
Affiliation(s)
- Kazuki Inoue
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, United States,Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, United States
| | - Yuhan Xia
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, United States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, United States,Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Graduate Program in Cell and Developmental Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, United States,*Correspondence: Baohong Zhao,
| |
Collapse
|
47
|
Lee LK, Eghbali M, Sapru A. A novel miRNA biomarker panel associated with mortality in pediatric patients with ARDS. Respir Res 2021; 22:169. [PMID: 34088312 PMCID: PMC8177032 DOI: 10.1186/s12931-021-01761-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/27/2021] [Indexed: 11/10/2022] Open
Abstract
We identified a novel microRNA biomarker panel consisting of 6 microRNAs predicting mortality in pediatric acute respiratory distress syndrome patients. Each of the identified mRNA have potential mechanistic importance in acute respiratory distress syndrome and may lead to the development of pharmacologic targets.
Collapse
Affiliation(s)
- Lisa K Lee
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, 757 Westwood Plaza, Suite 3325, Los Angeles, CA, 90095, USA.
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, 757 Westwood Plaza, Suite 3325, Los Angeles, CA, 90095, USA
| | - Anil Sapru
- Division of Pediatric Critical Care, Department of Pediatrics, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, MDCC 488, Los Angeles, CA, 90095, USA
| |
Collapse
|
48
|
Jiao P, Wang XP, Luoreng ZM, Yang J, Jia L, Ma Y, Wei DW. miR-223: An Effective Regulator of Immune Cell Differentiation and Inflammation. Int J Biol Sci 2021; 17:2308-2322. [PMID: 34239357 PMCID: PMC8241730 DOI: 10.7150/ijbs.59876] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) play a critical role in regulating various biological processes, such as cell differentiation and immune modulation by binding to their target genes. miR-223 is a miRNA with important functions and has been widely investigated in recent years. Under certain physiological conditions, miR-223 is regulated by different transcription factors, including sirtuin1 (Sirt1), PU.1 and Mef2c, and its biological functions are mediated through changes in its cellular or tissue expression. This review paper summarizes miR-223 biosynthesis and its regulatory role in the differentiation of granulocytes, dendritic cells (DCs) and lymphocytes, macrophage polarization, and endothelial and epithelial inflammation. In addition, it describes the molecular mechanisms of miR-223 in regulating lung inflammation, rheumatoid arthritis, enteritis, neuroinflammation and mastitis to provide insights into the existing molecular regulatory networks and therapies for inflammatory diseases in humans and animals.
Collapse
Affiliation(s)
- Peng Jiao
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Xing-Ping Wang
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Zhuo-Ma Luoreng
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Jian Yang
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Li Jia
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Da-Wei Wei
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| |
Collapse
|
49
|
Chen X, Liu Y, Gao Y, Shou S, Chai Y. The roles of macrophage polarization in the host immune response to sepsis. Int Immunopharmacol 2021; 96:107791. [PMID: 34162154 DOI: 10.1016/j.intimp.2021.107791] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/31/2022]
Abstract
Sepsis is a life-threatening clinical syndrome caused by infection. Its pathogenesis is complex and entails coagulation dysfunction, inflammation, and immune disorders. Macrophages are important components of innate and adaptive immunity that are highly heterogeneous and plastic. They can polarize into a multi-dimensional spectrum of phenotypes with different functions relating to immune regulation in response to changes in the microenvironment of specific tissues. We reviewed studies that examined the role of macrophage polarization with a focus on the classical activated (M1-like) and alternative activated (M2-like) macrophages as the two main phenotypes involved in the host immune response to sepsis. A complex regulatory network is involved in the process of macrophage polarization, which is influenced by a variety of signaling molecules, transcription factors, epigenetic modifications, and metabolic reprogramming. M1-like macrophages release large quantities of pro-inflammatory mediators, while M2-like macrophages release large quantities of anti-inflammatory mediators. An imbalance between M1-like and M2-like macrophages induces the occurrence and development of sepsis. Therefore, targeted regulation of the process of macrophage polarization could be a useful approach to normalize the immune balance of the host, offering a new treatment modality for different stages of sepsis.
Collapse
Affiliation(s)
- Xinsen Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yancun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yulei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanfen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
50
|
Extracellular Vesicles in Synovial Fluid from Rheumatoid Arthritis Patients Contain miRNAs with Capacity to Modulate Inflammation. Int J Mol Sci 2021; 22:ijms22094910. [PMID: 34066338 PMCID: PMC8125513 DOI: 10.3390/ijms22094910] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
In rheumatoid arthritis (RA), extracellular vesicles (EVs) are associated with both the propagation and attenuation of joint inflammation and destruction. However, the specific EV content responsible for these processes is largely unknown. Investigations into identifying EV content are confounded by the challenges in obtaining high-quality EV preparations from synovial fluid. Implementing a size exclusion chromatography-based method of EV isolation, coupled with small RNA sequencing, we accurately characterised EV miRNAs in synovial fluid obtained from RA patients and investigated the differences between joints with high- and low-grade inflammation. Synovial fluid was obtained from the joints of 12 RA patients and, based on leukocyte counts, classified as either high (n = 7)- or low (n = 5)-grade inflammation. Using size exclusion chromatography, EVs were purified and small RNA was extracted and sequenced on a NextSeq 500. Sequencing reads were aligned to miRBase v21, and differences in miRNA profiles between RA patients with high- and low-grade joint inflammation were analysed. In total, 1972 distinct miRNAs were identified from RA synovial fluid EVs. miRNAs with less than five reads in fewer than five patients were filtered out, leaving 318 miRNAs for analysis. Analysis of the most abundant miRNAs suggested that they negatively regulate multiple genes relevant to inflammation, including signal transducer and activator of transcription 3 (STAT3), which lies downstream of IL-6 and has a pro-inflammatory role in RA. Synovial fluid from joints with high-grade inflammation contained 3.5-fold more EV miRNA per mL of synovial fluid (p = 0.0017). Seventy-eight EV miRNAs were differentially expressed between RA joints with high- and low-grade inflammation, and pathway analysis revealed that their target genes were commonly involved a variety of processes, including cellular apoptosis, proliferation and migration. Of the 49 miRNAs that were elevated in joints with high-grade inflammation, pathway analysis revealed that genes involved in cytokine-mediated signalling pathways were significantly enriched targets. In contrast, genes associated with reactive oxygen species signalling were significantly enriched as targets of the 29 miRNAs elevated in joints with low-grade inflammation. Our study identified an abundance of EV miRNAs from the synovial fluid of RA patients with the potential to modulate inflammation. In doing so, we defined potential mechanisms by which synovial fluid EVs may contribute to RA pathophysiology.
Collapse
|