1
|
Lago T, Cardoso TM, Rocha A, Carvalho EM, Castellucci LC. Short communication: The miR-155a-5p is correlated with increased ROS and impaired apoptosis in macrophages infected by Leishmania braziliensis. PLoS One 2024; 19:e0298458. [PMID: 38381750 PMCID: PMC10880991 DOI: 10.1371/journal.pone.0298458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Cutaneous leishmaniasis (CL) caused by Leishmania braziliensis, is a disease characterized by well-limited ulcerated lesions with raised borders in exposed parts of the body. miRNAs are recognized for their role in the complex and plastic interaction between host and pathogens, either as part of the host's strategy to neutralize infection or as a molecular mechanism employed by the pathogen to modulate host inflammatory pathways to remain undetected. The mir155 targets a broad range of inflammatory mediators, following toll-like receptors (TLRs) signaling. In this work, we evaluated the effects of the expression of miR155a-5p in human macrophages infected with L. braziliensis. Our results show that miR155a-5p is inversely correlated with early apoptosis and conversely, seems to influence an increment in the oxidative burst in these cells. Altogether, we spotted a functional role of the miR155a-5p in CL pathogenesis, raising the hypothesis that an increased miR-155 expression by TLR ligands influences cellular mechanisms settled to promote both killing and control of parasite density after infection.
Collapse
Affiliation(s)
- Tainã Lago
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Brazil
| | - Thyago Marconi Cardoso
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Brazil
- Laboratório de Pesquisas Clínicas—LAPEC—Instituto Gonçalo Moniz-FIOCRUZ, Salvador, Bahia, Brazil
| | - Alan Rocha
- Laboratório de Pesquisas Clínicas—LAPEC—Instituto Gonçalo Moniz-FIOCRUZ, Salvador, Bahia, Brazil
| | - Edgar M. Carvalho
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Brazil
- Laboratório de Pesquisas Clínicas—LAPEC—Instituto Gonçalo Moniz-FIOCRUZ, Salvador, Bahia, Brazil
| | - Léa Cristina Castellucci
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Brazil
| |
Collapse
|
2
|
Liyanage TD, Nikapitiya C, De Zoysa M. Chitosan nanoparticles-based in vivo delivery of miR-155 modulates the Viral haemorrhagic septicaemia virus-induced antiviral immune responses in zebrafish (Danio rerio). FISH & SHELLFISH IMMUNOLOGY 2024; 144:109234. [PMID: 37984615 DOI: 10.1016/j.fsi.2023.109234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Viral haemorrhagic septicaemia virus (VHSV) is one of the highly pathogenic virus, which causes viral haemorrhagic septicaemia disease in both marine and freshwater fish. Micro RNA-155 (miRNA-155) is a multifunctional small non-coding RNA and it involves regulation of immune responses during viral infection. In this study, dre-miR-155 mimics were encapsulated into chitosan nanoparticles (CNPs). Resulted encapsulated product (miR-155-CNPs) was investigated for its immunomodulation role in zebrafish during experimentally challenged VHSV infection. Successful encapsulation of dre-miR-155 mimics into CNPs was confirmed through average nanoparticle (NPs) size (341.45 ± 10.00 nm), increased encapsulation efficiency percentage (98.80%), bound dre-miR-155 with chitosan, sustained release in vitro (up to 40%), and the integrity of RNA. Overexpressed miR-155 was observed in gills, muscle, and kidney tissues (5.42, 19.62, and 140.72-folds, respectively) after intraperitoneal delivery of miR-155-CNPs into zebrafish upon VHSV infection (miR-155-CNPs + VHSV). The miR-155-CNPs + VHSV infected fish had the highest cumulative survival (85%), which was associated with low viral copy numbers. The miR-155-overexpressing fish showed significantly decreased expression of ifnγ, irf2bpl, irf9, socs1a, il10, and caspase3, compared to that of the miR-155 inhibitor + VHSV infected fish group. In contrast, il1β, tnfα, il6, cd8a, and p53 expressions were upregulated in miR-155-overexpressed zebrafish compared to that of the control. The overall findings indicate the successful delivery of dre-miR-155 through miR-155-CNPs that enabled restriction of VHSV infection in zebrafish presumably by modulating immune gene expression.
Collapse
Affiliation(s)
- T D Liyanage
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea; Department of Microbiology and Immunology, University of Otago, 9054, Dunedin, New Zealand
| | - Chamilani Nikapitiya
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
3
|
Qin W, Saris A, van ’t Veer C, Roelofs JJTH, Scicluna BP, de Vos AF, van der Poll T. Myeloid miR-155 plays a limited role in antibacterial defense during Klebsiella-derived pneumosepsis and is dispensable for lipopolysaccharide- or Klebsiella-induced inflammation in mice. Pathog Dis 2023; 81:ftad031. [PMID: 37858304 PMCID: PMC10636497 DOI: 10.1093/femspd/ftad031] [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: 05/03/2023] [Revised: 08/02/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023] Open
Abstract
MicroRNA-155 (miR-155) plays a crucial role in regulating host inflammatory responses during bacterial infection. Previous studies have shown that constitutive miR-155 deficiency alleviates inflammation while having varying effects in different bacterial infection models. However, whether miR-155 in myeloid cells is involved in the regulation of inflammatory and antibacterial responses is largely elusive. Mice with myeloid cell specific miR-155 deficiency were generated to study the in vitro response of bone marrow-derived macrophages (BMDMs), alveolar macrophages (AMs) and peritoneal macrophages (PMs) to lipopolysaccharide (LPS), and the in vivo response after intranasal or intraperitoneal challenge with LPS or infection with Klebsiella (K.) pneumoniae via the airways. MiR-155-deficient macrophages released less inflammatory cytokines than control macrophages upon stimulation with LPS in vitro. However, the in vivo inflammatory cytokine response to LPS or K. pneumoniae was not affected by myeloid miR-155 deficiency. Moreover, bacterial outgrowth in the lungs was not altered in myeloid miR-155-deficient mice, but Klebsiella loads in the liver of these mice were significantly higher than in control mice. These data argue against a major role for myeloid miR-155 in host inflammatory responses during LPS-induced inflammation and K. pneumoniae-induced pneumosepsis but suggest that myeloid miR-155 contributes to host defense against Klebsiella infection in the liver.
Collapse
Affiliation(s)
- Wanhai Qin
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Anno Saris
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Cornelis van ’t Veer
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Brendon P Scicluna
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, MSD 2080, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080, Msida, Malta
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
4
|
Jafarzadeh A, Nemati M, Aminizadeh N, Bodhale N, Sarkar A, Jafarzadeh S, Sharifi I, Saha B. Bidirectional cytokine-microRNA control: A novel immunoregulatory framework in leishmaniasis. PLoS Pathog 2022; 18:e1010696. [PMID: 35925884 PMCID: PMC9351994 DOI: 10.1371/journal.ppat.1010696] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
As effector innate immune cells and as a host to the protozoan parasite Leishmania, macrophages play a dual role in antileishmanial immunoregulation. The 2 key players in this immunoregulation are the macrophage-expressed microRNAs (miRNAs) and the macrophage-secreted cytokines. miRNAs, as small noncoding RNAs, play vital roles in macrophage functions including cytokines and chemokines production. In the reverse direction, Leishmania-regulated cytokines alter miRNAs expression to regulate the antileishmanial functions of macrophages. The miRNA patterns vary with the time and stage of infection. The cytokine-regulated macrophage miRNAs not only help parasite elimination or persistence but also regulate cytokine production from macrophages. Based on these observations, we propose a novel immunoregulatory framework as a scientific rationale for antileishmanial therapy.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- * E-mail: (AJ); (BS)
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Najmeh Aminizadeh
- Department of Histology, School of Medicine, Islamic Azad University Branch of Kerman, Kerman
| | | | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bhaskar Saha
- National Centre For Cell Science, Pune, India
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India
- * E-mail: (AJ); (BS)
| |
Collapse
|
5
|
Crosstalk between macrophages and innate lymphoid cells (ILCs) in diseases. Int Immunopharmacol 2022; 110:108937. [PMID: 35779490 DOI: 10.1016/j.intimp.2022.108937] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022]
Abstract
Innate lymphoid cells (ILCs) and macrophages are tissue-resident cells that play important roles in tissue-immune homeostasis and immune regulation. ILCs are mainly distributed on the barrier surfaces of mammals to ensure immunity or tissue homeostasis following host, microbial, or environmental stimulation. Their complex relationships with different organs enable them to respond quickly to disturbances in environmental conditions and organ homeostasis, such as during infections and tissue damage. Gradually emerging evidence suggests that ILCs also play complex and diverse roles in macrophage development, homeostasis, polarization, inflammation, and viral infection. In turn, macrophages also determine the fate of ILCs to some extent, which indicates that network crossover between these interactions is a key determinant of the immune response. More work is needed to better define the crosstalk of ILCs with macrophages in different tissues and demonstrate how it is affected during inflammation and other diseases. Here, we summarize current research on the functional interactions between ILCs and macrophages and consider the potential therapeutic utility of these interactions for the benefit of human health.
Collapse
|
6
|
Safarzadeh M, Mohammadi-Yeganeh S, Ghorbani-Bidkorbeh F, Haji Molla Hoseini M. Chitosan based nanoformulation expressing miR-155 as a promising adjuvant to enhance Th1-biased immune responses. Life Sci 2022; 297:120459. [PMID: 35248524 DOI: 10.1016/j.lfs.2022.120459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/18/2022] [Accepted: 03/01/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND AIM MiR-155 could act as a key modulator of different aspects of immune system including Th1 responses. In this study, we designed chitosan nanoparticles containing miR-155-expressing plasmid and explored their effects as an adjuvant to enhance Th1 responses for potential future application against intracellular pathogens. METHODS Nanoparticles were formulated by complex coacervation method and characterized for physicochemical and functional characteristics. Transfection efficiency in Raw 264.7 cells, effects on miR-155 target genes and NO production were evaluated. The prepared nanoparticles were co-administered as an adjuvant with ovalbumin to immunize mice and finally production of IFN-γ and IL-4 were measured by ELISA in splenocyte recall responses. RESULTS The prepared nanoparticles had the mean size of 244 nm and zeta potential of +17 mV, respectively. Electrophoresis analysis indicated the high capability of nanoparticles to protect the plasmid from DNaseI degradation. Furthermore, nanoparticles showed an appropriate transfection efficiency in Raw 264.7 cells and could downregulate the expression of miR-155 target genes and also upregulate NO production. In vivo immunization examinations revealed successful shift of T cell responses toward Th1. CONCLUSION Our data suggests the high potential of chitosan nanoparticles containing miR-155-expressing plasmid as an adjuvant for significantly enhanced Th1-biased immune responses upon immunization with a given antigen.
Collapse
Affiliation(s)
- Mehrnoush Safarzadeh
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghorbani-Bidkorbeh
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Dhuri K, Gaddam RR, Vikram A, Slack FJ, Bahal R. Therapeutic Potential of Chemically Modified, Synthetic, Triplex Peptide Nucleic Acid-Based Oncomir Inhibitors for Cancer Therapy. Cancer Res 2021; 81:5613-5624. [PMID: 34548334 DOI: 10.1158/0008-5472.can-21-0736] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/20/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022]
Abstract
miRNA-155 (miR-155) is overexpressed in various types of lymphomas and leukemias, suggesting that targeting miR-155 could be a potential platform for the development of precision medicine. Here, we tested the anticancer activity of novel, chemically modified, triplex peptide nucleic acid (PNA)-based antimiRs compared with the current state-of-the-art conventional full-length antimiRs. Next-generation modified PNAs that bound miR-155 by Watson-Crick and Hoogsteen domains possessed superior therapeutic efficacy in vivo and ex vivo compared with conventional full-length anti-miR-155. The efficacy of anti-miR-155 targeting in multiple lymphoma cell lines was comprehensively corroborated by gene expression, Western blot analysis, and cell viability-based functional studies. Finally, preclinical testing in vivo in xenograft mouse models containing lymphoma cell lines demonstrated that treatment with the miR-155-targeting next-generation antimiR resulted in a significant decrease in miR-155 expression, followed by reduced tumor growth. These findings support the effective therapeutic application of chemically modified triplex PNAs to target miR-155 to treat lymphoma. Overall, the present proof-of-concept study further implicates the potential for next-generation triplex gamma PNAs to target other miRNAs for treating cancer. SIGNIFICANCE: This study demonstrates the utility of novel oncomiR inhibitors as cancer therapeutics, providing a new approach for targeting miRNAs and other noncoding RNAs.
Collapse
Affiliation(s)
- Karishma Dhuri
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| | - Ravinder Reddy Gaddam
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Iowa, Iowa City, Iowa
| | - Ajit Vikram
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Iowa, Iowa City, Iowa
| | - Frank J Slack
- Department of Pathology, HMS Initiative for RNA Medicine, BIDMC Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
8
|
Gao W, Yu T, Li G, Shu W, Jin Y, Zhang M, Yu X. Antioxidant Activity and Anti-Apoptotic Effect of the Small Molecule Procyanidin B1 in Early Mouse Embryonic Development Produced by Somatic Cell Nuclear Transfer. Molecules 2021; 26:molecules26206150. [PMID: 34684730 PMCID: PMC8540974 DOI: 10.3390/molecules26206150] [Citation(s) in RCA: 9] [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: 08/30/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 12/22/2022] Open
Abstract
As an antioxidant, procyanidin B1(PB1) can improve the development of somatic cell nuclear transfer (SCNT) embryos; PB1 reduces the level of oxidative stress (OS) during the in vitro development of SCNT embryos by decreasing the level of reactive oxygen species (ROS) and increasing the level of glutathione (GSH) and mitochondrial membrane potential (MMP). Metabolite hydrogen peroxide (H2O2) produces OS. Catalase (CAT) can degrade hydrogen peroxide so that it produces less toxic water (H2O) and oxygen (O2) in order to reduce the harm caused by H2O2. Therefore, we tested the CAT level in the in vitro development of SCNT embryos; it was found that PB1 can increase the expression of CAT, indicating that PB1 can offset the harm caused by oxidative stress by increasing the level of CAT. Moreover, if H2O2 accumulates excessively, it produces radical-(HO-) through Fe2+/3+ and damage to DNA. The damage caused to the DNA is mainly repaired by the protein encoded by the DNA damage repair gene. Therefore, we tested the expression of the DNA damage repair gene, OGG1. It was found that PB1 can increase the expression of OGG1 and increase the expression of protein. Through the above test, we proved that PB1 can improve the repairability of DNA damage. DNA damage can lead to cell apoptosis; therefore, we also tested the level of apoptosis of blastocysts, and we found that PB1 reduced the level of apoptosis. In summary, our results show that PB1 reduces the accumulation of H2O2 by decreasing the level of OS during the in vitro development of SCNT embryos and improves the repairability of DNA damage to reduce cell apoptosis. Our results have important significance for the improvement of the development of SCNT embryos in vitro and provide important reference significance for diseases that can be treated using SCNT technology.
Collapse
Affiliation(s)
- Wei Gao
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China; (W.G.); (Y.J.); (M.Z.)
- Group of Non-Human Primates of Reproductive and Stem Cell, Kunming Institute of Zoology, CAS, Kunming 650203, China; (T.Y.); (G.L.); (W.S.)
| | - Tingting Yu
- Group of Non-Human Primates of Reproductive and Stem Cell, Kunming Institute of Zoology, CAS, Kunming 650203, China; (T.Y.); (G.L.); (W.S.)
| | - Guomeng Li
- Group of Non-Human Primates of Reproductive and Stem Cell, Kunming Institute of Zoology, CAS, Kunming 650203, China; (T.Y.); (G.L.); (W.S.)
| | - Wei Shu
- Group of Non-Human Primates of Reproductive and Stem Cell, Kunming Institute of Zoology, CAS, Kunming 650203, China; (T.Y.); (G.L.); (W.S.)
| | - Yongxun Jin
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China; (W.G.); (Y.J.); (M.Z.)
| | - Mingjun Zhang
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China; (W.G.); (Y.J.); (M.Z.)
| | - Xianfeng Yu
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China; (W.G.); (Y.J.); (M.Z.)
- Correspondence: ; Tel.: +86-431-8783-6536
| |
Collapse
|
9
|
Guo J, Liao M, Wang J. TLR4 signaling in the development of colitis-associated cancer and its possible interplay with microRNA-155. Cell Commun Signal 2021; 19:90. [PMID: 34479599 PMCID: PMC8414775 DOI: 10.1186/s12964-021-00771-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/29/2021] [Indexed: 12/17/2022] Open
Abstract
Ulcerative colitis (UC) has closely been associated with an increased risk of colorectal cancer. However, the exact mechanisms underlying colitis-associated cancer (CAC) development remain unclear. As a classic pattern-recognition receptor, Toll like receptor (TLR)4 is a canonical receptor for lipopolysaccharide of Gram-negative bacteria (including two CAC-associated pathogens Fusobacterium nucleatum and Salmonella), and functions as a key bridge molecule linking oncogenic infection to colonic inflammatory and malignant processes. Accumulating studies verified the overexpression of TLR4 in colitis and CAC, and the over-expressed TLR4 might promote colitis-associated tumorigenesis via facilitating cell proliferation, protecting malignant cells against apoptosis, accelerating invasion and metastasis, as well as contributing to the creation of tumor-favouring cellular microenvironment. In recent years, considerable attention has been focused on the regulation of TLR4 signaling in the context of colitis-associated tumorigenesis. MicroRNA (miR)-155 and TLR4 exhibited a similar dynamic expression change during CAC development and shared similar CAC-promoting properties. The available data demonstrated an interplay between TLR4 and miR-155 in the context of different disorders or cell lines. miR-155 could augment TLR4 signaling through targeting negative regulators SOCS1 and SHIP1; and TLR4 activation would induce miR-155 expression via transcriptional and post-transcriptional mechanisms. This possible TLR4-miR-155 positive feedback loop might result in the synergistic accelerating effect of TLR4 and miR-155 on CAC development.![]() Video abstract
Collapse
Affiliation(s)
- Jie Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China.,New Medicine Innovation and Development Institute, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mengfan Liao
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China.,New Medicine Innovation and Development Institute, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jun Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China. .,New Medicine Innovation and Development Institute, Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
The Role of miR-155 in Nutrition: Modulating Cancer-Associated Inflammation. Nutrients 2021; 13:nu13072245. [PMID: 34210046 PMCID: PMC8308226 DOI: 10.3390/nu13072245] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/08/2023] Open
Abstract
Nutrition plays an important role in overall human health. Although there is no direct evidence supporting the direct involvement of nutrition in curing disease, for some diseases, good nutrition contributes to disease prevention and our overall well-being, including energy level, optimum internal function, and strength of the immune system. Lately, other major, but more silent players are reported to participate in the body’s response to ingested nutrients, as they are involved in different physiological and pathological processes. Furthermore, the genetic profile of an individual is highly critical in regulating these processes and their interactions. In particular, miR-155, a non-coding microRNA, is reported to be highly correlated with such nutritional processes. In fact, miR-155 is involved in the orchestration of various biological processes such as cellular signaling, immune regulation, metabolism, nutritional responses, inflammation, and carcinogenesis. Thus, this review aims to highlight those critical aspects of the influence of dietary components on gene expression, primarily on miR-155 and its role in modulating cancer-associated processes.
Collapse
|
11
|
Gao Y, Han T, Han C, Sun H, Yang X, Zhang D, Ni X. Propofol Regulates the TLR4/NF-κB Pathway Through miRNA-155 to Protect Colorectal Cancer Intestinal Barrier. Inflammation 2021; 44:2078-2090. [PMID: 34081253 DOI: 10.1007/s10753-021-01485-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 12/22/2022]
Abstract
Surgery for colorectal cancer (CRC) can cause damage to the intestinal mucosal barrier and lead to bacterial invasion. This study mainly analyzed whether propofol (PPF) could protect the intestinal mucosal barrier damage caused by CRC surgery, and explored its molecular mechanism. A mouse CRC model was constructed using azomethane and dextran sulfate sodium. During anesthesia, continuous intravenous injection of PPF was used for intervention. The influences of PPF on intestinal mucosal permeability and bacterial invasion were detected. The levels of microRNA (miR)-155, Toll-like receptor 4 (TLR4)/NF-κB in the intestinal mucosa, and the location of miR-155 were detected by fluorescence in situ hybridization (FISH). Mouse macrophages were used to analyze the regulation of miR-155 on the secretion of inflammatory cytokines through the TLR4/NF-κB pathway. PPF treatment promoted the expression of tight junction protein in the intestinal mucosa, protected the intestinal barrier, inhibited the translocation of intestinal bacteria, and increased the level of the beneficial bacterium Lactobacillus on the mucosal surface. In addition, PPF treatment could inhibit the expression of miR-155, TLR4/NF-KB, and reverse inflammatory response. miR-155 was expressed in macrophages of intestinal mucosa tissue. Overexpression of miR-155 promoted the nuclear translocation of NF-κB and the expression of inflammatory cytokines in macrophages. The use of VIPER to inhibit TLR4 reversed the pro-inflammatory effects of miR-155. PPF might inhibit the activation of the NF-κB pathway by downregulating miR-155 expression, thereby reducing the secretion of inflammatory cytokines. This might be the mechanism by which PPF protected the intestinal barrier of CRC surgical model mice.
Collapse
Affiliation(s)
- Yuhua Gao
- Department of Anesthesiology, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China.,School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Tao Han
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Cailing Han
- Department of Anesthesiology, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Hua Sun
- Department of Anesthesiology, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Xiaoxia Yang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Dongmei Zhang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Xinli Ni
- Department of Anesthesiology, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
12
|
Gholamrezaei M, Rouhani S, Mohebali M, Mohammadi-Yeganeh S, Haji Molla Hoseini M, Haghighi A, Lasjerdi Z, Hamidi F, Kazem Sharifi-Yazdi M. MicroRNAs Expression Induces Apoptosis of Macrophages in Response to Leishmania major (MRHO/IR/75/ER): An In-Vitro and In-Vivo Study. IRANIAN JOURNAL OF PARASITOLOGY 2020; 15:475-487. [PMID: 33884004 PMCID: PMC8039482 DOI: 10.18502/ijpa.v15i4.4851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background: We aimed to investigate the effect of miR-15a mimic and inhibitor of miR-155 expression on apoptosis induction in macrophages infected with Iranian strain of Leishmania major in-vitro and in-vivo. Methods: RAW 264.7 cells were infected with L. major promastigotes (MRHO/IR/75/ER), and then were treated with miRNAs. For in-vivo experiment, BALB/c mice were inoculated with L. major promastigotes, and then they were treated with miRNAs. For evaluation of miRNA therapeutic effect, in-vitro and in-vivo studies were performed using quantitative Real-time PCR, Flow cytometry, lesion size measurement, and Limiting Dilution Assay (LDA). This study was performed in Shahid Beheshti University of Medical Sciences in 2019. Results: In-vitro results of flow cytometry showed that using miR-15a mimic, miR-155 inhibitor or both of them increased apoptosis of macrophages. In in-vivo, size of lesion increased during experiment in control groups (P<0.05) while application of both miR-155 inhibitor and miR-15a mimic inhibited the increase in the size of lesions within 6 wk of experiment (P=0.85). LDA results showed that microRNA therapy could significantly decrease parasite load in mimic or inhibitor receiving groups compared to the control group (P<0.05). Conclusion: miR-155 inhibitor and miR-15a mimic in L. major infected macrophages can induce apoptosis and reduce parasite burden. Therefore, miRNA-based therapy can be proposed as new treatment for cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Mostafa Gholamrezaei
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Rouhani
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Center for Research of Endemic Parasites of Iran (CREPI), Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences,Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Haghighi
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Lasjerdi
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Hamidi
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
13
|
Lasjerdi Z, Ghanbarian H, Mohammadi Yeganeh S, Seyyed Tabaei SJ, Mohebali M, Taghipour N, Koochaki A, Hamidi F, Gholamrezaei M, Haghighi A. Comparative Expression Profile Analysis of Apoptosis-Related miRNA and Its Target Gene in Leishmania major Infected Macrophages. IRANIAN JOURNAL OF PARASITOLOGY 2020; 15:332-340. [PMID: 33082797 PMCID: PMC7548466 DOI: 10.18502/ijpa.v15i3.4197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background: Cutaneous Leishmaniasis (CL) is an emerging uncontrollable and neglected infectious disease worldwide including Iran. The aim of this study was to investigate the expression profile of apoptosis-related miRNA and its target gene in macrophages. Methods: This study was carried out in the Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran from January 2016 to November 2018. Applying literature reviews, bioinformatics software, and microarray expression analysis, we selected miRNA-24-3p interfering in apoptosis pathway. The expression profile of this miRNA and target gene were investigated in Leishmania major (MRHO/IR/75/ER)-infected primary and RAW 264.7 macrophages (IBRC-C10072) compared with non-infected macrophages (control group) using quantitative Real-time PCR. Results: Results of bioinformatics analysis showed that miR-24-3p as anti-apoptotic miRNA inhibits pro-apoptotic genes (Caspases 3 and 7). Microarray expression data presented in Gene Expression Omnibus (GEO) revealed a significant difference in the expression level of selected miRNA and its target gene between two groups. QRT-PCR results showed that the expression of miR-24-3p was upregulated in L. major infectioned macrophages that approved the results of bioinformatics and microarray analysis. Conclusion: Parasite can alter miRNAs expression pattern in the host cells to establish infection and its survival. Alteration in miRNAs levels likely plays an important role in regulating macrophage functions following L. major infection. These results could highlight current understanding and new insights concerning the gene expression in macrophages during leishmaniasis and will help to development of novel strategies for control and treatment of CL.
Collapse
Affiliation(s)
- Zohreh Lasjerdi
- Department of Medical Parasitology and Mycology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi Yeganeh
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Javad Seyyed Tabaei
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences Tehran, Iran
| | - Niloofar Taghipour
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ameneh Koochaki
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Hamidi
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Gholamrezaei
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Haghighi
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
MicroRNA-155 Participates in Smoke-Inhalation-Induced Acute Lung Injury through Inhibition of SOCS-1. Molecules 2020; 25:molecules25051022. [PMID: 32106541 PMCID: PMC7179228 DOI: 10.3390/molecules25051022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/18/2020] [Accepted: 02/23/2020] [Indexed: 12/11/2022] Open
Abstract
Smoke inhalation causes acute lung injury (ALI), a severe clinical disease with high mortality. Accumulating evidence indicates that microRNA-155 (miR-155) and suppressor of cytokine signaling 1 (SOCS-1), as mediators of inflammatory response, are involved in the pathogenesis of ALI. In this paper, we explored the proinflammatory mechanism of miR-155 in smoke-inhalation-induced ALI. Our data revealed that smoke inhalation induces miR-155 expression, and miR-155 knockout (KO) significantly ameliorates smoke-inhalation-induced lung injury in mice. Neutrophil infiltration and myeloperoxidase (MPO), macrophage inflammatory protein 2 (MIP-2) and keratinocyte chemoattractant (KC) expressions were decreased in miR-155–/– mice after smoke inhalation as well. Real-time RT-PCR and immunoblotting results showed that SOCS-1 level was remarkably increased in miR-155–/– mice after smoke exposure. Furthermore, the experiments performed in isolated miR-155 KO pulmonary neutrophils demonstrated that the lack of SOCS-1 enhanced inflammatory cytokines (MIP-2 and KC) secretion in response to smoke stimulation. In conclusion, smoke induces increased expression of miR-155, and miR-155 is involved in inflammatory response to smoke-inhalation-induced lung injury by inhibiting the expression of SOCS-1.
Collapse
|
15
|
Gao C, Dang S, Zhai J, Zheng S. Regulatory mechanism of microRNA-155 in chicken embryo fibroblasts in response to reticuloendotheliosis virus infection. Vet Microbiol 2020; 242:108610. [PMID: 32122614 DOI: 10.1016/j.vetmic.2020.108610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/09/2020] [Accepted: 02/09/2020] [Indexed: 01/17/2023]
Abstract
Reticuloendotheliosis virus (REV) infection of multiple avian species can lead to a number of diseases such as runting syndrome, immunosuppression and oncogenesis, causing major economic losses. MicroRNAs play important roles in post-transcriptional regulation, effectively inhibiting protein synthesis, and participating in many biological processes in cells, including proliferation, differentiation, apoptosis, lipometabolism, virus infection and replication, and tumorigenesis. Based on our previous high-throughput sequencing results, we explore the regulatory mechanisms of microRNA-155(miR-155) in chicken embryo fibroblasts (CEFs) in response to REV infection. Our results revealed expression of miR-155 in CEFs after REV infection upregulated in a time- and dose-dependent manner, indicating miR-155 plays a role in REV infection in CEFs indeed. After transfected with miR-155-mimic and miR-155-inhibitor, we found overexpression of miR-155 targeted caspase-6 and FOXO3a to inhibit apoptosis and accelerate cell cycle, thus improving viability of REV-infected CEFs. This result also verified the protective role of miR-155 in the viability of CEFs in the presence of REV. Knockdown of miR-155 also supported these above conclusions. Our findings uncover a new mechanism of REV pathogenesis in CEFs, and also provide a theoretical basis for uncovering new effective treatment and prevention methods for RE based on miR-155.
Collapse
Affiliation(s)
- Chang Gao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, NO. 59 Mucai Street, Harbin 150030, People's Republic of China.
| | - Shengyuan Dang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, NO. 59 Mucai Street, Harbin 150030, People's Republic of China.
| | - Jie Zhai
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, NO. 59 Mucai Street, Harbin 150030, People's Republic of China.
| | - Shimin Zheng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, NO. 59 Mucai Street, Harbin 150030, People's Republic of China.
| |
Collapse
|
16
|
Huang R, Cao Y, Li H, Hu Z, Zhang H, Zhang L, Su W, Xu Y, Liang L, Melgiri ND, Jiang L, Li X. miR-532-3p-CSF2RA Axis as a Key Regulator of Vulnerable Atherosclerotic Plaque Formation. Can J Cardiol 2019; 36:1782-1794. [PMID: 32473103 DOI: 10.1016/j.cjca.2019.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The most dangerous atherosclerotic plaques, referred to as "vulnerable," are most likely to trigger acute atherothrombotic events such as myocardial infarction (heart attack) and stroke. Our goal was to uncover the molecular drivers of vulnerable plaque formation. METHODS To elucidate the functional gene modules that drive vulnerable plaque formation, we performed a weighted gene coexpression network analysis integrated with a protein-protein interaction network analysis in human atherosclerotic carotid samples, which identified the candidate gene granulocyte-macrophage colony-stimulating factor 2 (GM-CSF) receptor alpha subunit (CSF2RA). Follow-up in vitro experiments were performed to elucidate the regulatory relationship between CSF2RA and the microRNA miR-532-3p as well as modifiers of macrophagic miR-532-3p-CSF2RA axis expression. Microarray and quantitative reverse transcription polymerase chain reaction (qRT-PCR) studies elucidated the effect of statins on carotid miR-532-3p-CSF2RA axis expression in patients with carotid atherosclerotic disease. Apoe-/-, Ldlr-/-, and Csf2ra mutant Apoe-/- mouse models of atherosclerosis were employed to assess the effects of agomiR-532-3p therapy in vivo. RESULTS The integrated weighted gene coexpression network analysis/protein-protein interaction network analysis revealed that the macrophagic GM-CSF receptor CSF2RA is significantly upregulated in macrophage-rich vulnerable plaques. Follow-up analysis identified the miR-532-3p-CSF2RA axis, as miR-532-3p downregulates CSF2RA via binding to CSF2RA's 3'UTR. Macrophagic miR-532-3p-CSF2RA dysregulation was enhanced via modified low-density lipoprotein or tumor necrosis factor α exposure in vitro. Moreover, this miR-532-3p-CSF2RA dysregulation was observed in human vulnerable plaques and Apoe-/- mouse plaques, effects rescued by statin therapy. In vivo, agomiR-532-3p therapy suppressed murine plaque formation and promoted plaque stabilization in a Csf2ra-dependent manner. CONCLUSION Macrophagic miR-532-3p-CSF2RA axis dysregulation is a key driver in vulnerable plaque formation.
Collapse
Affiliation(s)
- Rongzhong Huang
- Department of Gerontology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Cao
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Hongrong Li
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zicheng Hu
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Hong Zhang
- Department of Cardiology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Lujun Zhang
- Statistical Laboratory, Chuangxu Institute of Life Science, Chongqing, China; Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenhua Su
- Department of Cardiology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yu Xu
- Statistical Laboratory, Chuangxu Institute of Life Science, Chongqing, China
| | - Liwen Liang
- Department of Cardiology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Narayan D Melgiri
- Impactys Foundation for Biomedical Research, San Diego, California, USA
| | - Lihong Jiang
- Department of Cardiothoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Xingsheng Li
- Department of Gerontology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
17
|
Zhang B, Li B, Qin F, Bai F, Sun C, Liu Q. Expression of serum microRNA-155 and its clinical importance in patients with heart failure after myocardial infarction. J Int Med Res 2019; 47:6294-6302. [PMID: 31709859 PMCID: PMC7045684 DOI: 10.1177/0300060519882583] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Objective This study was conducted to investigate the level of microRNA-155 (miRNA-155) in patients with heart failure (HF) after myocardial infarction (MI) and its clinical importance. Methods Serum miRNA-155 levels were measured using quantitative reverse transcription (qRT)-PCR. The left ventricular ejection fraction (LVEF), left ventricular posterior wall thickness, and left ventricular end-diastolic diameter were measured by echocardiography. Serum amino-terminal pro-B-type natriuretic peptide (NT-proBNP) and other parameters were also analyzed. Results miRNA-155 levels in patients with HF were significantly higher than in control and MI groups. The area under the receiver operating characteristic curve of serum miRNA-155 in the diagnosis of HF after MI was 0.941, the cutoff value was 1.77, sensitivity was 92.73%, and specificity was 92.14%. NT-proBNP levels were significantly higher and LVEF was lower in patients with high versus low miRNA-155 expression. Conclusions Patients with HF after MI had elevated miRNA-155 levels and poor cardiac function, suggesting that determining miRNA-155 expression could be used to assess the severity of the disease.
Collapse
Affiliation(s)
- Baojian Zhang
- Cardiac Catheterization Laboratory, Department of Cardiology,
the Second Xiangya Hospital, Central South University, Changsha, China
- Coronary Care Unit, the Affiliated Hospital of Traditional
Chinese Medicine, Xinjiang Medical University, Urumqi, China
| | - Biao Li
- Cardiac Catheterization Laboratory, Department of Cardiology,
the Second Xiangya Hospital, Central South University, Changsha, China
| | - Fen Qin
- Cardiac Catheterization Laboratory, Department of Cardiology,
the Second Xiangya Hospital, Central South University, Changsha, China
| | - Fan Bai
- Cardiac Catheterization Laboratory, Department of Cardiology,
the Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Sun
- Cardiac Catheterization Laboratory, Department of Cardiology,
the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiming Liu
- Cardiac Catheterization Laboratory, Department of Cardiology,
the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Sui C, Zhang L, Hu Y. MicroRNA‑let‑7a inhibition inhibits LPS‑induced inflammatory injury of chondrocytes by targeting IL6R. Mol Med Rep 2019; 20:2633-2640. [PMID: 31322277 PMCID: PMC6691277 DOI: 10.3892/mmr.2019.10493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) is a type of degenerative joint disease that affects the health of the elderly. OA is characterized by articular cartilage degradation and joint inflammation. The present study aimed to investigate the role and mechanism of microRNA-let-7a (Let-7a) in OA by examining its role in lipopolysaccharide (LPS)-induced cartilage inflammatory injury in ATDC5 cells. ATDC5 cells were treated with various concentrations of LPS. The present results suggested that 5 and 10 µg/ml LPS significantly inhibited ATDC5 cell viability, and 5 µg/ml LPS was selected for further experiments. Reverse transcription-quantitative PCR (RT-qPCR) results suggested that treatment with LPS significantly induced the expression levels of multiple inflammatory factors, including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6 and IL-8, and increased the expression level of Let-7a in ATDC5 cells. IL-6 receptor (IL-6R) was identified to be a direct target of Let-7a using TargetScan and a dual-luciferase reporter assay. Subsequently, Cell Counting Kit-8 and flow cytometry analyses identified that Let-7a inhibitor could significantly promote cell viability and reduce cell apoptosis in ATDC5 cells treated with LPS, and these effects could be reversed by transfection with small interfering (si)RNA-IL-6R. ELISA was used to examine the expression of inflammatory factors in ATDC5 cells following treatment with LPS. Additionally, RT-qPCR and western blotting were performed to detect the mRNA and protein expression level of IL-6R and STAT3. The present results suggested that Let-7a inhibitor significantly reduced the expression level of TNF-α, IL-1β, IL-6 and IL-8 in ATDC5 cells, and this effect was reversed by transfecting siRNA-IL-6R. Moreover, RT-qPCR and western blot assay results suggested that Let-7a inhibitor significantly increased the expression level of IL-6R and phosphorylated STAT3, and these effects could be reversed by siRNA-IL-6R. Collectively, Let-7a inhibitor increased cell proliferation, reduced apoptosis and inhibited inflammatory response in ATDC5 cells treated with LPS. The present study provided a new potential therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Cong Sui
- Department of Orthopaedics (Trauma Orthopaedics Ward), The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Lecheng Zhang
- Department of Orthopaedics (Trauma Orthopaedics Ward), The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yong Hu
- Department of Orthopaedics (Trauma Orthopaedics Ward), The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
19
|
Milani R, Brognara E, Fabbri E, Manicardi A, Corradini R, Finotti A, Gasparello J, Borgatti M, Cosenza LC, Lampronti I, Dechecchi MC, Cabrini G, Gambari R. Targeting miR‑155‑5p and miR‑221‑3p by peptide nucleic acids induces caspase‑3 activation and apoptosis in temozolomide‑resistant T98G glioma cells. Int J Oncol 2019; 55:59-68. [PMID: 31180529 PMCID: PMC6561624 DOI: 10.3892/ijo.2019.4810] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/26/2019] [Indexed: 12/11/2022] Open
Abstract
The present study investigated the effects of the combined treatment of two peptide nucleic acids (PNAs), directed against microRNAs involved in caspase‑3 mRNA regulation (miR‑155‑5p and miR‑221‑3p) in the temozolomide (TMZ)‑resistant T98G glioma cell line. These PNAs were conjugated with an octaarginine tail in order to obtain an efficient delivery to treated cells. The effects of singularly administered PNAs or a combined treatment with both PNAs were examined on apoptosis, with the aim to determine whether reversion of the drug‑resistance phenotype was obtained. Specificity of the PNA‑mediated effects was analyzed by reverse transcription‑quantitative polymerase‑chain reaction, which demonstrated that the effects of R8‑PNA‑a155 and R8-PNA-a221 anti‑miR PNAs were specific. Furthermore, the results obtained confirmed that both PNAs induced apoptosis when used on the temozolomide‑resistant T98G glioma cell line. Notably, co‑administration of both anti‑miR‑155 and anti‑miR‑221 PNAs was associated with an increased proapoptotic activity. In addition, TMZ further increased the induction of apoptosis in T98G cells co‑treated with anti‑miR‑155 and anti‑miR‑221 PNAs.
Collapse
Affiliation(s)
- Roberta Milani
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Eleonora Brognara
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Enrica Fabbri
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Alex Manicardi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, I‑143214 Parma, Italy
| | - Roberto Corradini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, I‑143214 Parma, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | - Ilaria Lampronti
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| | | | - Giulio Cabrini
- Laboratory of Molecular Pathology, University‑Hospital of Verona, I‑37126 Verona, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I‑144121 Ferrara, Italy
| |
Collapse
|
20
|
Bayraktar R, Bertilaccio MTS, Calin GA. The Interaction Between Two Worlds: MicroRNAs and Toll-Like Receptors. Front Immunol 2019; 10:1053. [PMID: 31139186 PMCID: PMC6527596 DOI: 10.3389/fimmu.2019.01053] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are critical mediators of posttranscriptional regulation via their targeting of the imperfect antisense complementary regions of coding and non-coding transcripts. Recently, researchers have shown that miRNAs play roles in many aspects of regulation of immune cell function by targeting of inflammation-associated genes, including Toll-like receptors (TLRs). Besides this indirect regulatory role of miRNAs, they can also act as physiological ligands of specific TLRs and initiate the signaling cascade of immune response. In this review, we summarize the potential roles of miRNAs in regulation of TLR gene expression and TLR signaling, with a focus on the ability of miRNAs bind to TLRs.
Collapse
Affiliation(s)
- Recep Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
21
|
Guo J, Liu HB, Sun C, Yan XQ, Hu J, Yu J, Yuan Y, Du ZM. MicroRNA-155 Promotes Myocardial Infarction-Induced Apoptosis by Targeting RNA-Binding Protein QKI. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4579806. [PMID: 31191799 PMCID: PMC6525929 DOI: 10.1155/2019/4579806] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/12/2019] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
Abstract
Acute myocardial infarction (AMI) is the leading cause of sudden death worldwide. MicroRNA-155 (miR-155) has been reported to target antiapoptotic genes in various diseases models, but the functional role of miR-155 in response to MI injury needs further investigations. This study investigated the role of miR-155 in myocardial ischemia injury. TUNEL and flow cytometry were performed to measure cell apoptosis. Western blot analysis was employed to detect protein expressions of Bcl-2, XIAP, Bax, and caspase-3. qRT-PCR was used to quantify miRNA levels. We showed that miR-155 was dynamically elevated in murine hearts subjected to MI and in neonatal rat ventricular cardiomyocyte (NRVM) injury induced by hydrogen peroxide (H2O2). In response to H2O2, the silencing of miR-155 using AMO-155 (antisense inhibitor oligodeoxyribonucleotides) significantly increased cell viability and reduced cell apoptosis. Moreover, AMO-155 reversed the H2O2-induced downregulation of Bcl-2 and XIAP and upregulation of Bax and cleaved-caspase-3. Further study revealed that AMO-155 resulted in a decrease of H2O2-induced JC-1-labelled monomeric cell number. In addition, AMO-155 markedly decreased infarct size, ameliorated impaired cardiac function, and significantly reduced apoptotic cell percentages in MI mice heart. The RNA-binding protein Quaking (QKI) was predicted as a target gene of miR-155 through bioinformatic analysis, and AMO-155 attenuated the downregulation of QKI in H2O2-treated cardiomyocytes and MI mice heart. Knockdown of QKI by siRNA abolished the antiapoptotic effects of AMO-155. Taken together, miR-155 is upregulated in the MI heart and NRVMs in response to H2O2 stress, and downregulating of miR-155 protects cardiomyocytes against apoptosis. Mechanistically, it is probably due to the repression of QKI signaling pathway.
Collapse
Affiliation(s)
- Jing Guo
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Hui-Bin Liu
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Chuan Sun
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Xiu-Qing Yan
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Juan Hu
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Jie Yu
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Ye Yuan
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
| | - Zhi-Min Du
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, China
- Department of Clinical Pharmarcology, College of Pharmacy, Harbin Medical University, Harbin 150086, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
22
|
Lin J, Ma L, Zhang D, Gao J, Jin Y, Han Z, Lin D. Tumour biomarkers-Tracing the molecular function and clinical implication. Cell Prolif 2019; 52:e12589. [PMID: 30873683 PMCID: PMC6536410 DOI: 10.1111/cpr.12589] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/19/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022] Open
Abstract
In recent years, with the increase in cancer mortality caused by metastasis, and with the development of individualized and precise medical treatment, early diagnosis with precision becomes the key to decrease the death rate. Since detecting tumour biomarkers in body fluids is the most non‐invasive way to identify the status of tumour development, it has been widely investigated for the usage in clinic. These biomarkers include different expression or mutation in microRNAs (miRNAs), circulating tumour DNAs (ctDNAs), proteins, exosomes and circulating tumour cells (CTCs). In the present article, we summarized and discussed some updated research on these biomarkers. We overviewed their biological functions and evaluated their multiple roles in human and small animal clinical treatment, including diagnosis of cancers, classification of cancers, prognostic and predictive values for therapy response, monitors for therapy efficacy, and anti‐cancer therapeutics. Biomarkers including different expression or mutation in miRNAs, ctDNAs, proteins, exosomes and CTCs provide more choice for early diagnosis of tumour detection at early stage before metastasis. Combination detection of these tumour biomarkers may provide higher accuracy at the lowest molecule combination number for tumour early detection. Moreover, tumour biomarkers can provide valuable suggestions for clinical anti‐cancer treatment and execute monitoring of treatment efficiency.
Collapse
Affiliation(s)
- Jiahao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lie Ma
- Department of Respiratory Disease, The Navy General Hospital of PLA, Beijing, China
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiafeng Gao
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yipeng Jin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhihai Han
- Department of Respiratory Disease, The Navy General Hospital of PLA, Beijing, China
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
23
|
Nariman-Saleh-Fam Z, Saadatian Z, Daraei A, Mansoori Y, Bastami M, Tavakkoli-Bazzaz J. The intricate role of miR-155 in carcinogenesis: potential implications for esophageal cancer research. Biomark Med 2019; 13:147-159. [PMID: 30672305 DOI: 10.2217/bmm-2018-0127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
MiRNAs have immerged as essential modulators of key cellular procuresses involved in post-transcriptional regulation of the human transcriptome. They are essential components of complex regulatory networks that modulate most important physiological functions of cells. MicroRNA-155 (miR-155) is a multifaceted regulator of cell proliferation, cell cycle, development, immunity and inflammation that plays pivotal, and sometimes contradictory, roles in numerous cancers including esophageal cancer. Here, we review the intricate role of miR-155 in cancer by exemplifying carcinogenesis of various tumors, focusing on recent findings that may provide a link between miR-155 and esophageal cancer-related pathways.
Collapse
Affiliation(s)
- Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Saadatian
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Daraei
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Yaser Mansoori
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Milad Bastami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Tavakkoli-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Li M, Cui J, Niu W, Huang J, Feng T, Sun B, Yao H. Long non-coding PCED1B-AS1 regulates macrophage apoptosis and autophagy by sponging miR-155 in active tuberculosis. Biochem Biophys Res Commun 2019; 509:803-809. [PMID: 30621915 DOI: 10.1016/j.bbrc.2019.01.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/02/2019] [Indexed: 11/25/2022]
Abstract
Macrophages play a major role in the control and elimination of invading Mycobacterium tuberculosis (Mtb). Emerging studies have demonstrated that long non-coding RNAs (lncRNAs) are involved in resident macrophages in Mtb. However, the regulatory mechanism between lncRNAs and macrophages in tuberculosis (TB) remains unclear. In this study, we sought to investigate the effect of Mtb-associated lncRNA PCED1B-AS1 on macrophage apoptosis and autophagy. Our study first evaluated PCED1B-AS1 expression in the CD14+ monocytes from patients with active tuberculosis and from healthy individuals. It was found that PCED1B-AS1 expression was down-regulated in patients with active tuberculosis, accompanied by significant attenuated monocyte apoptosis and enhanced autophagy. In vitro, knockdown of PCED1B-AS1 reduced macrophage apoptosis and promoted autophagy. PCED1B-AS1 serves as an endogenous sponge to block miR-155 expression in macrophages by directly binding to miR-155. Furthermore, we demonstrated that overexpression of FOXO3/Rheb, target genes of miR-155, reversed the PCED1B-AS1-mediated effects on macrophage apoptosis and autophagy. Altogether, our data indicate that PCED1B-AS1 modulates macrophage apoptosis and autophagy by targeting the miR-155 axis in active TB.
Collapse
Affiliation(s)
- Mingying Li
- Fourth Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China
| | - Junwei Cui
- First Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China
| | - Wenyi Niu
- Fourth Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China
| | - Jian Huang
- Second Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China
| | - Tianjuan Feng
- Fourth Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China
| | - Bing Sun
- Second Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China
| | - Hengbo Yao
- Fourth Tuberculosis Internal Medicine Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China.
| |
Collapse
|
25
|
Chen H, Liu Gao MY, Zhang L, He FL, Shi YK, Pan XH, Wang H. MicroRNA-155 affects oxidative damage through regulating autophagy in endothelial cells. Oncol Lett 2018; 17:2237-2243. [PMID: 30675289 PMCID: PMC6341854 DOI: 10.3892/ol.2018.9860] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
MicroRNA-155 (miRNA-155) is a typical multifunctional miRNA, which serves a crucial role in the regulation of numerous vessel cells. However, its effects on dysfunctional endothelial cells have not been completely elucidated. In order to investigate the signaling pathway of miRNA-155-induced cell injury, H2O2 was used to establish an oxidative stress cell model, and miR-155 was transfected into H2O2-treated cells. The CCK8 assay was then employed to examine the effect of miR-155 on the cell proliferations of H2O2-treated cells, and the expressions of Microtubule Associated Protein 1 Light Chain 3 (LC3) and Sequestosome 1 (P62) were detected to examine the effect of miR-155 on the autophagy of Human umbilical vein endothelial cells, and then the formation of intracellular autophagosomes was observed. The results indicated that endothelial cell proliferation was promoted, and oxidant-induced injury was decreased when the expression of miR-155 was inhibited. In addition, the results also demonstrated that when the miR-155 inhibitor was used, the expression of LC3 was increased and the expression of P62 was decreased. This suggests that modulated miR-155 can prevent oxidative damage in endothelial cells, by regulating the level of autophagy. Furthermore, the present study also demonstrated that miR-155 regulated autophagy via promotion of the expression of the autophagy-related gene, Autophagy Related 5 (ATG5). In conclusion, the attenuated expression of miR-155 can decrease oxidant-induced injury and promote cell proliferation via upregulating autophagy, which subsequently affects the expression of ATG5. The present study provides a novel insight into microRNAs as potential therapeutics for the treatment of heart disease.
Collapse
Affiliation(s)
- Huifen Chen
- Department of Cardiology, Hengyang Central Hospital, Hengyang, Hunan 421000, P.R. China
| | - Mi Yang Liu Gao
- Department of Clinical Experiment, Kunming General Hospital, Kunming, Yunnan 650032, P.R. China
| | - Li Zhang
- Department of Geriatrics, Kunming General Hospital, Kunming, Yunnan 650032, P.R. China
| | - Fa Lian He
- Department of Clinical Experiment, Kunming General Hospital, Kunming, Yunnan 650032, P.R. China
| | - Yan Kun Shi
- Department of Geriatrics, Kunming General Hospital, Kunming, Yunnan 650032, P.R. China
| | - Xing Hua Pan
- Department of Clinical Experiment, Kunming General Hospital, Kunming, Yunnan 650032, P.R. China
| | - Hong Wang
- Department of Geriatrics, Kunming General Hospital, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
26
|
Ro Y, Jo G, Jung S, Lee E, Shin J, Lee J. Salmonella‑induced miR‑155 enhances necroptotic death in macrophage cells via targeting RIP1/3. Mol Med Rep 2018; 18:5133-5140. [DOI: 10.3892/mmr.2018.9525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/21/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Young‑Tae Ro
- Laboratory of Biochemistry, Graduate School of Medicine, Konkuk University, Chungju, Chungcheong 27478, Republic of Korea
| | - Guk‑Heui Jo
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| | - Sun‑Ah Jung
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| | - Eunjoo Lee
- Graduate School of East‑West Medical Science, Kyung Hee University, Yongin, Gyeonggi 17104, Republic of Korea
| | - Jongdae Shin
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| | - Joon Lee
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| |
Collapse
|
27
|
Wang JM, Qiu Y, Yang Z, Kim H, Qian Q, Sun Q, Zhang C, Yin L, Fang D, Back SH, Kaufman RJ, Yang L, Zhang K. IRE1α prevents hepatic steatosis by processing and promoting the degradation of select microRNAs. Sci Signal 2018; 11:11/530/eaao4617. [PMID: 29764990 DOI: 10.1126/scisignal.aao4617] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Obesity or a high-fat diet represses the endoribonuclease activity of inositol-requiring enzyme 1α (IRE1α), a transducer of the unfolded protein response (UPR) in cells under endoplasmic reticulum (ER) stress. An impaired UPR is associated with hepatic steatosis and nonalcoholic fatty liver disease (NAFLD), which is caused by lipid accumulation in the liver. We found that IRE1α was critical to maintaining lipid homeostasis in the liver by repressing the biogenesis of microRNAs (miRNAs) that regulate lipid mobilization. In mice fed normal chow, the endoribonuclease function of IRE1α processed a subset of precursor miRNAs in the liver, including those of the miR-200 and miR-34 families, such that IRE1α promoted their degradation through the process of regulated IRE1-dependent decay (RIDD). A high-fat diet in mice or hepatic steatosis in patients was associated with the S-nitrosylation of IRE1α and inactivation of its endoribonuclease activity. This resulted in an increased abundance of these miRNA families in the liver and, consequently, a decreased abundance of their targets, which included peroxisome proliferator-activated receptor α (PPARα) and the deacetylase sirtuin 1 (SIRT1), regulators of fatty acid oxidation and triglyceride lipolysis. IRE1α deficiency exacerbated hepatic steatosis in mice. The abundance of the miR-200 and miR-34 families was also increased in cultured, lipid-overloaded hepatocytes and in the livers of patients with hepatic steatosis. Our findings reveal a mechanism by which IRE1α maintains lipid homeostasis through its regulation of miRNAs, a regulatory pathway distinct from the canonical IRE1α-UPR pathway under acute ER stress.
Collapse
Affiliation(s)
- Jie-Mei Wang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA. .,Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, MI 48201, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhao Yang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Chunbin Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Lei Yin
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sung Hong Back
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA. .,Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
28
|
Liu Y, Zhang Y, Yang Q. Retracted
: Downregulated expression of microRNA‐329 inhibits apoptosis of nigral dopaminergic neurons by regulating CDKN2D expression via the FoxO3a signaling pathway in rats with Parkinson's disease. J Cell Physiol 2018; 233:8617-8629. [DOI: 10.1002/jcp.26608] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 03/22/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Yuan‐Yuan Liu
- Department of Neurologythe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouP.R. China
| | - Yi‐Nan Zhang
- Department of Neurologythe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouP.R. China
| | - Qing‐Shan Yang
- Department of Radiation Oncologythe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouLiaoning ProvinceP.R. China
| |
Collapse
|
29
|
Abstract
INTRODUCTION The aim of this study was to clarify the microRNA (miRNA) expression profiles of RAW264.7 macrophages infected by Candida albicans to elucidate the roles of differentially expressed miRNAs and to further explore the mechanisms underlying the immune response to C. albicans infection. METHODS High-throughput miRNA microarray analysis was performed to detect differentially expressed miRNAs in control and C. albicans-infected RAW264.7 cells. Quantitative real-time PCR analysis was used to verify the microarray results. Target genes of differentially expressed miRNAs were predicted with bioinformatics software. The cell biological processes and signaling pathways of these miRNA-targeted genes involved in C. albicans infection were predicted by gene ontology (GO) enrichment and pathway analyses. RESULTS Significant upregulation of eight miRNAs (mmu-miR-140-5p, mmu-miR-96-5p, mmu-miR-8109, mmu-miR-466i-3p, mmu-miR-222-5p, mmu-miR-301b-3p, mmu-miR-466g, and mmu-miR-7235-5p) and downregulation of eight miRNAs (mmu-miR-3154, mmu-miR-223-3p, mmu-miR-494-3p, mmu-miR-6908-5p, mmu-miR-188-5p, mmu-miR-6769b-5p, mmu-miR-7002-5p, and mmu-miR-1224-5p) were observed, as compared with the control (fold change ≥2.0 and P < 0.05). GO analysis revealed that both mmu-miR-140-5p and mmu-miR-223-3p participated in immune responses, inflammatory reactions, and cell apoptosis in C. albicans infection. Also, the MAPK signaling pathway was found to play an important role in the immune response against C. albicans infection. CONCLUSIONS This study revealed comprehensive expression and functional profiles of differentially expressed miRNAs in macrophage RAW264.7 cells infected by C. albicans. These findings should help to further elucidate the mechanisms underlying the immune response to C. albicans infection.
Collapse
|
30
|
Parkinson NJ, Buechner-Maxwell VA, Witonsky SG, Pleasant RS, Werre SR, Ahmed SA. Characterization of basal and lipopolysaccharide-induced microRNA expression in equine peripheral blood mononuclear cells using Next-Generation Sequencing. PLoS One 2017; 12:e0177664. [PMID: 28552958 PMCID: PMC5446123 DOI: 10.1371/journal.pone.0177664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 05/01/2017] [Indexed: 02/03/2023] Open
Abstract
The innate immune response to lipopolysaccharide contributes substantially to the morbidity and mortality of gram-negative sepsis. Horses and humans share an exquisite sensitivity to lipopolysaccharide and thus the horse may provide valuable comparative insights into this aspect of the inflammatory response. MicroRNAs, small non-coding RNA molecules acting as post-transcriptional regulators of gene expression, have key roles in toll-like receptor signaling regulation but have not been studied in this context in horses. The central hypothesis of this study was that lipopolysaccharide induces differential microRNA expression in equine peripheral blood mononuclear cells in a manner comparable to humans. Illumina Next Generation Sequencing was used to characterize the basal microRNA transcriptome in isolated peripheral blood mononuclear cells from healthy adult horses, and to evaluate LPS-induced changes in microRNA expression in cells cultured for up to four hours. Selected expression changes were validated using quantitative reverse-transcriptase PCR. Only miR-155 was significantly upregulated by LPS, changing in parallel with supernatant tumor necrosis factor-α concentration. Eight additional microRNAs, including miR-146a and miR-146b, showed significant expression change with time in culture without a clear LPS effect. Target predictions indicated a number of potential immunity-associated targets for miR-155 in the horse, including SOCS1, TAB2 and elements of the PI3K signaling pathway, suggesting that it is likely to influence the acute inflammatory response to LPS. Gene alignment showed extensive conservation of the miR-155 precursor gene and associated promoter regions between horses and humans. The basal and LPS-stimulated microRNA expression pattern characterized here were similar to those described in human leukocytes. As well as providing a resource for further research into the roles of microRNAs in immune responses in horses, this will facilitate inter-species comparative study of the role of microRNAs in the inflammatory cascade during endotoxemia and sepsis.
Collapse
Affiliation(s)
- Nicholas J. Parkinson
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, Virginia, United States of America
- * E-mail:
| | - Virginia A. Buechner-Maxwell
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, Virginia, United States of America
| | - Sharon G. Witonsky
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, Virginia, United States of America
| | - R. Scott Pleasant
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, Virginia, United States of America
| | - Stephen R. Werre
- Laboratory for Study Design and Statistical Analysis, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, Virginia, United States of America
| | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic and State University, Blacksburg, Virginia, United States of America
| |
Collapse
|
31
|
Kumar Kingsley SM, Vishnu Bhat B. Role of MicroRNAs in the development and function of innate immune cells. Int Rev Immunol 2017; 36:154-175. [DOI: 10.1080/08830185.2017.1284212] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- S. Manoj Kumar Kingsley
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - B. Vishnu Bhat
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
32
|
miR-155 Regulated Inflammation Response by the SOCS1-STAT3-PDCD4 Axis in Atherogenesis. Mediators Inflamm 2016; 2016:8060182. [PMID: 27843203 PMCID: PMC5098093 DOI: 10.1155/2016/8060182] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/01/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022] Open
Abstract
Inflammation response plays a critical role in all phases of atherosclerosis (AS). Increased evidence has demonstrated that miR-155 mediates inflammatory mediators in macrophages to promote plaque formation and rupture. However, the precise mechanism of miR-155 remains unclear in AS. Here, we also found that miR-155 and PDCD4 were elevated in the aortic tissue of atherosclerotic mice and ox-LDL treated RAW264.7 cells. Further studies showed that miR-155 not only directly inhibited SOCS1 expression, but also increased the expression of p-STAT and PDCD4, as well as the production of proinflammation mediators IL-6 and TNF-α. Downregulation of miR-155 and PDCD4 and upregulation of SOCS1 obviously decreased the IL-6 and TNF-α expression. In addition, inhibition of miR-155 levels in atherosclerotic mice could notably reduce the IL-6 and TNF-α level in plasma and aortic tissue, accompanied with increased p-STAT3 and PDCD4 and decreased SOCS1. Thus, miR-155 might mediate the inflammation in AS via the SOCS1-STAT3-PDCD4 axis. These results provide a rationale for intervention of intracellular miR-155 as possible antiatherosclerotic targets.
Collapse
|
33
|
Liepelt A, Naarmann-de Vries IS, Simons N, Eichelbaum K, Föhr S, Archer SK, Castello A, Usadel B, Krijgsveld J, Preiss T, Marx G, Hentze MW, Ostareck DH, Ostareck-Lederer A. Identification of RNA-binding Proteins in Macrophages by Interactome Capture. Mol Cell Proteomics 2016; 15:2699-714. [PMID: 27281784 DOI: 10.1074/mcp.m115.056564] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Indexed: 12/11/2022] Open
Abstract
Pathogen components, such as lipopolysaccharides of Gram-negative bacteria that activate Toll-like receptor 4, induce mitogen activated protein kinases and NFκB through different downstream pathways to stimulate pro- and anti-inflammatory cytokine expression. Importantly, post-transcriptional control of the expression of Toll-like receptor 4 downstream signaling molecules contributes to the tight regulation of inflammatory cytokine synthesis in macrophages. Emerging evidence highlights the role of RNA-binding proteins (RBPs) in the post-transcriptional control of the innate immune response. To systematically identify macrophage RBPs and their response to LPS stimulation, we employed RNA interactome capture in LPS-induced and untreated murine RAW 264.7 macrophages. This combines RBP-crosslinking to RNA, cell lysis, oligo(dT) capture of polyadenylated RNAs and mass spectrometry analysis of associated proteins. Our data revealed 402 proteins of the macrophage RNA interactome including 91 previously not annotated as RBPs. A comparison with published RNA interactomes classified 32 RBPs uniquely identified in RAW 264.7 macrophages. Of these, 19 proteins are linked to biochemical activities not directly related to RNA. From this group, we validated the HSP90 cochaperone P23 that was demonstrated to exhibit cytosolic prostaglandin E2 synthase 3 (PTGES3) activity, and the hematopoietic cell-specific LYN substrate 1 (HCLS1 or HS1), a hematopoietic cell-specific adapter molecule, as novel macrophage RBPs. Our study expands the mammalian RBP repertoire, and identifies macrophage RBPs that respond to LPS. These RBPs are prime candidates for the post-transcriptional regulation and execution of LPS-induced signaling pathways and the innate immune response. Macrophage RBP data have been deposited to ProteomeXchange with identifier PXD002890.
Collapse
Affiliation(s)
- Anke Liepelt
- From the ‡Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Isabel S Naarmann-de Vries
- From the ‡Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Nadine Simons
- From the ‡Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Katrin Eichelbaum
- §European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Sophia Föhr
- §European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Stuart K Archer
- ¶EMBL-Australia Collaborating Group, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Garran Rd, Acton (Canberra) ACT 2601, Australia
| | - Alfredo Castello
- §European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Björn Usadel
- ‖Institute of Biology I, RWTH Aachen, Worringer Weg 2, 52074 Aachen, Germany
| | - Jeroen Krijgsveld
- §European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Thomas Preiss
- ¶EMBL-Australia Collaborating Group, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Garran Rd, Acton (Canberra) ACT 2601, Australia; **Victor Chang Cardiac Research Institute, Darlinghurst (Sydney), New South Wales 2010, Australia
| | - Gernot Marx
- From the ‡Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Matthias W Hentze
- §European Molecular Biology Laboratory (EMBL), Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Dirk H Ostareck
- From the ‡Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany;
| | - Antje Ostareck-Lederer
- From the ‡Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany;
| |
Collapse
|
34
|
Huang Q, Xiao B, Ma X, Qu M, Li Y, Nagarkatti P, Nagarkatti M, Zhou J. MicroRNAs associated with the pathogenesis of multiple sclerosis. J Neuroimmunol 2016; 295-296:148-61. [DOI: 10.1016/j.jneuroim.2016.04.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 12/14/2022]
|
35
|
Larrea E, Sole C, Manterola L, Goicoechea I, Armesto M, Arestin M, Caffarel MM, Araujo AM, Araiz M, Fernandez-Mercado M, Lawrie CH. New Concepts in Cancer Biomarkers: Circulating miRNAs in Liquid Biopsies. Int J Mol Sci 2016; 17:ijms17050627. [PMID: 27128908 PMCID: PMC4881453 DOI: 10.3390/ijms17050627] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/18/2016] [Accepted: 04/18/2016] [Indexed: 12/19/2022] Open
Abstract
The effective and efficient management of cancer patients relies upon early diagnosis and/or the monitoring of treatment, something that is often difficult to achieve using standard tissue biopsy techniques. Biological fluids such as blood hold great possibilities as a source of non-invasive cancer biomarkers that can act as surrogate markers to biopsy-based sampling. The non-invasive nature of these “liquid biopsies” ultimately means that cancer detection may be earlier and that the ability to monitor disease progression and/or treatment response represents a paradigm shift in the treatment of cancer patients. Below, we review one of the most promising classes of circulating cancer biomarkers: microRNAs (miRNAs). In particular, we will consider their history, the controversy surrounding their origin and biology, and, most importantly, the hurdles that remain to be overcome if they are really to become part of future clinical practice.
Collapse
Affiliation(s)
- Erika Larrea
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - Carla Sole
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - Lorea Manterola
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - Ibai Goicoechea
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - María Armesto
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - María Arestin
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - María M Caffarel
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| | - Angela M Araujo
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - María Araiz
- Hematology Department, Donostia Hospital, 20014 San Sebastián, Spain.
| | | | - Charles H Lawrie
- Molecular Oncology, Biodonostia Research Institute, 20014 San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, Oxford OX3 9DU, UK.
| |
Collapse
|