1
|
Hart M, Diener C, Rheinheimer S, Kehl T, Keller A, Lenhof HP, Meese E. Expanding the immune-related targetome of miR-155-5p by integrating time-resolved RNA patterns into miRNA target prediction. RNA Biol 2025; 22:1-9. [PMID: 39760255 PMCID: PMC11730359 DOI: 10.1080/15476286.2025.2449775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/14/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025] Open
Abstract
The lack of a sufficient number of validated miRNA targets severely hampers the understanding of their biological function. Even for the well-studied miR-155-5p, there are only 239 experimentally validated targets out of 42,554 predicted targets. For a more complete assessment of the immune-related miR-155 targetome, we used an inverse correlation of time-resolved mRNA profiles and miR-155-5p expression of early CD4+ T cell activation to predict immune-related target genes. Using a high-throughput miRNA interaction reporter (HiTmIR) assay we examined 90 target genes and confirmed 80 genes as direct targets of miR-155-5p. Our study increases the current number of verified miR-155-5p targets approximately threefold and exemplifies a method for verifying miRNA targetomes as a prerequisite for the analysis of miRNA-regulated cellular networks.
Collapse
Affiliation(s)
- Martin Hart
- Institute of Human Genetics, Saarland University (USAAR), Homburg, Germany
- Center of Human and Molecular Biology (ZHMB), Saarland University (USAAR), Saarbrücken, Germany
| | - Caroline Diener
- Institute of Human Genetics, Saarland University (USAAR), Homburg, Germany
| | | | - Tim Kehl
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University (USAAR), Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University (USAAR), Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)–Helmholtz Centre for Infection Research (HZI), Saarland University Campus, Saarbrücken, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University (USAAR), Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University (USAAR), Homburg, Germany
| |
Collapse
|
2
|
Sun W, Hu K, Song Z, An R, Liang X. One-Pot Detection of miRNA by Dual Rolling Circle Amplification at Ambient Temperature with High Specificity and Sensitivity. BIOSENSORS 2025; 15:317. [PMID: 40422056 DOI: 10.3390/bios15050317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/07/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025]
Abstract
Rolling circle amplification (RCA) at ambient temperature is prone to false positive signals during nucleic acid detection, which makes it challenging to establish an efficient RCA detection method. The false positive signals are primarily caused by binding of non-target nucleic acids to the circular single-stranded template, leading to non-specific amplification. Here, we present an RCA method for miRNA detection at 37 °C using two circular ssDNAs, each of which is formed by ligating the intramolecularly formed nick (without any splint) in a secondary structure. The specific target recognition is realized by utilizing low concentrations (0.1 nM) of circular ssDNA1 (C1). A phosphorothioate modification is present at G*AATTC on C1 to generate a nick for primer extension during the primer self-generated rolling circle amplification (PG-RCA). The fragmented amplification products are used as primers for the following RCA that serves as signal amplification using circular ssDNA2 (C2). Notably, the absence of splints and the low concentration of C1 significantly inhibits non-target binding, thus minimizing false positive signals. A high concentration (10 nM) of C2 is used to carry out linear rolling circle amplification (LRCA), which is highly specific. This strategy demonstrates a good linear response to 0.01-100 pM of miRNA with a detection limit of 7.76 fM (miR-155). Moreover, it can distinguish single-nucleotide mismatch in the target miRNA, enabling the rapid one-pot detection of miRNA at 37 °C. Accordingly, this method performs with high specificity and sensitivity. This approach is suitable for clinical serum sample analysis and offers a strategy for developing specific biosensors and diagnostic tools.
Collapse
Affiliation(s)
- Wenhua Sun
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China
| | - Kunling Hu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China
| | - Ziting Song
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China
| | - Ran An
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, No. 1 Wenhai Road, Qingdao 266237, China
| | - Xingguo Liang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, No. 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
3
|
Joldes C, Jimbu L, Mesaros O, Zdrenghea M, Fetica B. Tumor-Associated Macrophages as Key Modulators of Disease Progression in Diffuse Large B-Cell Lymphoma. Biomedicines 2025; 13:1099. [PMID: 40426926 PMCID: PMC12108958 DOI: 10.3390/biomedicines13051099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/17/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
With the advent of new therapeutic approaches, there is hope that anticancer treatment will eventually be possible without the use of chemotherapy. Efficient immunotherapeutic options have recently emerged in many cancers, offering a less aggressive approach, with overall better tolerance, making them also suitable for frail patients. Response to immunotherapy relies on the availability, functionality, and efficacy of the host's immune effector mechanisms. One of the key factors determining the efficacy of immunotherapy is the tumor microenvironment, which encompasses various immune effectors, including macrophages, which play a crucial role in regulating immune responses through phagocytosis and antigen presentation. Macrophages are prototypically divided, according to their polarization, into either the pro-inflammatory M1 type or the anti-inflammatory M2 type. In the tumor microenvironment, M2-polarized macrophages, known as tumor-associated macrophages (TAMs), are the predominant phenotype and are associated with tumor progression. The M1/M2 paradigm contributes to the understanding of tumor progression. Due to the variable microenvironment, the mechanisms regulating TAMs can vary across different cancers. Variations in TAM polarization may account for the different treatment responses in patients with similar diseases. This paper investigates the connection between TAMs, disease progression, and treatment responses in the most frequent solid hematologic cancer, diffuse large B-cell lymphoma.
Collapse
Affiliation(s)
- Corina Joldes
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Street, 400012 Cluj-Napoca, Romania; (L.J.); (O.M.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Bogdan Fetica
- Department of Pathology, Ion Chiricuta Oncology Institute, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania;
| |
Collapse
|
4
|
Pan Y, Zheng J, Ren Z, Yuan W, Zhao J, Zheng C, Zeng Y. RTA activates the activity of the miR-155 promoter region and promotes the growth and invasion of endothelial cells through the regulatory network of miR-155/GATA3/STAT3. Int J Biol Macromol 2025; 310:143536. [PMID: 40288717 DOI: 10.1016/j.ijbiomac.2025.143536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Kaposi's sarcoma (KS) is a malignant tumor primarily derived from endothelial cells. KS is the most common malignant tumor in AIDS patients and is aggressive. Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is the etiological basis of KS. However, the underlying mechanism is still unclear. In this study, the KSHV virus was used to infect the human umbilical vein-fused endothelial cell line EAhy926 and the human ovarian microvascular endothelial cell line HOMEC. KSHV promoted the growth and invasion of endothelial cells and induced tumorigenesis in nude mice. Then, the growth-promoting and invasive effects of the protein replication and transcription activator (RTA) encoded by KSHV on endothelial cells were clarified. In addition, this study found that RTA can promote the expression of miR-155 by activating the activity of miR-155 promoter region. Previous studies have confirmed the inhibitory effect of miR-155 on GATA3 and GATA3 on STAT3. On this basis, this study made it clear that RTA can regulate the expression of miR-155/GATA3/STAT3, and then promote the growth and invasion of endothelial cells through this regulatory network. The purpose of this study is to explore the role of RTA in the growth and invasion of endothelial cells, as well as the regulation and specific regulation mechanism of RTA on the expression of miR-155/GATA3/STAT3, so as to determine whether RTA promotes the growth and invasion of endothelial cells through the regulation network of miR-155, so as to open up a new way for the treatment of diseases related to KSHV.
Collapse
Affiliation(s)
- Yangyang Pan
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524037, China; State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Jun Zheng
- Department of Stomatology, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524037, China
| | - Zuodong Ren
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524037, China
| | - Wumei Yuan
- Key Laboratory of Xinjiang Endemic and Ethnic Disease, School of Medicine, Shihezi University, Shihezi 832002, China
| | - Juan Zhao
- Key Laboratory of Xinjiang Endemic and Ethnic Disease, School of Medicine, Shihezi University, Shihezi 832002, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Yan Zeng
- Precision Clinical Laboratory, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524037, China.
| |
Collapse
|
5
|
da Silva-Neto PV, de Carvalho JCS, Toro DM, Oliveira BTM, Cominal JG, Castro RC, Almeida MA, Prado CM, Arruda E, Frantz FG, Ramos AP, Ciancaglini P, Martins RB, da Silveira JC, Almeida F, Malmegrim KCR, Sorgi CA. TREM-1-Linked Inflammatory Cargo in SARS-CoV-2-Stimulated Macrophage Extracellular Vesicles Drives Cellular Senescence and Impairs Antibacterial Defense. Viruses 2025; 17:610. [PMID: 40431622 PMCID: PMC12115590 DOI: 10.3390/v17050610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/29/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has significantly affected global health, with severe inflammatory responses leading to tissue damage and persistent symptoms. Macrophage-derived extracellular vesicles (EVs) are involved in the modulation of immune responses, but their involvement in SARS-CoV-2-induced inflammation and senescence remains unclear. Triggering receptors expressed on myeloid cell-1 (TREM-1) are myeloid cell receptors that amplify inflammation, described as a biomarker of the severity and mortality of COVID-19. This study investigated the composition and effects of macrophage-derived EVs stimulated by SARS-CoV-2 (MφV-EVs) on the recipient cell response. Our results, for the first time, show that SARS-CoV-2 stimulation modifies the cargo profile of MφV-EVs, enriching them with TREM-1 and miRNA-155 association, along with MMP-9 and IL-8/CXCL8. These EVs carry senescence-associated secretory phenotype (SASP) components, promote cellular senescence, and compromise antibacterial defenses upon internalization. Our findings provide evidence that MφV-EVs are key drivers of inflammation and immune dysfunction, underscoring their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Pedro V. da Silva-Neto
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Jonatan C. S. de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Diana M. Toro
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (D.M.T.); (E.A.)
| | - Bianca T. M. Oliveira
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
| | - Juçara G. Cominal
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
| | - Ricardo C. Castro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Maria A. Almeida
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos-FZEA, Universidade de São Paulo-USP, Pirassununga 13635-900, SP, Brazil; (M.A.A.); (C.M.P.); (J.C.d.S.)
| | - Cibele M. Prado
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos-FZEA, Universidade de São Paulo-USP, Pirassununga 13635-900, SP, Brazil; (M.A.A.); (C.M.P.); (J.C.d.S.)
| | - Eurico Arruda
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (D.M.T.); (E.A.)
| | - Fabiani G. Frantz
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Ana P. Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
| | - Ronaldo B. Martins
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Juliano C. da Silveira
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos-FZEA, Universidade de São Paulo-USP, Pirassununga 13635-900, SP, Brazil; (M.A.A.); (C.M.P.); (J.C.d.S.)
| | - Fausto Almeida
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
| | - Kelen C. R. Malmegrim
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Carlos A. Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
- Programa de Pós-graduação em Imunologia Básica e Aplicada-PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas-UFAM, Manaus 69080-900, AM, Brazil
| |
Collapse
|
6
|
Hering C, Conover GM. Advancing Ischemic Stroke Prognosis: Key Role of MiR-155 Non-Coding RNA. Int J Mol Sci 2025; 26:3947. [PMID: 40362186 PMCID: PMC12071504 DOI: 10.3390/ijms26093947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Ischemic stroke (IS) is the leading cause of long-term disability and the second leading cause of death worldwide. It remains a significant clinical problem because only supportive therapies exist, such as thrombolytic agents and surgical thrombectomy, which do not restore function. Understanding the molecular pathogenesis of IS, including dysfunction in oxidative homeostasis, apoptosis, neuroinflammation and neuroprotection, is crucial to developing therapies. Non-coding RNAs (ncRNAs) are master regulators, and one ncRNA that stands out is miR-155, a pro-inflammatory micro-RNA elevated in stroke. This review addresses the biological mechanisms reported in the literature that support using miR-155 as a biomarker and therapeutic agent to treat IS in patients.
Collapse
Affiliation(s)
| | - Gloria M. Conover
- Department of Medical Education, College of Medicine, Texas A&M University, Bryan, TX 77807, USA;
| |
Collapse
|
7
|
Li W, Yang C, Xu J, Ran D, Wang C. MIR155HG suppresses the osteogenic differentiation of bone marrow mesenchymal stem cells through regulating miR-155-5p and DKK1 expression. J Orthop Surg Res 2025; 20:392. [PMID: 40251598 PMCID: PMC12008851 DOI: 10.1186/s13018-025-05798-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Increasing evidence has demonstrated that non-coding RNAs, including the lncRNA MIR155HG, are involved in the pathogenesis of postmenopausal osteoporosis (PMOP). In the current study, we studied MIR155HG function in regulation of osteogenic differentiation and tried to reveal the underlying mechanisms. METHODS Forty blood samples taken from 20 PMOP patients (PMOP group) and 20 postmenopausal individuals without osteoporosis (control group) were used to compare the contents of MIR155HG and miR-155-5p via RT-PCR. Alizarin red S staining and ALP staining were used to evaluate the osteogenic differentiation potential of bone marrow mesenchymal stem cells (BMSCs). RESULTS Elevated levels of MIR155HG and miR-155-5p were observed in the blood samples of the PMOP group. Upregulation of MIR155HG resulted in decreased expression of OPN, OSX, ALP, RUNX2 and β-catenin but increased DKK1 expression, together with decreased Alizarin red S + and ALP + staining areas. However, downregulation of DKK1 did not obviously change the above indices induced by MIR155HG upregulation. Further experiments revealed that MIR155HG caused an increase in the expression of miR-155-5p, which also serves as an inhibitor of the osteogenic differentiation of BMSCs through binding to β-catenin. Consistent with DKK1 knockdown, downregulation of miR-155-5p only also did not obviously reverse the repressive effect of MIR155HG on osteoblastic differentiation, but downregulation of DKK1 and miR-155-5p synchronously restored the osteogenic differentiation ability of BMSCs suppressed by MIR155HG overexpression. CONCLUSION MIR155HG suppressed the osteoblastic differentiation of BMSCs by regulating miR-155-5p and DKK1 expression. Either inhibition of miR-155-5p and DKK1 or direct suppression of MIR155HG may be effective approaches for treating PMOP.
Collapse
Affiliation(s)
- Weimin Li
- Department of Orthopedic, The Fourth People's Hospital of Guiyang, Guiyang Guizhou, 550002, China
| | - Cheng Yang
- Department of Orthopedic, Guizhou Hospital of Beijing Jishuitan Hospital, Guiyang Guizhou, 550014, China
| | - Jiamu Xu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Dongcheng Ran
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Chunqing Wang
- Department of Traumatology and Orthopedics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, China.
| |
Collapse
|
8
|
Golovina E, Kokavec J, Kazantsev D, Yurikova O, Bajecny M, Savvulidi FG, Simersky R, Lenobel R, Tost J, Herynek V, Sefc L, Sebela M, Klener P, Zemanova Z, Stopka T, Vargova KS. Deficiency of miR-155 in Leukemic B-Cells Results in Cell Cycle Arrest and Deregulation of MIR155HG/TP53INP1/CDKN1A/CCND1 network. Arch Med Res 2025; 56:103124. [PMID: 39591901 DOI: 10.1016/j.arcmed.2024.103124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/15/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Cell cycle progression and leukemia development are tightly regulated processes in which even a small imbalance in the expression of cell cycle regulatory molecules and microRNAs (miRNAs) can lead to an increased risk of cancer/leukemia development. Here, we focus on the study of a ubiquitous, multifunctional, and oncogenic miRNA-hsa-miR-155-5p (miR-155, MIR155HG), which is overexpressed in malignancies including chronic lymphocytic leukemia (CLL). Nonetheless, the precise mechanism of how miR-155 regulates the cell cycle in leukemic cells remains the subject of extensive research. METHODS We edited the CLL cell line MEC-1 by CRISPR/Cas9 to introduce a short deletion within the MIR155HG gene. To describe changes at the transcriptome and miRNome level in miR-155-deficient cells, we performed mRNA-seq/miRNA-seq and validated changes by qRT-PCR. Flow cytometry was used to measure cell cycle kinetics. A WST-1 assay, hemocytometer, and Annexin V/PI staining assessed cell viability and proliferation. RESULTS The limited but phenotypically robust miR-155 modification impaired cell proliferation, cell cycle, and cell ploidy. This was accompanied by overexpression of the negative cell cycle regulator p21/CDKN1A and Cyclin D1 (CCND1). We confirmed the overexpression of canonical miR-155 targets such as PU.1, FOS, SHIP-1, TP53INP1 and revealed new potential targets (FCRL5, ISG15, and MX1). CONCLUSIONS We demonstrate that miR-155 deficiency impairs cell proliferation, cell cycle, transcriptome, and miRNome via deregulation of the MIR155HG/TP53INP1/CDKN1A/CCND1 axis. Our CLL model is valuable for further studies to manipulate miRNA levels to revert highly aggressive leukemic cells to nearly benign or non-leukemic types.
Collapse
Affiliation(s)
- Elena Golovina
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Juraj Kokavec
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Dmitry Kazantsev
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Oxana Yurikova
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Kazakhstan
| | - Martin Bajecny
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic; The Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Filipp Georgijevic Savvulidi
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University, Prague, Kamýcká, Czech Republic
| | - Radim Simersky
- Department of Chemical Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Rene Lenobel
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences and Palacký University, Olomouc, Czech Republic
| | - Jorg Tost
- Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie Francois Jacob, Universite Paris-Saclay, Évry, France
| | - Vit Herynek
- The Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ludek Sefc
- The Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Sebela
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zuzana Zemanova
- Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Stopka
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Karina Savvulidi Vargova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
9
|
O’Dowd K, Vatandour S, Ahamed SS, Boulianne M, Dozois CM, Gagnon CA, Barjesteh N, Abdul-Careem MF. Characterization of microRNA candidates at the primary site of infectious bronchitis virus infection: A comparative study of in vitro and in vivo avian models. PLoS One 2025; 20:e0319153. [PMID: 40067877 PMCID: PMC11896067 DOI: 10.1371/journal.pone.0319153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/28/2025] [Indexed: 03/15/2025] Open
Abstract
Infectious bronchitis virus (IBV) is an important avian pathogen with a positive-sense single-stranded RNA genome. IBV is the causative agent of infectious bronchitis (IB), a primarily respiratory disease affecting chickens, with the ability to disseminate to other organ systems, such as the gastrointestinal, renal, lymphoid, and reproductive systems. Tracheal epithelial cells are the primary target of IBV, and these cells play a vital role in the effective induction of the antiviral response and eventual clearance of IBV. The host immune system is regulated by a number of different molecular players, including micro-ribonucleic acids (microRNAs), which are small, conserved, non-coding RNA molecules that regulate gene expression of complementary messenger RNA (mRNA) sequences, resulting in gene silencing through translational repression or target degradation. The goal of this study was to characterize and compare the microRNA expression profiles in chicken tracheal epithelial cells (cTECs) in vitro and the trachea in vivo upon IBV Delmarva/1639 (DMV/1639) or IBV Massachusetts 41 (Mass41) infections. We hypothesized that IBV infection influences the expression of the host microRNA expression profiles. cTECs and young specific pathogen-free (SPF) chickens were infected with IBV DMV/1639 or IBV Mass41 and the microRNA expression at 3 and 18 hours post-infection (hpi) in the cTECs and at 4 and 11 days post-infection (dpi) in the trachea were determined using small RNA-sequencing (RNA-seq). We found that the profile of differentially expressed (DE) microRNAs is largely dependent on the IBV strain and time point of sample collection. Furthermore, we predicted the interaction between host microRNA and IBV viral RNA using microRNA-RNA interaction prediction platforms. We identified several candidate microRNAs suitable for future functional studies, such as gga-miR-155, gga-miR-1388a, gga-miR-7/7b and gga-miR-21-5p. Characterizing the interaction between IBV and the host cells at the level of microRNA regulation provides further insight into the regulatory mechanisms involved in viral infection and host defense in chickens following IBV infection.
Collapse
Affiliation(s)
- Kelsey O’Dowd
- Health Research Innovation Centre, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Safieh Vatandour
- Department of Animal and Poultry Science, Islamic Azad University, Qaemshahr Branch, Qaemshahr, Mazandaran, Iran
| | - Sadhiya S. Ahamed
- Health Research Innovation Centre, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Martine Boulianne
- Swine and Poultry Infectious Diseases Research Centre – Fonds de recherche du Québec (CRIPA-FRQ), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Charles M. Dozois
- Swine and Poultry Infectious Diseases Research Centre – Fonds de recherche du Québec (CRIPA-FRQ), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Institut National de Recherche Scientifique-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Carl A. Gagnon
- Swine and Poultry Infectious Diseases Research Centre – Fonds de recherche du Québec (CRIPA-FRQ), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Molecular Diagnostic and Virology Laboratories, Centre de diagnostic vétérinaire de l’Université de Montréal (CDVUM), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Neda Barjesteh
- Swine and Poultry Infectious Diseases Research Centre – Fonds de recherche du Québec (CRIPA-FRQ), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Mohamed Faizal Abdul-Careem
- Health Research Innovation Centre, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Zhang RL, Wang WM, Li JQ, Li RW, Zhang J, Wu Y, Liu Y. The role of miR-155 in cardiovascular diseases: Potential diagnostic and therapeutic targets. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 24:200355. [PMID: 39760132 PMCID: PMC11699627 DOI: 10.1016/j.ijcrp.2024.200355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025]
Abstract
Cardiovascular diseases (CVDs), such as atherosclerotic cardiovascular diseases, heart failure (HF), and acute coronary syndrome, represent a significant threat to global health and impose considerable socioeconomic burdens. The intricate pathogenesis of CVD involves various regulatory mechanisms, among which microRNAs (miRNAs) have emerged as critical posttranscriptional regulators. In particular, miR-155 has demonstrated differential expression patterns across a spectrum of CVD and is implicated in the etiology and progression of arterial disorders. This systematic review synthesizes current evidence on the multifaceted roles of miR-155 in the modulation of genes and pathological processes associated with CVD. We delineate the potential of miR-155 as a diagnostic biomarker and therapeutic target, highlighting its significant regulatory influence on conditions such as atherosclerosis, aneurysm, hypertension, HF, myocardial hypertrophy, and oxidative stress. Our analysis underscores the transformative potential of miR-155 as a target for intervention in cardiovascular medicine, warranting further investigation into its clinical applicability.
Collapse
Affiliation(s)
- Rui-Lin Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Wei-Ming Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ji-Qiang Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Run-Wen Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
11
|
Zhang X, Cai Y, Sit BHM, Jian RX, Malki Y, Zhang Y, Ong CCY, Li Q, Lam RPK, Rainer TH. Cell-Free Nucleic Acids for Early Diagnosis of Acute Ischemic Stroke: A Systematic Review and Meta-Analysis. Int J Mol Sci 2025; 26:1530. [PMID: 40003998 PMCID: PMC11855205 DOI: 10.3390/ijms26041530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Rapid identification of acute ischemic stroke (AIS) is challenging in both pre-hospital and hospital settings. We aimed to identify the most promising cell-free nucleic acids (cfNAs) as diagnostic biomarkers for IS within 72 h from symptom onset. We searched PubMed, Web of Science, EMBASE, and Cochrane Library for published articles that evaluated blood cfNAs in the early diagnosis of AIS until 10 May 2023. The diagnostic performances of individual cfNAs were pooled by random-effects meta-analysis based on the fold change of biomarkers' level between AIS and non-AIS patients. Of 2955 records, 66 articles reporting 143 different cfNAs met the inclusion criteria. The median sample size was 110, and 21.4% of the studies performed validation. Among selected high-quality studies, miR-106b-5p, miR-124, miR-155, lncRNA H19, and cfDNA showed good diagnostic performance. Data from four studies on cfDNA involving 355 AIS patients and 97 controls were pooled in the meta-analysis, which showed a significant fold change between AIS and controls (pooled ratio 1.48, 95% confidence interval 1.23-1.79, p < 0.001). This review highlights that cfDNA, miR-106b-5p, miR-124, miR-155, and lncRNA H19 are the most promising biomarkers for AIS diagnosis, and further research is needed for verification.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Yuee Cai
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Brian Hon Man Sit
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Rain Xiaoyu Jian
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Yasine Malki
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China;
| | - Yilin Zhang
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Christopher Chi Yat Ong
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Qianyun Li
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Rex Pui Kin Lam
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Timothy Hudson Rainer
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| |
Collapse
|
12
|
Emerson JI, Shi W, Paredes-Larios J, Walker WG, Hutton JE, Cristea IM, Marzluff WF, Conlon FL. X-Chromosome-Linked miRNAs Regulate Sex Differences in Cardiac Physiology. Circ Res 2025; 136:258-275. [PMID: 39772608 PMCID: PMC11781965 DOI: 10.1161/circresaha.124.325447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Males and females exhibit distinct anatomic and functional characteristics of the heart, predisposing them to specific disease states. METHODS We identified microRNAs (miRNAs/miR) with sex-differential expression in mouse hearts. RESULTS Four conserved miRNAs are present in a single locus on the X-chromosome and are expressed at higher levels in females than males. We show miRNA, miR-871, is responsible for decreased expression of the protein SRL (sarcalumenin) in females. SRL is involved in calcium signaling, and we show it contributes to differences in electrophysiology between males and females. miR-871 overexpression mimics the effects of the cardiac physiology of conditional cardiomyocyte-specific Srl-null mice. Inhibiting miR-871 with an antagomir in females shortened ventricular repolarization. The human orthologue of miR-871, miR-888, coevolved with the SRL 3' untranslated region and regulates human SRL. CONCLUSIONS These data highlight the importance of sex-differential miRNA mechanisms in mediating sex-specific functions and their potential relevance to human cardiac diseases.
Collapse
Affiliation(s)
- James I. Emerson
- Department of Biochemistry & Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Wei Shi
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jose Paredes-Larios
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William G. Walker
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Josiah E. Hutton
- Department of Molecular Biology, Princeton University, Lew Thomas Laboratory, Princeton, NJ 08544, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Lew Thomas Laboratory, Princeton, NJ 08544, USA
| | - William F. Marzluff
- Department of Biochemistry & Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Science, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
13
|
Khosrojerdi M, Azad FJ, Yadegari Y, Ahanchian H, Azimian A. The role of microRNAs in atopic dermatitis. Noncoding RNA Res 2024; 9:1033-1039. [PMID: 39022685 PMCID: PMC11254505 DOI: 10.1016/j.ncrna.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 07/20/2024] Open
Abstract
Atopic dermatitis (AD), known as eczema, is a chronic inflammatory skin condition affecting millions worldwide. This abstract provides an overview of the clinical features and underlying pathogenesis of AD, highlighting the role of specific microRNAs (miRNAs) in its development and progression. AD presents with distinct clinical manifestations that evolve with age, starting in infancy with dry, itchy skin and red patches, which can lead to sleep disturbances. In childhood, the rash spreads to flexural areas, resulting in lichenification. In adulthood, lesions may localize to specific areas, including the hands and eyelids. Pruritus (itchiness) is a hallmark symptom, often leading to excoriations and increased vulnerability to skin infections. The pathogenesis of AD is multifaceted, involving genetic, immunological, and environmental factors. Skin barrier dysfunction, immune dysregulation, genetic predisposition, microbiome alterations, and environmental triggers contribute to its development. Recent research has uncovered the role of miRNAs, such as miR-10a-5p, miR-29b, miR-124, miR-143, miR-146a-5p, miR-151a, miR-155, and miR-223, in AD pathogenesis. These microRNAs play crucial roles in regulating various aspects of immune responses, keratinocyte dynamics, and inflammation. MicroRNA-10a-5p orchestrates keratinocyte proliferation and differentiation, while miR-29b regulates keratinocyte apoptosis and barrier integrity. MicroRNA-124 exhibits anti-inflammatory effects by targeting the NF-κB signaling pathway. MicroRNANA-143 counters allergic inflammation by modulating IL-13 signaling. MicroRNA-146a-5p regulates immune responses and correlates with IgE levels in AD. MicroRNA-151a shows diagnostic potential and modulates IL-12 receptor β2. MicroRNA-155 plays a central role in immune responses and Th17 cell differentiation, offering diagnostic and therapeutic potential. MicroRNA-223 is linked to prenatal smoke exposure and immune modulation in AD. Understanding these microRNAs' intricate roles in AD pathogenesis promises more effective treatments, personalized approaches, and enhanced diagnostic tools. Further research into these molecular orchestrators may transform the landscape of AD management, improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Mahsa Khosrojerdi
- Department of Immunology and Allergy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farahzad Jabbari Azad
- Department of Immunology and Allergy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yaser Yadegari
- Department of Immunology and Allergy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Ahanchian
- Department of Immunology and Allergy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Azimian
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
14
|
Xu H, Li X, Liu K, Huang P, Liu XJ. PM2.5 Promotes Macrophage-Mediated Inflammatory Response Through Airway Epithelial Cell-Derived Exosomal miR-155-5p. J Inflamm Res 2024; 17:8555-8567. [PMID: 39539727 PMCID: PMC11559224 DOI: 10.2147/jir.s482509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Background Airway epithelial cells (AECs) and alveolar macrophages are involved in airway inflammation. The direct effects of atmospheric fine-particulate-matter (PM2.5) on airway cells, such as AECs and alveolar macrophages, have been widely investigated, but the effect of cell-cell interaction on inflammatory response remains unclear. Exosomes play a crucial role in intercellular communication. However, the cellular interaction of exosomes in PM2.5-induced airway inflammation is unclear. Methods The PM2.5-induced human bronchial epithelial (BEAS-2B) cells and phorbol 12-myristate 13-acetate-induced macrophages (Mφ) were co-cultured and then the expression of IL-6, IL-1β, TNF-α and miRNA-155-5p were detected. Exosomes from PM2.5-exposed BEAS-2B cells were then co-cultured with Mφ to detect the expression of miR-155-5p and inflammatory cytokines, as well as cytokine signaling inhibitor-1 (SOCS1)/NFκB, and to detect the effect of the exosome inhibitor GW4869. Results After the co-culture of PM2.5-induced BEAS-2B cells and Mφ, the expression of Mφ-derived IL-6, IL-1β, and TNF-α, as well as miRNA-155-5p were upregulated. The expression of miRNA-155-5p was upregulated in BEAS-2B and BEAS-2B cell-derived exosomes after exposure to PM2.5. Furthermore, co-culturing exosomes derived from PM2.5-exposed BEAS-2B cells with Mφ, upregulated miR-155-5p and inflammatory cytokines, decreased cytokine signaling inhibitor-1 (SOCS1) expression, and activated NF-κB. In addition, adding exosome inhibitor GW4869 to PM2.5-interfered BEAS-2B cells co-culture with Mφ downregulated miRNA-155-5p expression, inhibited NF-κB, and reduced the levels of inflammatory factors. Conclusion PM2.5 promotes Mφ inflammation by upregulating miRNA-155-5P in exosomes obtained from BEAS-2B cells through miR-155-5P/SOCS1/NF-κB pathway. Exosomal miRNAs mediate cellular communication between BEAS-2B cells and Mφ, which may be a new mechanism of PM2.5-stimulated pulmonary inflammatory response.
Collapse
Affiliation(s)
- Hui Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xin Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Kai Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Ping Huang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xiao-Ju Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
15
|
Capetini VC, Quintanilha BJ, Garcia BREV, Rogero MM. Dietary modulation of microRNAs in insulin resistance and type 2 diabetes. J Nutr Biochem 2024; 133:109714. [PMID: 39097171 DOI: 10.1016/j.jnutbio.2024.109714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
The prevalence of type 2 diabetes is increasing worldwide. Various molecular mechanisms have been proposed to interfere with the insulin signaling pathway. Recent advances in proteomics and genomics indicate that one such mechanism involves the post-transcriptional regulation of insulin signaling by microRNA (miRNA). These noncoding RNAs typically induce messenger RNA (mRNA) degradation or translational repression by interacting with the 3' untranslated region (3'UTR) of target mRNA. Dietary components and patterns, which can either enhance or impair the insulin signaling pathway, have been found to regulate miRNA expression in both in vitro and in vivo studies. This review provides an overview of the current knowledge of how dietary components influence the expression of miRNAs related to the control of the insulin signaling pathway and discusses the potential application of these findings in precision nutrition.
Collapse
Affiliation(s)
- Vinícius Cooper Capetini
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil; Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, Institute of Pharmaceutical Science, Department of Pharmacology, King's College London, London, United Kingdom.
| | - Bruna Jardim Quintanilha
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| | - Bruna Ruschel Ewald Vega Garcia
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Marcelo Macedo Rogero
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| |
Collapse
|
16
|
Guo Z, Tian JJ, Wang Y, Jiang L, Chen Y, Dai HJ, Wang L, Zhang Y. MiR-155-5p regulates autophagy and apoptosis of glioma cells through RICTOR. Transl Cancer Res 2024; 13:5509-5521. [PMID: 39525027 PMCID: PMC11543026 DOI: 10.21037/tcr-24-543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024]
Abstract
Background Glioma characterized by the high degree of drug resistance and the poor prognosis is the most common primary malignant tumors of the brain. And miRNA is involved in a variety of biological behaviors of tumors, enhancing or inhibiting the occurrence and development of tumors. Therefore, the present study aims to explore whether miR-155-5p can regulate autophagy and apoptosis of glioma through RICTOR. Methods The significantly differential gene miR-155-5p was identified from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo) databases GSE165937 and GSE138764 using bioinformatics analysis, and its expression was validated by quantitative real-time polymerase chain reaction (qRT-PCR). The putative target genes of miR-155-5p were predicted through interrogation of relevant databases, followed by identification of key target genes. Subsequently, core target genes were selected for functional enrichment analysis. The U87MG cell line was utilized as the experimental model and divided into Negative Control1 (NC1) group, Mimic group, Negative Control2 (NC2) group, Inhibitor group, and NC + 3-methyladenine (3-MA) group. The expression levels of miR-155-5p, RICTOR, P62, LC-3, Bax, Bcl-2, and Caspase-3 were assessed using qRT-PCR, cellular fluorescence imaging, and Western blotting; while apoptosis in the U87MG cell line was evaluated via flow cytometry. Results The results showed that miR-155-5P was highly expressed in glioma cells, which could inhibit the expression of Bax, Caspase-3, LCII/LCI and Beclin-1, and increase the expression of Bcl2 and P62. Flow cytometry and cell fluorescence were used to verify the above results. Moreover, when U87MG cells treated with miR-155-5p inhibitor were inhibited by 3-MA, the results showed that miR-155-5p enhanced the anti-apoptotic ability of U87MG cells by regulating autophagy. In addition, the bioinformatics results show that miR-155-5p survival prognosis in glioma into a strong negative correlation, while the survival prognosis of RICTOR in glioma showed a strong positive correlation. The core target genes Kyoto Encyclopedia of Genes and Genomes (KEGG) mainly occurred in PI3K-AKT signaling pathway; in addition, qRT-PCR and Western blot confirmed the regulatory effect of miR-155-5P on RICTOR. Conclusions MiR-155-5p regulates autophagy and apoptosis-related proteins in glioma cells through RICTOR, affecting the occurrence and development of glioma.
Collapse
Affiliation(s)
- Zhao Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jing-Jie Tian
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yao Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Lei Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yang Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - He-Jun Dai
- Department of Neurosurgery, Danyang Hospital Affiliated to Nantong University, Zhenjiang, China
| | - Lei Wang
- Department of Emergency Center, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yi Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai, China
- Department of Neurosurgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Cao S, Wang S, Luo H, Guo J, Xuan L, Sun L. The effect of macrophage-cardiomyocyte interactions on cardiovascular diseases and development of potential drugs. Mol Biol Rep 2024; 51:1056. [PMID: 39417949 DOI: 10.1007/s11033-024-09944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
The interaction between macrophages and cardiomyocytes plays an important role not only in maintaining cardiac homeostasis, but also in the development of many cardiovascular diseases (CVDs), such as myocardial infarction (MI) and heart failure (HF). In addition to supporting cardiomyocytes, macrophages and cardiomyocytes have a close and complex relationship. By studying their cross-talk, we can better understand novel mechanisms and target pathogenic mechanisms, and improve the treatment of CVDs. We review macrophage-cardiomyocyte communication through connexin 43 (Cx43)-containing gap junctions (GJs) directly, secreted protein factors indirectly, and discuss the implications of these interactions in cardiac homeostasis and the development of various CVDs, including MI, HF, arrhythmia, cardiac fibrosis and myocarditis. In this section, we review various drugs that work by modulating cytokines or other proteins to reduce inflammation in CVDs. The clinical findings from targeting inflammation in CVDs are also discussed. Additionally, we examine the challenges and opportunities for improving our understanding of macrophage-cardiomyocyte coupling as it relates to pathophysiological disease processes, extending our research scope, and helping identify new molecular targets and improve the effectiveness of existing therapies.
Collapse
Affiliation(s)
- Shoupeng Cao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Shengjie Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Huishan Luo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jianjun Guo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Lina Xuan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medicial University, Harbin, 157 Baojian Road, Nangang District, 150081, heilongjiang, China.
| | - Lihua Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medicial University, Harbin, 157 Baojian Road, Nangang District, 150081, heilongjiang, China.
| |
Collapse
|
18
|
Iacomino N, Tarasco MC, Berni A, Ronchi J, Mantegazza R, Cavalcante P, Foti M. Non-Coding RNAs in Myasthenia Gravis: From Immune Regulation to Personalized Medicine. Cells 2024; 13:1550. [PMID: 39329732 PMCID: PMC11430632 DOI: 10.3390/cells13181550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Myasthenia gravis (MG) is an antibody-mediated autoimmune disorder characterized by altered neuromuscular transmission, which causes weakness and fatigability in the skeletal muscles. The etiology of MG is complex, being associated with multiple genetic and environmental factors. Over recent years, progress has been made in understanding the immunological alterations implicated in the disease, but the exact pathogenesis still needs to be elucidated. A pathogenic interplay between innate immunity and autoimmunity contributes to the intra-thymic MG development. Epigenetic changes are critically involved in both innate and adaptive immune response regulation. They can act as (i) pathological factors besides genetic predisposition and (ii) co-factors contributing to disease phenotypes or patient-specific disease course/outcomes. This article reviews the role of non-coding RNAs (ncRNAs) as epigenetic factors implicated in MG. Particular attention is dedicated to microRNAs (miRNAs), whose expression is altered in MG patients' thymuses and circulating blood. The long ncRNA (lncRNA) contribution to MG, although not fully characterized yet, is also discussed. By summarizing the most recent and fast-growing findings on ncRNAs in MG, we highlight the therapeutic potential of these molecules for achieving immune regulation and their value as biomarkers for the development of personalized medicine approaches to improve disease care.
Collapse
Affiliation(s)
- Nicola Iacomino
- Neurology 4–Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (N.I.); (M.C.T.); (A.B.); (R.M.)
| | - Maria Cristina Tarasco
- Neurology 4–Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (N.I.); (M.C.T.); (A.B.); (R.M.)
- Ph.D. Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Alessia Berni
- Neurology 4–Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (N.I.); (M.C.T.); (A.B.); (R.M.)
| | - Jacopo Ronchi
- Ph.D. Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy;
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- BicOMICs, University of Milano-Bicocca, 20900 Monza, Italy
| | - Renato Mantegazza
- Neurology 4–Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (N.I.); (M.C.T.); (A.B.); (R.M.)
| | - Paola Cavalcante
- Neurology 4–Neuroimmunology and Neuromuscolar Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (N.I.); (M.C.T.); (A.B.); (R.M.)
| | - Maria Foti
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- BicOMICs, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
19
|
Espina-Ordoñez M, Balderas-Martínez YI, Torres-Machorro AL, Herrera I, Maldonado M, Romero Y, Toscano-Marquez F, Pardo A, Selman M, Cisneros J. Mir-155-5p targets TP53INP1 to promote proliferative phenotype in hypersensitivity pneumonitis lung fibroblasts. Noncoding RNA Res 2024; 9:865-875. [PMID: 38586316 PMCID: PMC10997802 DOI: 10.1016/j.ncrna.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/11/2024] [Accepted: 02/19/2024] [Indexed: 04/09/2024] Open
Abstract
Background Hypersensitivity pneumonitis (HP) is an inflammatory disorder affecting lung parenchyma and often evolves into fibrosis (fHP). The altered regulation of genes involved in the pathogenesis of the disease is not well comprehended, while the role of microRNAs in lung fibroblasts remains unexplored. Methods We used integrated bulk RNA-Seq and enrichment pathway bioinformatic analyses to identify differentially expressed (DE)-miRNAs and genes (DEGs) associated with HP lungs. In vitro, we evaluated the expression and potential role of miR-155-5p in the phenotype of fHP lung fibroblasts. Loss and gain assays were used to demonstrate the impact of miR-155-5p on fibroblast functions. In addition, mir-155-5p and its target TP53INP1 were analyzed after treatment with TGF-β, IL-4, and IL-17A. Results We found around 50 DEGs shared by several databases that differentiate HP from control and IPF lungs, constituting a unique HP lung transcriptional signature. Additionally, we reveal 18 DE-miRNAs that may regulate these DEGs. Among the candidates likely associated with HP pathogenesis was miR-155-5p. Our findings indicate that increased miR-155-5p in fHP fibroblasts coincides with reduced TP53INP1 expression, high proliferative capacity, and a lack of senescence markers compared to IPF fibroblasts. Induced overexpression of miR-155-5p in normal fibroblasts remarkably increases the proliferation rate and decreases TP53INP1 expression. Conversely, miR-155-5p inhibition reduces proliferation and increases senescence markers. TGF-β, IL-4, and IL-17A stimulated miR-155-5p overexpression in HP lung fibroblasts. Conclusion Our findings suggest a distinctive signature of 53 DEGs in HP, including CLDN18, EEF2, CXCL9, PLA2G2D, and ZNF683, as potential targets for future studies. Likewise, 18 miRNAs, including miR-155-5p, could be helpful to establish differences between these two pathologies. The overexpression of miR-155-5p and downregulation of TP53INP1 in fHP lung fibroblasts may be involved in his proliferative and profibrotic phenotype. These findings may help differentiate and characterize their pathogenic features and understand their role in the disease.
Collapse
Affiliation(s)
- Marco Espina-Ordoñez
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, Piso 1, Circuito de Posgrados, Ciudad Universidad, Coyoacán, C.P 04510, CDMX, Mexico
| | - Yalbi Itzel Balderas-Martínez
- Laboratorio de Biología Computacional, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| | - Ana Lilia Torres-Machorro
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| | - Iliana Herrera
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| | - Mariel Maldonado
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| | - Yair Romero
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Fernanda Toscano-Marquez
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Moisés Selman
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| | - José Cisneros
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, 14080, Mexico
| |
Collapse
|
20
|
Maqbool M, Hussain MS, Shaikh NK, Sultana A, Bisht AS, Agrawal M. Noncoding RNAs in the COVID-19 Saga: An Untold Story. Viral Immunol 2024; 37:269-286. [PMID: 38968365 DOI: 10.1089/vim.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024] Open
Affiliation(s)
- Mudasir Maqbool
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Nusrat K Shaikh
- Department of Quality Assurance, Smt. N. M. Padalia Pharmacy College, Ahmedabad, India
| | - Ayesha Sultana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya University, Mangalore, India
| | - Ajay Singh Bisht
- Shri Guru Ram Rai University School of Pharmaceutical Sciences, Dehradun, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K. R. Mangalam University, Gurugram, India
| |
Collapse
|
21
|
Aghajani Mir M. Illuminating the pathogenic role of SARS-CoV-2: Insights into competing endogenous RNAs (ceRNAs) regulatory networks. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 122:105613. [PMID: 38844190 DOI: 10.1016/j.meegid.2024.105613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
The appearance of SARS-CoV-2 in 2019 triggered a significant economic and health crisis worldwide, with heterogeneous molecular mechanisms that contribute to its development are not yet fully understood. Although substantial progress has been made in elucidating the mechanisms behind SARS-CoV-2 infection and therapy, it continues to rank among the top three global causes of mortality due to infectious illnesses. Non-coding RNAs (ncRNAs), being integral components across nearly all biological processes, demonstrate effective importance in viral pathogenesis. Regarding viral infections, ncRNAs have demonstrated their ability to modulate host reactions, viral replication, and host-pathogen interactions. However, the complex interactions of different types of ncRNAs in the progression of COVID-19 remains understudied. In recent years, a novel mechanism of post-transcriptional gene regulation known as "competing endogenous RNA (ceRNA)" has been proposed. Long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and viral ncRNAs function as ceRNAs, influencing the expression of associated genes by sequestering shared microRNAs. Recent research on SARS-CoV-2 has revealed that disruptions in specific ceRNA regulatory networks (ceRNETs) contribute to the abnormal expression of key infection-related genes and the establishment of distinctive infection characteristics. These findings present new opportunities to delve deeper into the underlying mechanisms of SARS-CoV-2 pathogenesis, offering potential biomarkers and therapeutic targets. This progress paves the way for a more comprehensive understanding of ceRNETs, shedding light on the intricate mechanisms involved. Further exploration of these mechanisms holds promise for enhancing our ability to prevent viral infections and develop effective antiviral treatments.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
22
|
Toropko M, Chuvpilo S, Karabelsky A. miRNA-Mediated Mechanisms in the Generation of Effective and Safe Oncolytic Viruses. Pharmaceutics 2024; 16:986. [PMID: 39204331 PMCID: PMC11360794 DOI: 10.3390/pharmaceutics16080986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression by inhibiting the translation of target transcripts. The expression profiles of miRNAs vary in different tissues and change with the development of diseases, including cancer. This feature has begun to be used for the modification of oncolytic viruses (OVs) in order to increase their selectivity and efficacy. OVs represent a relatively new class of anticancer drugs; they are designed to replicate in cancer tumors and destroy them. These can be natural viruses that can replicate within cancer tumor cells, or recombinant viruses created in laboratories. There are some concerns regarding OVs' toxicity, due to their ability to partially replicate in healthy tissues. In addition, lytic and immunological responses upon OV therapy are not always sufficient, so various OV editing methods are used. This review discusses the latest results of preclinical and clinical studies of OVs, modifications of which are associated with the miRNA-mediated mechanism of gene silencing.
Collapse
Affiliation(s)
- Mariia Toropko
- Gene Therapy Department, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia; (S.C.); (A.K.)
| | | | | |
Collapse
|
23
|
Wystrychowski G, Simka-Lampa K, Witkowska A, Sobecko E, Skubis-Sikora A, Sikora B, Wojtyna E, Golda A, Gwizdek K, Wróbel M, Sędek Ł, Górczyńska-Kosiorz S, Szweda-Gandor N, Trautsolt W, Francuz T, Kruszniewska-Rajs C, Gola J. Selected microRNA Expression and Protein Regulator Secretion by Adipose Tissue-Derived Mesenchymal Stem Cells and Metabolic Syndrome. Int J Mol Sci 2024; 25:6644. [PMID: 38928349 PMCID: PMC11204268 DOI: 10.3390/ijms25126644] [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: 05/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The role of adipose mesenchymal stem cells (Ad-MSCs) in metabolic syndrome remains unclear. We aimed to assess the expression of selected microRNAs in Ad-MSCs of non-diabetic adults in relation to Ad-MSC secretion of protein regulators and basic metabolic parameters. Ten obese, eight overweight, and five normal weight subjects were enrolled: 19 females and 4 males; aged 43.0 ± 8.9 years. Ad-MSCs were harvested from abdominal subcutaneous fat. Ad-MSC cellular expressions of four microRNAs (2-ΔCt values) and concentrations of IL-6, IL-10, VEGF, and IGF-1 in the Ad-MSC-conditioned medium were assessed. The expressions of miR-21, miR-122, or miR-192 did not correlate with clinical parameters (age, sex, BMI, visceral fat, HOMA-IR, fasting glycemia, HbA1c, serum lipids, CRP, and eGFR). Conversely, the expression of miR-155 was lowest in obese subjects (3.69 ± 2.67 × 10-3 vs. 7.07 ± 4.42 × 10-3 in overweight and 10.25 ± 7.05 × 10-3 in normal weight ones, p = 0.04). The expression of miR-155 correlated inversely with BMI (sex-adjusted r = -0.64; p < 0.01), visceral adiposity (r = -0.49; p = 0.03), and serum CRP (r = -0.63; p < 0.01), whereas it correlated positively with serum HDL cholesterol (r = 0.51; p = 0.02). Moreover, miR-155 synthesis was associated marginally negatively with Ad-MSC secretion of IGF-1 (r = -0.42; p = 0.05), and positively with that of IL-10 (r = 0.40; p = 0.06). Ad-MSC expression of miR-155 appears blunted in visceral obesity, which correlates with Ad-MSC IGF-1 hypersecretion and IL-10 hyposecretion, systemic microinflammation, and HDL dyslipidemia. Ad-MSC studies in metabolic syndrome should focus on miR-155.
Collapse
Affiliation(s)
| | - Klaudia Simka-Lampa
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | | | - Ewelina Sobecko
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | - Aleksandra Skubis-Sikora
- Department of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.S.-S.); (B.S.)
| | - Bartosz Sikora
- Department of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.S.-S.); (B.S.)
| | - Ewa Wojtyna
- Institute of Medical Sciences, University of Opole, 45-040 Opole, Poland;
| | - Agnieszka Golda
- Alfamed General Practice, 41-100 Siemianowice Slaskie, Poland;
| | - Katarzyna Gwizdek
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Marta Wróbel
- Department of Internal Medicine, Diabetology and Cardiometabolic Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Łukasz Sędek
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Sylwia Górczyńska-Kosiorz
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Nikola Szweda-Gandor
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Wanda Trautsolt
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | - Celina Kruszniewska-Rajs
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (C.K.-R.); (J.G.)
| | - Joanna Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (C.K.-R.); (J.G.)
| |
Collapse
|
24
|
Shahin H, Belcastro L, Das J, Perdiki Grigoriadi M, Saager RB, Steinvall I, Sjöberg F, Olofsson P, Elmasry M, El-Serafi AT. MicroRNA-155 mediates multiple gene regulations pertinent to the role of human adipose-derived mesenchymal stem cells in skin regeneration. Front Bioeng Biotechnol 2024; 12:1328504. [PMID: 38562669 PMCID: PMC10982420 DOI: 10.3389/fbioe.2024.1328504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction: The role of Adipose-derived mesenchymal stem cells (AD-MSCs) in skin wound healing remains to be fully characterized. This study aims to evaluate the regenerative potential of autologous AD-MSCs in a non-healing porcine wound model, in addition to elucidate key miRNA-mediated epigenetic regulations that underlie the regenerative potential of AD-MSCs in wounds. Methods: The regenerative potential of autologous AD-MSCs was evaluated in porcine model using histopathology and spatial frequency domain imaging. Then, the correlations between miRNAs and proteins of AD-MSCs were evaluated using an integration analysis in primary human AD-MSCs in comparison to primary human keratinocytes. Transfection study of AD-MSCs was conducted to validate the bioinformatics data. Results: Autologous porcine AD-MSCs improved wound epithelialization and skin properties in comparison to control wounds. We identified 26 proteins upregulated in human AD-MSCs, including growth and angiogenic factors, chemokines and inflammatory cytokines. Pathway enrichment analysis highlighted cell signalling-associated pathways and immunomodulatory pathways. miRNA-target modelling revealed regulations related to genes encoding for 16 upregulated proteins. miR-155-5p was predicted to regulate Fibroblast growth factor 2 and 7, C-C motif chemokine ligand 2 and Vascular cell adhesion molecule 1. Transfecting human AD-MSCs cell line with anti-miR-155 showed transient gene silencing of the four proteins at 24 h post-transfection. Discussion: This study proposes a positive miR-155-mediated gene regulation of key factors involved in wound healing. The study represents a promising approach for miRNA-based and cell-free regenerative treatment for difficult-to-heal wounds. The therapeutic potential of miR-155 and its identified targets should be further explored in-vivo.
Collapse
Affiliation(s)
- Hady Shahin
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
- Faculty of Biotechnology, Modern Sciences and Arts University, October City, Cairo, Egypt
| | - Luigi Belcastro
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Jyotirmoy Das
- Bioinformatics Unit, Core Facility (KEF), Faculty of Medicine and Health Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, SciLife Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Rolf B. Saager
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Ingrid Steinvall
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Folke Sjöberg
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| | - Pia Olofsson
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Moustafa Elmasry
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Ahmed T. El-Serafi
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| |
Collapse
|
25
|
Krause C, Bergmann E, Schmidt SV. Epigenetic modulation of myeloid cell functions in HIV and SARS-CoV-2 infection. Mol Biol Rep 2024; 51:342. [PMID: 38400997 PMCID: PMC10894183 DOI: 10.1007/s11033-024-09266-2] [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: 11/29/2023] [Accepted: 01/18/2024] [Indexed: 02/26/2024]
Abstract
Myeloid cells play a vital role in innate immune responses as they recognize and phagocytose pathogens like viruses, present antigens, produce cytokines, recruit other immune cells to combat infections, and contribute to the attenuation of immune responses to restore homeostasis. Signal integration by pathogen recognition receptors enables myeloid cells to adapt their functions by a network of transcription factors and chromatin remodelers. This review provides a brief overview of the subtypes of myeloid cells and the main epigenetic regulation mechanisms. Special focus is placed on the epigenomic alterations in viral nucleic acids of HIV and SARS-CoV-2 along with the epigenetic changes in the host's myeloid cell compartment. These changes are important as they lead to immune suppression and promote the progression of the disease. Finally, we highlight some promising examples of 'epidrugs' that modulate the epigenome of immune cells and could be used as therapeutics for viral infections.
Collapse
Affiliation(s)
- Carolyn Krause
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- Department of Microbiology and Immunology, the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Eva Bergmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Susanne Viktoria Schmidt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
26
|
Zhang L, Zhang C, Zheng J, Wang Y, Wei X, Yang Y, Zhao Q. miR-155-5p/Bmal1 Modulates the Senescence and Osteogenic Differentiation of Mouse BMSCs through the Hippo Signaling Pathway. Stem Cell Rev Rep 2024; 20:554-567. [PMID: 38150082 PMCID: PMC10837250 DOI: 10.1007/s12015-023-10666-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND The core clock gene brain and muscle ARNT like-1 (Bmal1) is involved in the regulation of bone tissue aging. However, current studies are mostly limited to the establishment of the association between Bmal1 and bone senescence, without in-depth exploration of its main upstream and downstream regulatory mechanisms. METHODS The luciferase reporter assay, RT-qPCR and Western blotting were performed to detect the interaction between miR-155-5p and Bmal1. The effects of miR-155-5p and Bmal1 on the aging and osteogenic differentiation ability of mouse bone marrow mesenchymal stem cells (BMSCs) were investigated by cell counting kit-8 (CCK-8) assay, flow cytometry, β-gal staining, alkaline phosphatase quantitative assay and alizarin red staining in vitro. The potential molecular mechanism was identified by ChIP-Seq, RNA-seq database analysis and immunofluorescence staining. RESULTS The expression of Bmal1 declined with age, while the miR-155-5p was increased. miR-155-5p and Bmal1 repressed each other's expression, and miR-155-5p targeted the Bmal1. Besides, miR-155-5p inhibited the proliferation and osteogenic differentiation of BMSCs, promoted cell apoptosis and senescence, inhibited the expression and nuclear translocation of YAP and TAZ. However, Bmal1 facilitated the osteogenic differentiation and suppressed the aging of BMSCs, meanwhile inactivated the Hippo pathway. Moreover, YAP inhibitors abrogated the positive regulation of aging and osteogenic differentiation in BMSCs by miR-155-5p and Bmal1. CONCLUSION In mouse BMSCs, miR-155-5p and Bmal1 regulated the aging and osteogenic differentiation ability of BMSCs mainly through the Hippo signaling pathway. Our findings provide new insights for the interventions in bone aging.
Collapse
Affiliation(s)
- Lanxin Zhang
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Chengxiaoxue Zhang
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Jiawen Zheng
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Yuhong Wang
- Department of Stomatology, West China Fourth Hospital, Sichuan University, 18, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Xiaoyu Wei
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Yuqing Yang
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Qing Zhao
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China.
| |
Collapse
|
27
|
Peng L, Chen S, Lin H, Wan C, Li X, Xu S, Li S. Bisphenol A exposure exacerbates tracheal inflammatory injury in selenium-deficient chickens by regulating the miR-155/TRAF3/ROS pathway. Int J Biol Macromol 2023; 253:127501. [PMID: 37866585 DOI: 10.1016/j.ijbiomac.2023.127501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Abstract
Bisphenol A (BPA) is an endocrine disruptor. Excessive BPA intake can damage the structure and function of the respiratory tract. Dietary selenium (Se) deficiency may also cause immune tissue damage. To investigate the potential mechanism of BPA on tracheal damage in selenium-deficient chickens and the role of microRNAs (miRNAs) in this process, we established in vitro and in vivo Se deficiency and BPA exposure models and screened out miR-155 for follow-up experiments. We further predicted and confirmed the targeting relationship between miR-155 and TRAF3 using TargetScan and dual luciferase assays and found that miR-155 was highly expressed and caused inflammatory damage. Further studies showed that BPA exposure increased airway oxidative stress, activated the NF-κB pathway, and caused inflammation and immune damage in selenium-deficient chickens, but down-regulating miR-155 and NAC treatment could reverse this phenomenon. This suggested that these pathways are regulated by the miR-155/TRAF3/ROS axis. In conclusion, BPA exposure aggravates airway inflammation in selenium-deficient chickens by regulating miR-155/TRAF3/ROS. This study revealed the mechanism of BPA exposure combined with Se deficiency in tracheal inflammatory injury in chickens and enriched the theoretical basis of BPA injury in poultry.
Collapse
Affiliation(s)
- Lin Peng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shasha Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Chunyan Wan
- National Selenium-rich Product Quality Supervision and Inspection Center, Enshi 445000, PR China
| | - Xiang Li
- National Selenium-rich Product Quality Supervision and Inspection Center, Enshi 445000, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
28
|
Gu F, Huang X, Huang W, Zhao M, Zheng H, Wang Y, Chen R. The role of miRNAs in Behçet's disease. Front Immunol 2023; 14:1249826. [PMID: 37860009 PMCID: PMC10584330 DOI: 10.3389/fimmu.2023.1249826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/12/2023] [Indexed: 10/21/2023] Open
Abstract
The symptoms of Behçet's disease (BD), a multisystemic condition with autoimmune and inflammation as hallmarks, include arthritis, recurring oral and vaginal ulcers, skin rashes and lesions, and involvement of the nervous, gastrointestinal, and vascular systems. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), may be important regulators of inflammation and autoimmune disease. These ncRNAs are essential to the physiological and pathophysiological disease course, and miRNA in particular has received significant attention for its role and function in BD and its potential use as a diagnostic biomarker in recent years. Although promising as therapeutic targets, miRNAs must be studied further to fully comprehend how miRNAs in BD act biologically.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuanyin Wang
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, China
| | - Ran Chen
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, China
| |
Collapse
|
29
|
Moutabian H, Radi UK, Saleman AY, Adil M, Zabibah RS, Chaitanya MNL, Saadh MJ, Jawad MJ, Hazrati E, Bagheri H, Pal RS, Akhavan-Sigari R. MicroRNA-155 and cancer metastasis: Regulation of invasion, migration, and epithelial-to-mesenchymal transition. Pathol Res Pract 2023; 250:154789. [PMID: 37741138 DOI: 10.1016/j.prp.2023.154789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/25/2023]
Abstract
Among the leading causes of death globally has been cancer. Nearly 90% of all cancer-related fatalities are attributed to metastasis, which is the growing of additional malignant growths out of the original cancer origin. Therefore, a significant clinical need for a deeper comprehension of metastasis exists. Beginning investigations are being made on the function of microRNAs (miRNAs) in the metastatic process. Tiny non-coding RNAs called miRNAs have a crucial part in controlling the spread of cancer. Some miRNAs regulate migration, invasion, colonization, cancer stem cells' properties, the epithelial-mesenchymal transition (EMT), and the microenvironment, among other processes, to either promote or prevent metastasis. One of the most well-conserved and versatile miRNAs, miR-155 is primarily distinguished by overexpression in a variety of illnesses, including malignant tumors. It has been discovered that altered miR-155 expression is connected to a number of physiological and pathological processes, including metastasis. As a result, miR-155-mediated signaling pathways were identified as possible cancer molecular therapy targets. The current research on miR-155, which is important in controlling cancer cells' invasion, and metastasis as well as migration, will be summarized in the current work. The crucial significance of the lncRNA/circRNA-miR-155-mRNA network as a crucial regulator of carcinogenesis and a player in the regulation of signaling pathways or related genes implicated in cancer metastasis will be covered in the final section. These might provide light on the creation of fresh treatment plans for controlling cancer metastasis.
Collapse
Affiliation(s)
- Hossein Moutabian
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Mv N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | | | - Ebrahi Hazrati
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rashmi Saxena Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
30
|
Fan H, Liu S, Jiao B, Liang X. Low‑dose ionizing radiation attenuates high glucose‑induced hepatic apoptosis and immune factor release via modulation of a miR‑155‑SOCS1 axis. Mol Med Rep 2023; 28:171. [PMID: 37503757 PMCID: PMC10433713 DOI: 10.3892/mmr.2023.13058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Abstract
Diabetic liver injury (DLI) can result in several diseases of the liver, including steatohepatitis, liver fibrosis, cirrhosis, and liver cancer. Low‑dose ionizing radiation (LDIR) has hormetic effects in normal/disease conditions. However, whether LDIR has a beneficial effect on DLI has not been assessed previously. MicroRNA (miR)‑155 and its target gene suppressor of cytokine signaling 1 (SOCS1) play critical roles in modulating hepatic proliferation, apoptosis, and immunity. However, whether a miR‑155‑SOCS1 axis is involved in high glucose (HG) induced hepatic damage remains to be determined. In the present study, mouse hepatocyte AML12 cells were treated with 30 mM glucose (HG), 75 mGy X‑ray (LDIR), or HG plus LDIR. The expression levels of miR‑155 and SOCS1 were determined by reverse transcription‑quantitative PCR and western blotting. Additionally, apoptosis was measured using flow cytometry. The release of inflammatory factors, including TNF‑α, IL‑1β, IL‑6, IL‑10, and IFN‑γ, after HG and/or LDIR treatment was detected by ELISA. The results showed that HG may induce hepatic apoptosis by upregulating the levels of miR‑155 and downregulating the levels of SOCS1. HG also stimulated the secretion of TNF‑α, IL‑1β, IL‑6, and IL‑10. However, LDIR blocked the HG‑induced activation of a miR‑155‑SOCS1 axis and suppressed the release of inflammatory factors. These results indicated that a miR‑155‑SOCS1 axis plays a role in HG‑induced liver injury, and LDIR may exert a hepatoprotective effect by regulating the miR‑155‑SOCS1 axis.
Collapse
Affiliation(s)
- Hongqiong Fan
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shanshan Liu
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Benzheng Jiao
- Department of Nuclear Medicine, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xinyue Liang
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
31
|
Figueroa MAC, Lujambio IM, Gutiérrez TA, Hernández MFP, Ramírez EYE, Guzmán DJ, Sánchez MFL, Morales HFG, Samudio HJG, Sánchez FS, Flores MD, Zamarripa CAJ, Mendoza CCC, Hernández MEO, Velázquez CMO, Flores MS, Orozco DVH, Moreno GYC, Cruz M, de Jesús Peralta Romero J. Association of the rs5186 polymorphism of the AGTR1 gene with decreased eGFR in patients with type 2 diabetes from Mexico City. Nefrologia 2023; 43:546-561. [PMID: 37996337 DOI: 10.1016/j.nefroe.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/10/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Early biomarkers search for Diabetic Kidney Disease (DKD) in patients with Type 2 Diabetes Mellitus (T2DM), as genetic markers to identify vulnerable carriers of the disease even before Glomerular Filtration Rate (GFR) decline or microalbuminuria development, has been relevant during the last few years. The rs5186 (A116C) polymorphism of the Angiotensin II Receptor Type I gene (AGTR1), has been associated to multiple effects of renal injury risk, commonly detected in patients with Diabetes Mellitus (DM). It has been described that rs5186 could have an effect in stability proteins that assemble Angiotensin II Receptor Type I (AT1), modifying its action, which is why it should be considered as a risk factor for Chronic Kidney Disease (CKD), characterized by a GFR progressive reduction. Even though, the association between rs5186 AGTR1 gene polymorphism and DKD in patients with T2DM has been controversial, inconclusive, and even absent. This disputable issue might be as a result of association studies in which many and varied clinical phenotypes included are contemplated as CKD inductors and enhancers. Although, the sample sizes studied in patients with T2DM are undersized and did not have a strict inclusion criteria, lacking of biochemical markers or KDOQI classification, which have hindered its examination. OBJECTIVE The aim of our study was to establish an association between rs5186 AGTR1 gene polymorphism and GFR depletion, assessed as a risk factor to DKD development in patients with T2DM. METHODS We analyzed 297 not related patients with T2DM, divided into 221 controls (KDOQI 1) and 76 cases (KDOQI 2). Arterial pressure, anthropometric and biochemical parameters were measured. rs5186 of AGTR1 genotyping was performed by TaqMan assay real-time PCR method. Allele and genotype frequencies, and Hardy-Weinberg equilibrium were measured. Normality test for data distribution was analyzed by Shapiro-Wilk test, variable comparison by Student's t-test for continuous variables, and Chi-squared test for categorical variables; ANOVA test was used for mean comparison of more than two groups. Effect of rs5186 to DKD was estimated by multiple heritability adjustment models for risk variables of DKD. Statistical significance was indicated by p<0.05. Data was analyzed using Statistical Package STATA v11 software. RESULTS Dominant and Over-dominant models showed a likelihood ratio to GFR depletion of 1.89 (1.05-3.39, p=0.031) and 2.01 (1.08-3.73, p=0.023) in patients with T2DM. Risk factor increased to 2.54 (1.10-5.89) in women in Over-dominant model. CONCLUSION In clinical practice, most of nephropathies progress at a slow pace into a total breakdown of renal function, even asymptomatic. This is the first study, reporting that rs5186 polymorphism of AGTR1 gene contribution to GFR depletion, and this could be evaluated as a predisposing factor for DKD in patients with T2DM.
Collapse
Affiliation(s)
- Manuel Alejandro Contreras Figueroa
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - Irene Mendoza Lujambio
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - Teresa Alvarado Gutiérrez
- Coordinación Clínica de Educación e Investigación en Salud de la Unidad de Medicina Familiar 31, Instituto Mexicano del Seguro Social, Delegación sur, Ciudad de México, México
| | - María Fernanda Pérez Hernández
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México; Red de Medicina Para la Educación, el Desarrollo y la Investigación Científica de Iztacala. MEDICI, Facultad de Estudios Superiores Iztacala, UNAM, Estado de México, México
| | - Evelyn Yazmín Estrada Ramírez
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Departamento de Nefrología del Hospital de Especialidades "Dr. Antonio Fraga Mouret", CMN La Raza, IMSS, Ciudad de México, México
| | - Dominga Jiménez Guzmán
- Departamento de Nefrología del Hospital de Especialidades "Dr. Bernardo Sepúlveda" CMN Siglo XXI, IMSS, Ciudad de México, México; Jefatura de la Unidad de Consulta Externa de la UMAE, Hospital de Alta Especialidad Médica "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México
| | - María Fernanda Lucas Sánchez
- Secretaría de Enseñanza Clínica, Internado y Servicio Social. Facultad de Medicina UNAM, Ciudad de México, México; Becaria de la Dirección General de Calidad y Educación en Salud, Secretaría de Salud, México
| | - Hannia Fernanda González Morales
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Red de Medicina Para la Educación, el Desarrollo y la Investigación Científica de Iztacala. MEDICI, Facultad de Estudios Superiores Iztacala, UNAM, Estado de México, México
| | - Héctor Jaime Gómez Samudio
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México
| | - Fernando Suarez Sánchez
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México
| | - Margarita Díaz Flores
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México
| | - Carlos Alberto Jiménez Zamarripa
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - Claudia Camelia Calzada Mendoza
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - María Esther Ocharán Hernández
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - Cora Mariana Orozco Velázquez
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Secretaría de Enseñanza Clínica, Internado y Servicio Social. Facultad de Medicina UNAM, Ciudad de México, México
| | - Mariana Soto Flores
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Departamento de Formación Integral e Institucional, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México, México
| | - Daniela Vicenta Hernández Orozco
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México; Departamento de Formación Integral e Institucional, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México, México
| | - Gabriela Yanet Cortés Moreno
- Coordinación Nacional de Investigación, Subdirección de Servicios de salud de Petróleos Mexicanos, PEMEX, Ciudad de México, México
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México
| | - José de Jesús Peralta Romero
- Unidad de Investigación Médica en Bioquímica, Unidad Médica de Alta Especialidad "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, México.
| |
Collapse
|
32
|
Link J, Thon C, Petkevicius V, Steponaitiene R, Malfertheiner P, Kupcinskas J, Link A. The Translational Impact of Plant-Derived Xeno-miRNA miR-168 in Gastrointestinal Cancers and Preneoplastic Conditions. Diagnostics (Basel) 2023; 13:2701. [PMID: 37627960 PMCID: PMC10453613 DOI: 10.3390/diagnostics13162701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/30/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION Diet is one of the most important factors contributing to the multistep process of carcinogenesis. The clinical relevance of exogenous food-derived xeno-microRNAs (miRNAs) in human diseases is poorly understood. In this study, we aimed to evaluate the potential clinical relevance of the xeno-miRNA miR-168 in the gastric mucosa along the preneoplastic conditions and gastric carcinogenesis. METHODS For a systematic analysis, we included stomach tissues from patients with different pathologies, including normal mucosa (N), chronic non-atrophic (CNAG) and atrophic gastritis (CAG) and intestinal metaplasia (IM) (n = 72), matched non-tumorous (NT) and tumorous (T) gastric cancer (GC) tissues (n = 81), matched colorectal cancer (CRC) tissues (n = 40), and colon mucosa and faeces from controls and IBD patients. RESULTS miR-168 was reproducibly detectable in all samples studied, with the highest levels in the proximal upper GI and in non-tumorous compared to tumorous tissues in both GC and CRC. There was no difference related to H. pylori positivity or inflammation grade, while higher miR-168 levels were observed in patients with moderate or severe AG/IM or OLGIM3/4. Survival analysis showed only a small, non-significant trend towards worse overall survival for patients with the highest to lowest miR-168 levels, while no differences were related to Lauren's classification. CONCLUSIONS Food-derived xeno miRNAs are reproducibly detectable in the gastric and colonic mucosa. Although the clinically relevant function remains to be elucidated, higher levels of miR-168 in patients with moderate and severe IM merit further investigation.
Collapse
Affiliation(s)
- Jastin Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, 39120 Magdeburg, Germany (C.T.); (P.M.)
| | - Cosima Thon
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, 39120 Magdeburg, Germany (C.T.); (P.M.)
| | - Vytenis Petkevicius
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (V.P.); (R.S.); (J.K.)
| | - Ruta Steponaitiene
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (V.P.); (R.S.); (J.K.)
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, 39120 Magdeburg, Germany (C.T.); (P.M.)
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, 80539 Munich, Germany
| | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (V.P.); (R.S.); (J.K.)
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, 39120 Magdeburg, Germany (C.T.); (P.M.)
| |
Collapse
|
33
|
Hukowska-Szematowicz B, Ostrycharz E, Dudzińska W, Roszkowska P, Siennicka A, Wojciechowska-Koszko I. Digital PCR (dPCR) Quantification of miR-155-5p as a Potential Candidate for a Tissue Biomarker of Inflammation in Rabbits Infected with Lagovirus europaeus/Rabbit Hemorrhagic Disease Virus (RHDV). Viruses 2023; 15:1578. [PMID: 37515264 PMCID: PMC10386091 DOI: 10.3390/v15071578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/07/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
MicroRNAs (miRNAs, miRs) are a group of small, 17-25 nucleotide, non-coding RNA sequences that, in their mature form, regulate gene expression at the post-transcriptional level. They participate in many physiological and pathological processes in both humans and animals. One such process is viral infection, in which miR-155 participates in innate and adaptive immune responses to a broad range of inflammatory mediators. Recently, the study of microRNA has become an interesting field of research as a potential candidate for biomarkers for various processes and disease. To use miRNAs as potential biomarkers of inflammation in viral diseases of animals and humans, it is necessary to improve their detection and quantification. In a previous study, using reverse transcription real-time quantitative PCR (RT-qPCR), we showed that the expression of ocu-miR-155-5p in liver tissue was significantly higher in rabbits infected with Lagovirus europaeus/Rabbit Hemorrhagic Disease Virus (RHDV) compared to healthy rabbits. The results indicated a role for ocu-miR-155-5p in Lagovirus europaeus/RHDV infection and reflected hepatitis and the impairment/dysfunction of this organ during RHD. MiR-155-5p was, therefore, hypothesized as a potential candidate for a tissue biomarker of inflammation and examined in tissues in Lagovirus europaeus/RHDV infection by dPCR. The objective of the study is the absolute quantification of ocu-miR-155-5p in four tissues (liver, lung, kidney, and spleen) of rabbits infected with Lagovirus europaeus/RHDV by digital PCR, a robust technique for the precise and direct quantification of small amounts of nucleic acids, including miRNAs, without standard curves and external references. The average copy number/µL (copies/µL) of ocu-miRNA-155-5p in rabbits infected with Lagovirus europaeus GI.1a/Rossi in the liver tissue was 12.26 ± 0.14, that in the lung tissue was 48.90 ± 9.23, that in the kidney tissue was 16.92 ± 2.89, and that in the spleen was 25.10 ± 0.90. In contrast, in the tissues of healthy control rabbits, the average number of copies/µL of ocu-miRNA-155-5p was 5.07 ± 1.10 for the liver, 23.52 ± 2.77 for lungs, 8.10 ± 0.86 for kidneys, and 42.12 ± 3.68 for the spleen. The increased expression of ocu-miRNA-155-5p in infected rabbits was demonstrated in the liver (a fold-change of 2.4, p-value = 0.0003), lung (a fold-change of 2.1, p-value = 0.03), and kidneys (a fold-change of 2.1, p-value = 0.01), with a decrease in the spleen (a fold-change of 0.6, p-value = 0.002). In the study of Lagovirus europaeus/RHDV infection and in the context of viral infections, this is the first report that shows the potential use of dPCR for the sensitive and absolute quantification of microRNA-155-5p in tissues during viral infection. We think miR-155-5p may be a potential candidate for a tissue biomarker of inflammation with Lagovirus europaeus/RHDV infection. Our report presents a new path in discovering potential candidates for the tissue biomarkers of inflammation.
Collapse
Affiliation(s)
- Beata Hukowska-Szematowicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, 71-412 Szczecin, Poland
| | - Ewa Ostrycharz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, 71-412 Szczecin, Poland
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Wioleta Dudzińska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, Żołnierska 54, 71-210 Szczecin, Poland
| | - Paulina Roszkowska
- Department of Diagnostic Immunology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Aldona Siennicka
- Department of Laboratory Diagnostics, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Iwona Wojciechowska-Koszko
- Department of Diagnostic Immunology, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
34
|
Kern AE, Ortmayr G, Assinger A, Starlinger P. The role of microRNAs in the different phases of liver regeneration. Expert Rev Gastroenterol Hepatol 2023; 17:959-973. [PMID: 37811642 DOI: 10.1080/17474124.2023.2267422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
INTRODUCTION Since the first discovery of microRNAs (miRs) extensive evidence reveals their indispensable role in different patho-physiological processes. They are recognized as critical regulators of hepatic regeneration, as they modulate multiple complex signaling pathways affecting liver regeneration. MiR-related translational suppression and degradation of target mRNAs and proteins are not limited to one specific gene, but act on multiple targets. AREAS COVERED In this review, we are going to explore the role of miRs in the context of liver regeneration and discuss the regulatory effects attributed to specific miRs. Moreover, specific pathways crucial for liver regeneration will be discussed, with a particular emphasis on the involvement of miRs within the respective signaling cascades. EXPERT OPINION The considerable amount of studies exploring miR functions in a variety of diseases paved the way for the development of miR-directed therapeutics. Clinical implementation has already shown promising results, but additional research is warranted to assure safe and efficient delivery. Nevertheless, given the broad functional properties of miRs and their critical involvement during hepatic regeneration, they represent an attractive treatment target to promote liver recovery after hepatic resection.
Collapse
Affiliation(s)
- Anna Emilia Kern
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Gregor Ortmayr
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Patrick Starlinger
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, MN, USA
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Mehdipour M, Shahidi M, Anbari F, Mirzaei H, Jafari S, Kholghi A, Lotfi E, Manifar S, Mashhadiabbas F. Salivary level of microRNA-146a and microRNA-155 biomarkers in patients with oral lichen planus versus oral squamous cell carcinoma. BMC Oral Health 2023; 23:433. [PMID: 37386382 PMCID: PMC10308694 DOI: 10.1186/s12903-023-03155-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 06/20/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Oral lichen planus (OLP) is a chronic inflammatory disease of the oral mucosa, which has potential for malignant transformation. MicroRNAs play an important role in immunopathogenesis of OLP, and may be used for prediction of its malignant transformation. This study aimed to assess the salivary level of microRNA-146a and microRNA-155 biomarkers in patients with OLP and oral squamous cell carcinoma (OSCC). METHODS In this case-control study, unstimulated saliva samples were collected from 60 patients, including 15 patients with dysplastic OLP, 15 OLP patients without dysplasia, 15 patients with OSCC, and 15 healthy controls according to the Navazesh technique. After RNA extraction, the expression of microRNA-146a and microRNA-155 was quantified by real-time quantitative polymerase chain reaction (RT-qPCR). The data were analyzed by the Kruskal-Wallis and Dunn-Bonferroni tests. RESULTS The difference in expression of microRNA-146a and microRNA-155 among the four groups was significant (P < 0.05). Pairwise comparisons of the groups showed significantly higher expression of microRNA-146a in OLP (P = 0.004) and dysplastic OLP (P = 0.046) patients compared with the control group. Up-regulation of this biomarker in OSCC patients was not significant compared with the control group (P = 0.076). Up-regulation of micro-RNA-155 was only significant in OLP group, compared with the control group (P = 0.009). No other significant differences were found (P > 0.05). CONCLUSION Considering the altered expression of MicroRNA-146a and microRNA-155 in dysplastic OLP and OSCC, their altered expression may serve as an alarming sign of malignancy. However, further investigations are still required.
Collapse
Affiliation(s)
- Masoumeh Mehdipour
- Oral Medicine Department, School of Dentistry, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin, Chamran high way, Tehran, 1983963113, Iran
| | - Minoo Shahidi
- Hematology and blood banking Department, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Anbari
- Oral Medicine Department, School of Dentistry, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin, Chamran high way, Tehran, 1983963113, Iran
| | - Homa Mirzaei
- Oral Medicine Department, School of Dentistry, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin, Chamran high way, Tehran, 1983963113, Iran.
| | - Soudeh Jafari
- Oral Medicine Department, School of Dentistry, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin, Chamran high way, Tehran, 1983963113, Iran
| | - Azam Kholghi
- Department of Medical Biotechnology, Faculty of Allied Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Lotfi
- Department of Medical Biotechnology, Faculty of Allied Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Manifar
- Oral Medicine Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mashhadiabbas
- Oral Pathology Department, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Stabile R, Cabezas MR, Verhagen MP, Tucci FA, van den Bosch TPP, De Herdt MJ, van der Steen B, Nigg AL, Chen M, Ivan C, Shimizu M, Koljenović S, Hardillo JA, Verrijzer CP, Baatenburg de Jong RJ, Calin GA, Fodde R. The deleted in oral cancer (DOC1 aka CDK2AP1) tumor suppressor gene is downregulated in oral squamous cell carcinoma by multiple microRNAs. Cell Death Dis 2023; 14:337. [PMID: 37217493 DOI: 10.1038/s41419-023-05857-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023]
Abstract
Cyclin-dependent kinase 2-associated protein 1 (CDK2AP1; also known as deleted in oral cancer or DOC1) is a tumor suppressor gene known to play functional roles in both cell cycle regulation and in the epigenetic control of embryonic stem cell differentiation, the latter as a core subunit of the nucleosome remodeling and histone deacetylation (NuRD) complex. In the vast majority of oral squamous cell carcinomas (OSCC), expression of the CDK2AP1 protein is reduced or lost. Notwithstanding the latter (and the DOC1 acronym), mutations or deletions in its coding sequence are extremely rare. Accordingly, CDK2AP1 protein-deficient oral cancer cell lines express as much CDK2AP1 mRNA as proficient cell lines. Here, by combining in silico and in vitro approaches, and by taking advantage of patient-derived data and tumor material in the analysis of loss of CDK2AP1 expression, we identified a set of microRNAs, namely miR-21-5p, miR-23b-3p, miR-26b-5p, miR-93-5p, and miR-155-5p, which inhibit its translation in both cell lines and patient-derived OSCCs. Of note, no synergistic effects were observed of the different miRs on the CDK2AP1-3-UTR common target. We also developed a novel approach to the combined ISH/IF tissue microarray analysis to study the expression patterns of miRs and their target genes in the context of tumor architecture. Last, we show that CDK2AP1 loss, as the result of miRNA expression, correlates with overall survival, thus highlighting the clinical relevance of these processes for carcinomas of the oral cavity.
Collapse
Affiliation(s)
- Roberto Stabile
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mario Román Cabezas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mathijs P Verhagen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Francesco A Tucci
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | | | - Maria J De Herdt
- Department of Otorhinolaryngology and Head & Neck Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Berdine van der Steen
- Department of Otorhinolaryngology and Head & Neck Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Alex L Nigg
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Meng Chen
- Department of Translational Molecular Pathology and Center of Department of Translational Molecular Pathology, and Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Department of Translational Molecular Pathology and Center of Department of Translational Molecular Pathology, and Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Caris Life Science, Irving, TX, USA
| | - Masayoshi Shimizu
- Department of Translational Molecular Pathology and Center of Department of Translational Molecular Pathology, and Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Senada Koljenović
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Pathology, Antwerp University Hospital, 2650, Edegem, Belgium
| | - Jose A Hardillo
- Department of Otorhinolaryngology and Head & Neck Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - C Peter Verrijzer
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert J Baatenburg de Jong
- Department of Otorhinolaryngology and Head & Neck Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - George A Calin
- Department of Translational Molecular Pathology and Center of Department of Translational Molecular Pathology, and Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Riccardo Fodde
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
37
|
Papadopoulos KI, Papadopoulou A, Aw TC. Beauty and the beast: host microRNA-155 versus SARS-CoV-2. Hum Cell 2023; 36:908-922. [PMID: 36847920 PMCID: PMC9969954 DOI: 10.1007/s13577-023-00867-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/29/2023] [Indexed: 02/28/2023]
Abstract
Severe acute respiratory coronavirus 2 (SARS-CoV-2) infection in the young and healthy usually results in an asymptomatic or mild viral syndrome, possibly through an erythropoietin (EPO)-dependent, protective evolutionary landscape. In the old and in the presence of co-morbidities, however, a potentially lethal coronavirus disease 2019 (COVID-19) cytokine storm, through unrestrained renin-angiotensin aldosterone system (RAAS) hyperactivity, has been described. Multifunctional microRNA-155 (miR-155) elevation in malaria, dengue virus (DENV), the thalassemias, and SARS-CoV-1/2, plays critical antiviral and cardiovascular roles through its targeted translational repression of over 140 genes. In the present review, we propose a plausible miR-155-dependent mechanism whereby the translational repression of AGRT1, Arginase-2 and Ets-1, reshapes RAAS towards Angiotensin II (Ang II) type 2 (AT2R)-mediated balanced, tolerable, and SARS-CoV-2-protective cardiovascular phenotypes. In addition, it enhances EPO secretion and endothelial nitric oxide synthase activation and substrate availability, and negates proinflammatory Ang II effects. Disrupted miR-155 repression of AT1R + 1166C-allele, significantly associated with adverse cardiovascular and COVID-19 outcomes, manifests its decisive role in RAAS modulation. BACH1 and SOCS1 repression creates an anti-inflammatory and cytoprotective milieu, robustly inducing antiviral interferons. MiR-155 dysregulation in the elderly, and in comorbidities, allows unimpeded RAAS hyperactivity to progress towards a particularly aggressive COVID-19 course. Elevated miR-155 in thalassemia plausibly engenders a favorable cardiovascular profile and protection against malaria, DENV, and SARS-CoV-2. MiR-155 modulating pharmaceutical approaches could offer novel therapeutic options in COVID-19.
Collapse
Affiliation(s)
- K. I. Papadopoulos
- THAI StemLife, 566/3 Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, Bangkok, 10310 Thailand
| | - A. Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63 Lund, Sweden
| | - T. C. Aw
- Department of Laboratory Medicine, Changi General Hospital, 2 Simei Street 3, Singapore, 529889 Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228 Singapore
| |
Collapse
|
38
|
Fawzy MS, Ibrahiem AT, Bayomy NA, Makhdoom AK, Alanazi KS, Alanazi AM, Mukhlef AM, Toraih EA. MicroRNA-155 and Disease-Related Immunohistochemical Parameters in Cutaneous Melanoma. Diagnostics (Basel) 2023; 13:1205. [PMID: 36980512 PMCID: PMC10047208 DOI: 10.3390/diagnostics13061205] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Cutaneous melanoma is a severe and life-threatening form of skin cancer with growing incidences. While novel interventions have improved prognoses for these patients, early diagnosis of targeted treatment remains the most effective approach. MicroRNAs have grown to good use as potential biomarkers for early detection and as targets for treatment. miR-155 is well-studied for its role in tumor cell survival and proliferation in various tissues, although its role in melanoma remains controversial. In silico data analysis was performed in the dbDEMC v.3 to identify differentially expressed miRNA. We validated gene targets in melanoma using TarBase v8.0 and miRPath v3.0 and determined protein-protein interactions of the target genes. One hundred forty patients (age range 21-90 years) with cutaneous melanoma who underwent resection were included. Molecular assessment using Real-Time RT-qPCR, clinicopathological associations, and a literature review for the different roles of miR-155 in melanoma were performed. Analysis of the dbDEMC reveals controversial findings. While there is evidence of upregulation of miR-155 in primary and metastatic melanoma samples, others suggest decreased expression in later-stage melanoma and cases with brain metastasis. miR-155 has been overexpressed in prior cases of melanoma and precancerous lesions, and it was found to be dysregulated when compared to benign nevi. While miR-155 expression was associated with favorable outcomes in some studies, others showed an association with metastasis. Patients with high levels of miR-155 also noted reduction after receiving anti-PD-1 treatment, correlated with more prolonged overall survival. In our patient's cohort, 22.9% relapsed during treatment, and 45% developed recurrence, associated with factors such as lymph node infiltration, high mitotic index, and positive staining for CD117. Although overall analysis revealed miR-155 downregulation in melanoma specimens compared to non-cancer tissues, increased expression of miR-155 was associated with cases of superficial spreading melanoma subtype (p = 0.005) and any melanoma with a high mitotic rate (p = 0.010). The analysis did not identify optimum cutoff values to predict relapse, recurrence, or mortality. In conclusion, miR-155 could have, in part, a potential prognostic utility in cutaneous melanoma. Further mechanistic studies are required to unravel the multifunctional role of miR-155 in melanoma.
Collapse
Affiliation(s)
- Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia
| | - Afaf T. Ibrahiem
- Department of Pathology, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia;
| | - Naglaa A. Bayomy
- Department of Anatomy, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia;
| | - Amin K. Makhdoom
- Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; (A.K.M.); (K.S.A.); (A.M.A.); (A.M.M.)
| | - Khalid S. Alanazi
- Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; (A.K.M.); (K.S.A.); (A.M.A.); (A.M.M.)
| | - Abdulaziz M. Alanazi
- Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; (A.K.M.); (K.S.A.); (A.M.A.); (A.M.M.)
| | - Abdulaziz M. Mukhlef
- Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; (A.K.M.); (K.S.A.); (A.M.A.); (A.M.M.)
| | - Eman A. Toraih
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
39
|
Park S, Kim M, Park M, Jin Y, Lee SJ, Lee H. Specific upregulation of extracellular miR-6238 in particulate matter-induced acute lung injury and its immunomodulation. JOURNAL OF HAZARDOUS MATERIALS 2023; 445:130466. [PMID: 36455323 DOI: 10.1016/j.jhazmat.2022.130466] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/03/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening diseases characterized by a severe inflammatory response and the destruction of alveolar epithelium and endothelium. ALI/ARDS is caused by pathogens and toxic environmental stimuli, such as particulate matter (PM). However, the general symptoms of ALI/ARDS are similar, and determining the cause of lung injury is often challenging. In this study, we investigated whether there is a critical miRNA that characterizes PM-induced ALI. We found that the expression of miR-6238 is specifically upregulated in lung tissue and lung-derived extracellular vesicles (EVs) in response to PM exposure. Notably, bacterial endotoxin (Lipopolysaccharide; LPS or peptidoglycan; PTG) does not induce the expression of miR-6238 in the lung. Instead, the expression of miR-155 is dramatically increased in LPS-induced ALI. We further demonstrated that human lung epithelial cells and macrophages predominantly produce miR-6238 and miR-155, respectively. Mechanistically, EV-miR-6238 is effectively internalized into alveolar macrophages (AMs) and regulates inflammatory responses in vivo. CXCL3 is a main target of miR-6238 in AMs and modulates neutrophil infiltration into the lung alveoli. Collectively, our findings suggest that miR-6238 is a novel regulator of pulmonary inflammation and a putative biomarker that distinguishes PM-induced ALI from endotoxin (LPS/PTG)-mediated ALI.
Collapse
Affiliation(s)
- Sujeong Park
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, South Korea
| | - Miji Kim
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, South Korea
| | - Minkyung Park
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, South Korea; Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, South Korea
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Seon-Jin Lee
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, South Korea; Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, South Korea.
| | - Heedoo Lee
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, South Korea.
| |
Collapse
|
40
|
Gao Y, Yong F, Yan M, Wei Y, Wu X. miR-361 and miR-34a suppress foot-and-mouth disease virus proliferation by activating immune response signaling in PK-15 cells. Vet Microbiol 2023; 280:109725. [PMID: 36996618 DOI: 10.1016/j.vetmic.2023.109725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/11/2023] [Accepted: 03/18/2023] [Indexed: 03/22/2023]
Abstract
Foot-and-mouth disease (FMD) severely impacts cloven-hoofed live-stock production, leading to serious economic losses and international restriction on the trade of animals and animal products worldwide. MiRNAs serve key roles in viral immunity and regulation. However, the knowledge about miRNAs regulation in FMDV infection is still limited. In this study, we found that FMDV infection caused rapid cytopathic in PK-15 cell. To investigate the miRNAs' function in FMDV infection, we performed knockdown of endogenous Dgcr8 using its specific siRNA and found that interference of Dgcr8 inhibited cellular miRNA expression and increased FMDV production, including viral capsid proteins expression, viral genome copies and virus titer, suggesting that miRNAs play an important role in FMDV infection. To obtain a full perspective on miRNA expression profiling after FMDV infection, we performed miRNA sequencing and found that FMDV infection caused inhibition of miRNA expression in PK-15 cells. Together with the target prediction result, miR-34a and miR-361 were screened for further study. Function study showed that no matter plasmid or mimics-mediated overexpression of miR-34a and miR-361 both suppressed FMDV replication, while inhibition of endogenous miR-34a and miR-361 expression using specific inhibitors significantly increased FMDV replication. Further study showed that miR-34a and miR-361 stimulated IFN-β promoter activity and activated interferon-stimulated response element (ISRE). In addition, ELISA test found that miR-361 and miR-34a increased secretion level of IFN-β and IFN-γ, which may contribute to repression of FMDV replication. This study preliminary revealed that miR-361 and miR-34a inhibited FMDV proliferation via stimulating immune response.
Collapse
|
41
|
Innocenti T, Bigagli E, Lynch EN, Galli A, Dragoni G. MiRNA-Based Therapies for the Treatment of Inflammatory Bowel Disease: What Are We Still Missing? Inflamm Bowel Dis 2023; 29:308-323. [PMID: 35749310 DOI: 10.1093/ibd/izac122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Indexed: 02/05/2023]
Abstract
Micro-RNAs (miRNAs) are noncoding RNAs usually 24-30 nucleotides long that play a central role in epigenetic mechanisms of inflammatory diseases and cancers. Recently, several studies have assessed the involvement of miRNAs in the pathogenesis of inflammatory bowel disease (IBD) and colitis-associated neoplasia. Particularly, it has been shown that many members of miRNAs family are involved in the pathways of inflammation and fibrogenesis of IBD; therefore, their use as inflammatory and fibrosis biomarkers has been postulated. In light of these results, the role of miRNAs in IBD therapy has been proposed and is currently under investigation with many in vitro and in vivo studies, murine models, and a phase 2a trial. The accumulating data have pushed miRNA-based therapy closer to clinical practice, although many open questions remain. With this systematic review, we discuss the current knowledge about the therapeutic effects of miRNAs mimicking and inhibition, and we explore the new potential targets of miRNA family for the treatment of inflammation and fibrosis in IBD.
Collapse
Affiliation(s)
- Tommaso Innocenti
- IBD Referral Center, Gastroenterology Department, Careggi University Hospital, Florence, Italy.,Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elisabetta Bigagli
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Erica Nicola Lynch
- IBD Referral Center, Gastroenterology Department, Careggi University Hospital, Florence, Italy.,Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Andrea Galli
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Gabriele Dragoni
- IBD Referral Center, Gastroenterology Department, Careggi University Hospital, Florence, Italy.,Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
42
|
Aggeletopoulou I, Mouzaki A, Thomopoulos K, Triantos C. miRNA Molecules-Late Breaking Treatment for Inflammatory Bowel Diseases? Int J Mol Sci 2023; 24:2233. [PMID: 36768556 PMCID: PMC9916785 DOI: 10.3390/ijms24032233] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of non-coding RNAs that play a critical role in regulating epigenetic mechanisms in inflammation-related diseases. Inflammatory bowel diseases (IBDs), which primarily include ulcerative colitis (UC) and Crohn's disease (CD), are characterized by chronic recurrent inflammation of intestinal tissues. Due to the multifactorial etiology of these diseases, the development of innovative treatment strategies that can effectively maintain remission and alleviate disease symptoms is a major challenge. In recent years, evidence for the regulatory role of miRNAs in the pathogenetic mechanisms of various diseases, including IBD, has been accumulating. In light of these findings, miRNAs represent potential innovative candidates for therapeutic application in IBD. In this review, we discuss recent findings on the role of miRNAs in regulating inflammatory responses, maintaining intestinal barrier integrity, and developing fibrosis in clinical and experimental IBD. The focus is on the existing literature, indicating potential therapeutic application of miRNAs in both preclinical experimental IBD models and translational data in the context of clinical IBD. To date, a large and diverse data set, which is growing rapidly, supports the potential use of miRNA-based therapies in clinical practice, although many questions remain unanswered.
Collapse
Affiliation(s)
- Ioanna Aggeletopoulou
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece
| | - Konstantinos Thomopoulos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, 26504 Patras, Greece
| |
Collapse
|
43
|
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
|
44
|
Ziętara KJ, Lejman J, Wojciechowska K, Lejman M. The Importance of Selected Dysregulated microRNAs in Diagnosis and Prognosis of Childhood B-Cell Precursor Acute Lymphoblastic Leukemia. Cancers (Basel) 2023; 15:428. [PMID: 36672378 PMCID: PMC9856444 DOI: 10.3390/cancers15020428] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is a frequent type of childhood hematological malignancy. The disease is classified into several subtypes according to genetic abnormalities. MicroRNAs (miRNAs) are involved in pathological processes (e.g., proliferation, apoptosis, differentiation). A miRNA is a group of short non-coding RNAs with relevant regulatory effects on gene expression achieved by suppression of the translation or degradation of messenger RNA (mRNA). These molecules act as tumor suppressors and/or oncogenes in the pathogenesis of pediatric leukemias. The characteristic features of miRNAs are their stable form and the possibility of secretion to the circulatory system. The role of miRNA in BCP-ALL pathogenesis is still emerging, but several studies have suggested using miRNA expression profiles as biomarkers for diagnosis, prognosis, and response to therapy in leukemia. The dysregulation of some miRNAs involved in childhood acute lymphoid leukemia, such as miR-155, miR-200c, miR-100, miR-181a, miR125b, and miR146a is discussed, showing their possible employment as therapeutic targets. In the current review, the capabilities of miRNAs in non-invasive diagnostics and their prognostic potential as biomarkers are presented.
Collapse
Affiliation(s)
- Karolina Joanna Ziętara
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Jan Lejman
- Independent Public Health Care Facility of The Ministry of Internal Affairs and Administration in Lublin, 20-331 Lublin, Poland
| | - Katarzyna Wojciechowska
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
45
|
Mahdavi Anari SR, Kheirkhah B, Amini K, Roozafzai F. Expression of MicroRNA-155 in Patients with Non-Hodgkin Lymphoma, Coronavirus Disease 2019, or Both: A Cross-Sectional Study. IRANIAN JOURNAL OF MEDICAL SCIENCES 2023; 48:26-34. [PMID: 36688191 PMCID: PMC9843467 DOI: 10.30476/ijms.2022.91669.2282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 11/23/2021] [Accepted: 12/10/2021] [Indexed: 01/24/2023]
Abstract
Background Non-Hodgkin lymphoma (NHL) is the eleventh leading cause of cancer-related death in the world. Diffuse large B-cell lymphoma (DLBCL) is the most common type of NHL. Up to winter 2021-2022, the death toll caused by the coronavirus disease 2019 (COVID-19) has exceeded 5.6 million worldwide. Possible molecular mechanisms involved in the systemic inflammation, and cytokine storm in COVID-19 patients are still not fully understood. MicroRNA-155 (miR-155) plays a role in the post-transcriptional gene regulation of hematopoiesis, oncogenesis, and inflammation. The present study aimed to evaluate the expression of miR-155 in patients with DLBCL and/or COVID-19. Methods A cross-sectional study was conducted from July to December 2020 in Tehran (Iran) to evaluate the expression of miR-155 in adult patients diagnosed with DLBCL and/or COVID-19. The real-time polymerase chain reaction technique was used to evaluate the expression of miR-155 in the sera of 92 adults who were either healthy or suffering from DLBCL and/or COVID-19. Relative quantification of gene expression was calculated in terms of cycle threshold (Ct) value. Data were analyzed using SPSS software, and P<0.05 was considered statistically significant. Results The expression of miR-155 was not associated with the sex or age of the participants. In comparison with healthy individuals (-ΔCt -1.92±0.25), the expression of miR-155 increased in patients with COVID-19 (1.95±0.14), DLBCL (2.25±0.16), or both (4.33±0.65). Conclusion The expression of miR-155 increased in patients with DLBCL and/or COVID-19.
Collapse
Affiliation(s)
| | - Babak Kheirkhah
- Department of Biology, Sirjan Branch, Islamic Azad University, Sirjan, Iran,
Department of Microbiology, Kerman Branch, Islamic Azad University, Kerman, Iran
| | - Kumarss Amini
- Department of Microbiology, Saveh Branch, Islamic Azad University, Saveh, Iran
| | - Farzin Roozafzai
- Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Jahangir M, Kahrizi MS, Natami M, Moaref Pour R, Ghoreishizadeh S, Hemmatzadeh M, Mohammadi H, Shomali N, Sandoghchian Shotorbani S. MicroRNA-155 acts as a potential prognostic and diagnostic factor in patients with ankylosing spondylitis by modulating SOCS3. Mol Biol Rep 2023; 50:553-563. [PMID: 36350418 DOI: 10.1007/s11033-022-08033-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a progressive inflammatory disease. Our primary objective was to explore the role of miR-155 and its targeted factors in AS pathogenesis. METHODS AND RESULTS PBMCs were isolated from 30 AS patients and 30 healthy individuals using the Ficoll-hypaque isolation approach. The expression of miR-155 and its associated targets, including Suppressor Of Cytokine Signaling 3 (SOCS3), STAT3, and IL-21, were determined using qT-qPCR. Then, PBMCs were cultured, and the effect of miR-155, SOCS3 siRNA (to suppress its expression), pEFSOCS3 (enforced expression), and their combination were investigated by qRT-PCR and western blotting. We also treated the cultured PBMCs with Brefeldin A, a potent inhibitor of cytokine secretion, to determine its effect on IL-21 expression and secretion. In addition, the association between miR-155 and patients' clinicopathological features was examined. The results showed that miR-155, IL-21, and STAT3 were increased in patients with AS, while SOCS3 had decreasing expression trend. It was also determined that miR-155 alleviates SOCS3 expression and increases IL-21 and STAT3 expression; it had a prominent effect when combined with SOCS3 siRNA. Besides, we showed that simultaneous transfection of miR-155 and pEFSOCS3 had no significant effect on IL-21 and STAT3 expression, revealing that miR-155 could alleviate the enforced expression of SOCS3. It was also proven that Brefledine A led to IL-21 up-regulation or accumulation while relieving its secretion. Also, a significant correlation between miR-155 and pathological features of AS patients was found. CONCLUSION miR-155 acts as a potential prognostic and diagnostic biomarker. Its up-regulation leads to the down-regulation of SOCS3 and increased expression of IL-21 and STAT3 as characteristic of TH-17 lymphocytes, leading to worsening inflammatory conditions in patients with AS.
Collapse
Affiliation(s)
| | | | - Mohammad Natami
- Department of Urology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raziyeh Moaref Pour
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maryam Hemmatzadeh
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siamak Sandoghchian Shotorbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
47
|
Rai KR, Liao Y, Cai M, Qiu H, Wen F, Peng M, Wang S, Liu S, Guo G, Chi X, Maarouf M, Chen Y, Huang S, Chen JL. MIR155HG Plays a Bivalent Role in Regulating Innate Antiviral Immunity by Encoding Long Noncoding RNA-155 and microRNA-155-5p. mBio 2022; 13:e0251022. [PMID: 36321836 PMCID: PMC9765511 DOI: 10.1128/mbio.02510-22] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/13/2022] [Indexed: 01/25/2023] Open
Abstract
MIR155HG encodes a precursor RNA of microRNA-155 (miRNA-155). We previously identified this RNA also as a long noncoding RNA (lncRNA) that we call lncRNA-155. To define the functions of miRNA-155 and lncRNA-155, we generated miRNA-155 knockout (KO) mice lacking only 19 bp of the miRNA-155 core sequence without affecting the expression of lncRNA-155. Surprisingly, compared with the miRNA-155KO mice, previously generated lncRNA-155KO mice were more susceptible to both influenza virus (RNA virus) and pseudorabies virus (DNA virus) infection, as characterized by lower survival rate, higher body weight loss, and higher viral load. We found that miRNA-155-5p enhanced antiviral responses by positively regulating activation of signal transducer and activator of transcription 1 (STAT1), but the STAT1 activity differed greatly in the animals (lncRNA-155KO < miRNA-155KO < wild type). In line with this, expression levels of several critical interferon-stimulated genes (ISGs) were also significantly different (lncRNA-155KO < miRNA-155KO < wild type). We found that lncRNA-155 augmented interferon beta (IFN-β) production during the viral infection, but miRNA-155 had no significant effect on the virus-induced IFN-β expression. Furthermore, we observed that lncRNA-155 loss in mice resulted in dramatic inhibition of virus-induced activation of interferon regulatory factor 3 compared to both miRNA-155KO and wild-type (WT) animals. Moreover, lncRNA-155 still significantly suppressed the viral infection even though the miRNA-155 derived from lncRNA-155 was deleted or blocked. These results reveal that lncRNA-155 and miRNA-155 regulate antiviral responses through distinct mechanisms, indicating a bivalent role for MIR155HG in innate immunity. IMPORTANCE Here, we found that lncRNA-155KO mice lacking most of the lncRNA-155 sequences along with pre-miRNA-155, were more susceptible to influenza virus or pseudorabies virus infection than miRNA-155KO mice lacking only 19 bp of the miRNA-155 core sequence without affecting the expression of lncRNA-155, as evidenced by faster body weight loss, poorer survival, and higher viral load, suggesting an additional role of lncRNA-155 in regulating viral pathogenesis besides via processing miRNA-155. Congruously, miRNA-155-deleted lncRNA-155 significantly attenuated the viral infection. Mechanistically, we demonstrated miRNA-155-5p potentiated antiviral responses by promoting STAT1 activation but could not directly regulate the IFN-β expression. In contrast, lncRNA-155 enhanced virus-induced IFN-β production by regulating the activation of interferon regulatory factor 3. This finding reveals a bivalent role of MIR155HG in regulating antiviral responses through encoding lncRNA-155 and miRNA-155-5p and provides new insights into complicated mechanisms underlying interaction between virus and host innate immunity.
Collapse
Affiliation(s)
- Kul Raj Rai
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yuan Liao
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mengjuan Cai
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Haori Qiu
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Faxin Wen
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Min Peng
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Song Wang
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shasha Liu
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Guijie Guo
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaojuan Chi
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mohamed Maarouf
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yuhai Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Ji-Long Chen
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
48
|
Landeros N, Gonzalez-Hormazabal P, Pérez-Moreno P, Tapia JC, Jara L. A Single Variant in Pri-miRNA-155 Associated with Susceptibility to Hereditary Breast Cancer Promotes Aggressiveness in Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms232315418. [PMID: 36499743 PMCID: PMC9735695 DOI: 10.3390/ijms232315418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Variants in genes encoding for microRNAs have been associated with their deregulation in breast cancer (BC). Sequencing of microRNAs deregulated in BC was performed using DNA from Chilean patients with a strong family history and negative for mutations in BRCA1/BRCA2. Seventeen variants were identified, three of which were selected for a case-control association study: rs376491654 (miR-335), rs755634302 (miR-497), and rs190708267 (miR-155). For rs190708267 C>T, the heterozygous T allele was detected in four BC cases and absent in controls, while homozygous TT cases were not detected. Variants were modelled in silico, cloned in a plasmid, expressed in BC cell lines, and functional in vitro assays were performed. Overexpression of the miR-155-T allele increased mature miR-155-5p levels in both BC cell lines, suggesting that its presence alters pre-miR-155 processing. Moreover, BC cells overexpressing the miR-155-T allele showed increased proliferation, migration, and resistance to cisplatin-induced death compared to miR-155-C overexpressing cells. Of note, the 3′UTR of APC, GSK3β, and PPP1CA genes, all into the canonical Wnt signaling pathway, were identified as direct targets. APC and GSK3β mRNA levels decreased while PP1 levels increased. These results suggest a pathogenic role of the variant rs190708267 (miR-155) in BRCA 1/2 negative BC, conferring susceptibility and promoting traits of aggressiveness.
Collapse
Affiliation(s)
- Natalia Landeros
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Patricio Gonzalez-Hormazabal
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Pablo Pérez-Moreno
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Julio C Tapia
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Lilian Jara
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| |
Collapse
|
49
|
Song M, Cui X, Zhang J, Li Y, Li J, Zang Y, Li Q, Yang Q, Chen Y, Cai W, Weng X, Wang Y, Zhu X. Shenlian extract attenuates myocardial ischaemia-reperfusion injury via inhibiting M1 macrophage polarization by silencing miR-155. PHARMACEUTICAL BIOLOGY 2022; 60:2011-2024. [PMID: 36239618 PMCID: PMC9578494 DOI: 10.1080/13880209.2022.2117828] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
CONTEXT Shenlian extract (SL) is a combination of Salvia miltiorrhiza Bge. (Labiatae) and Andrographis paniculata (Burm. F.) Wall. Ex Nees (Acanthaceae) extracts, which promote blood circulation and clear endogenous heat toxins. Myocardial ischaemia-reperfusion injury (MI/RI) is aggravated myocardial tissue damage induced by reperfusion therapy after myocardial infarction. OBJECTIVES This study explores the effect of SL on MI/RI and the underlying mechanism. MATERIALS AND METHODS Primary peritoneal macrophages (pMACs) were treated with LPS and SL (5, 10 or 20 μg/mL) for 24 h. The myocardial ischaemia-reperfusion (MI/R) model was established after administration of different doses of SL (90, 180 or 360 mg/kg). Myocardial tissue injury was assessed by methylthiazolyl tetrazolium (TTC) staining and levels of creatine kinase (CK), lactate dehydrogenase (LDH) and superoxide dismutase (SOD) in mice. The double immunofluorescence staining of iNOS/F4/80 and CD86/F4/80 was used to detect macrophage M1 polarization. The levels of miR-155, inflammatory factors and chemokines were detected by qRT-PCR or ELISA. CD86, iNOS, SOCS3, JAK2, p-JAK2, STAT3 and p-STAT3 proteins expressions in macrophages were analyzed by western blotting. Conditioned medium transfer systems were designed to unite M1 macrophages with H/R cardiomyocytes, and cell apoptosis was detected by TUNEL staining, western blotting or immunohistochemistry. RESULTS SL reduced apoptosis, diminished CK and LDH levels, raised SOD concentration and decreased infarct size in the MI/R model. Meanwhile, SL decreased miR-155 level, inhibited M1 macrophage polarization and inflammation. Furthermore, SL promoted SOCS3 expression and blocked JAK2/STAT3 pathway in vitro. CONCLUSIONS SL may be a promising TCM candidate for MI/RI. The underlying mechanisms could be associated with inhibition of M1 macrophage polarization via down-regulating miR-155.
Collapse
Affiliation(s)
- Min Song
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Xihe Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Jing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Jingjing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Yuanlong Zang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Weiyan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Yajie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, China
| |
Collapse
|
50
|
McDonald SJ, Cranford TL, VanderVeen BN, Cardaci TD, Velázquez KT, Enos RT, Chatzistamou I, Fan D, Murphy EA. miR155 deficiency reduces breast tumor burden in the MMTV-PyMT mouse model. Physiol Genomics 2022; 54:433-442. [PMID: 36121133 PMCID: PMC9602813 DOI: 10.1152/physiolgenomics.00057.2022] [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: 04/26/2022] [Revised: 08/25/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022] Open
Abstract
miRNA155 (miR155) has emerged as an important regulator of breast cancer (BrCa) development. Studies have consistently noted an increase in miR155 levels in serum and/or tissues in patients with BrCa. However, what is less clear is whether this increase in miR155 is a reflection of oncogenic or tumor suppressive properties. To study the effects of miR155 in a transgenic model of BrCA, we developed an MMTV-PyMT mouse deficient in miR155 (miR155-/- PyMT). miR155-/- mice (n = 11) exhibited reduced tumor number and volume palpations at ∼14-18 wk of age compared with miR155 sufficient littermates (n = 12). At 19 wk, mammary glands were excised from tumors for RT-PCR, and tumors were counted, measured, and weighed. miR155-/- PyMT mice exhibited reduced tumor volume, number, and weight, which was confirmed by histopathological analysis. There was an increase in apoptosis with miR155 deficiency and a decrease in proliferation. As expected, miR155 deficiency resulted in upregulated gene expression of suppressor of cytokine signaling 1 (Socs1)-its direct target. There was a reduction in gene expression of macrophage markers (CD68, Adgre1, Itgax, Mrc1) with miR-155-/- and this was confirmed with immunofluorescence staining for F4/80. miR155-/- increased expression of M1 macrophage marker Nos2 and reduced expression of M2 macrophage markers IL-10, IL-4, Arg1, and MMP9. Overall, miR155 deficiency reduced BrCA and improved the tumor microenvironment through the reduction of genes associated with protumorigenic processes. However, given the inconsistencies in the literature, additional studies are needed before any attempts are made to harness miR155 as a potential oncogenic or tumor suppressive miRNA.NEW & NOTEWORTHY To examine the effects of miR155 in a transgenic model of breast cancer, we developed an MMTV-PyMT mouse-deficient in miR155. We demonstrate that global loss of miR155 resulted in blunted tumor growth through modulating the tumor microenvironment. Specifically, miR155-deficient mice had smaller and less invasive tumors, an increase in apoptosis and a decrease in proliferation, a reduction in tumor-associated macrophages, and the expression of genes associated with protumoral processes.
Collapse
Affiliation(s)
- Sierra J McDonald
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Taryn L Cranford
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
- Precision Medicine Initiatives, Caris Life Sciences, Phoenix, Arizona
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
- AcePre, LLC, Columbia, South Carolina
| | - Thomas D Cardaci
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Kandy T Velázquez
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Reilly T Enos
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina
- AcePre, LLC, Columbia, South Carolina
| | - E Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
- AcePre, LLC, Columbia, South Carolina
| |
Collapse
|