1
|
Brook B, Goetz M, Duval V, Micol R, Dowling DJ. mRNA vaccines: miRNA-based controlled biodistribution and directed adjuvantation. Trends Immunol 2025:S1471-4906(25)00078-X. [PMID: 40268657 DOI: 10.1016/j.it.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
The development of ionizable mRNA-lipid nanoparticle (mRNA-LNP) nucleic acid carriers facilitated the clinical translation of the Coronavirus 2019 (COVID-19) mRNA vaccines BNT162b2 and mRNA-1273. Here, we discuss insights into rational improvements to mRNA vaccines, focusing on LNP modifications for mRNA-LNP biodistribution control, miRNA-based biodistribution control of encoded transcripts, and precision adjuvantation strategies.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Morgan Goetz
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Valerie Duval
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - Romain Micol
- Combined Therapeutics Incorporated, Boston, MA 02135, USA
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Yang H, Thompson B. Widespread changes to the translational landscape in a maize microRNA biogenesis mutant. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 119:1986-2000. [PMID: 38963711 DOI: 10.1111/tpj.16902] [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: 11/19/2023] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
MicroRNAs are short, non-coding RNAs that repress gene expression in both plants and animals and have diverse functions related to growth, development, and stress responses. The ribonuclease, DICER-LIKE1 (DCL1) is required for two steps in plant miRNA biogenesis: cleavage of the primary miRNAs (pri-miRNAs) to release a hairpin structure, called the precursor miRNA (pre-miRNA) and cleavage of the pre-miRNA to generate the miRNA/miRNA* duplex. The mature miRNA guides the RNA-induced silencing complex to target RNAs with complementary sequences, resulting in translational repression and/or RNA cleavage of target mRNAs. However, the relative contribution of translational repression versus mRNA degradation by miRNAs remains unknown at the genome-level in crops, especially in maize. The maize fuzzy tassel (fzt) mutant contains a hypomorphic mutation in DCL1 resulting in broad developmental defects. While most miRNAs are reduced in fzt, the levels of miRNA-targeted mRNAs are not dramatically increased, suggesting that translational regulation by miRNAs may be common. To gain insight into the repression mechanism of plant miRNAs, we combined ribosome profiling and RNA-sequencing to globally survey miRNA activities in maize. Our data indicate that translational repression contributes significantly to regulation of most miRNA targets and that approximately one-third of miRNA targets are regulated primarily at the translational level. Surprisingly, ribosomes appear altered in fzt mutants suggesting that DCL1 may also have a role in ribosome biogenesis. Thus, DICER-LIKE1 shapes the translational landscape in plants through both miRNA-dependent and miRNA-independent mechanisms.
Collapse
Affiliation(s)
- Hailong Yang
- Biology Department, East Carolina University, Greenville, North Carolina, USA
| | - Beth Thompson
- Biology Department, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
3
|
Pozzo LD, Xu Z, Lin S, Wang J, Wang Y, Enechojo OS, Abankwah JK, Peng Y, Chu X, Zhou H, Bian Y. Role of epigenetics in the regulation of skin aging and geroprotective intervention: A new sight. Biomed Pharmacother 2024; 174:116592. [PMID: 38615608 DOI: 10.1016/j.biopha.2024.116592] [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: 02/15/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
Multiple epigenetic factors play a regulatory role in maintaining the homeostasis of cutaneous components and are implicated in the aging process of the skin. They have been associated with the activation of the senescence program, which is the primary contributor to age-related decline in the skin. Senescent species drive a series of interconnected processes that impact the immediate surroundings, leading to structural changes, diminished functionality, and heightened vulnerability to infections. Geroprotective medicines that may restore the epigenetic balance represent valid therapeutic alliances against skin aging. Most of them are well-known Western medications such as metformin, nicotinamide adenine dinucleotide (NAD+), rapamycin, and histone deacetylase inhibitors, while others belong to Traditional Chinese Medicine (TCM) remedies for which the scientific literature provides limited information. With the help of the Geroprotectors.org database and a comprehensive analysis of the referenced literature, we have compiled data on compounds and formulae that have shown potential in preventing skin aging and have been identified as epigenetic modulators.
Collapse
Affiliation(s)
- Lisa Dal Pozzo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhe Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shan Lin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jida Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ogbe Susan Enechojo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Joseph Kofi Abankwah
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanfei Peng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoqian Chu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huifang Zhou
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
4
|
Asbjornsdottir B, Sigurdsson S, Miranda-Ribera A, Fiorentino M, Konno T, Lan J, Gudmundsson LS, Gottfredsson M, Lauth B, Birgisdottir BE, Fasano A. Evaluating Prophylactic Effect of Bovine Colostrum on Intestinal Barrier Function in Zonulin Transgenic Mice: A Transcriptomic Study. Int J Mol Sci 2023; 24:14730. [PMID: 37834178 PMCID: PMC10572565 DOI: 10.3390/ijms241914730] [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: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The intestinal barrier comprises a single layer of epithelial cells tightly joined to form a physical barrier. Disruption or compromise of the intestinal barrier can lead to the inadvertent activation of immune cells, potentially causing an increased risk of chronic inflammation in various tissues. Recent research has suggested that specific dietary components may influence the function of the intestinal barrier, potentially offering a means to prevent or mitigate inflammatory disorders. However, the precise mechanism underlying these effects remains unclear. Bovine colostrum (BC), the first milk from cows after calving, is a natural source of nutrients with immunomodulatory, anti-inflammatory, and gut-barrier fortifying properties. This novel study sought to investigate the transcriptome in BC-treated Zonulin transgenic mice (Ztm), characterized by dysbiotic microbiota, intestinal hyperpermeability, and mild hyperactivity, applying RNA sequencing. Seventy-five tissue samples from the duodenum, colon, and brain of Ztm and wild-type (WT) mice were dissected, processed, and RNA sequenced. The expression profiles were analyzed and integrated to identify differentially expressed genes (DEGs) and differentially expressed transcripts (DETs). These were then further examined using bioinformatics tools. RNA-seq analysis identified 1298 DEGs and 20,952 DETs in the paired (Ztm treatment vs. Ztm control) and reference (WT controls) groups. Of these, 733 DEGs and 10,476 DETs were upregulated, while 565 DEGs and 6097 DETs were downregulated. BC-treated Ztm female mice showed significant upregulation of cingulin (Cgn) and claudin 12 (Cldn12) duodenum and protein interactions, as well as molecular pathways and interactions pertaining to tight junctions, while BC-treated Ztm males displayed an upregulation of transcripts like occludin (Ocln) and Rho/Rac guanine nucleotide exchange factor 2 (Arhgf2) and cellular structures and interfaces, protein-protein interactions, and organization and response mechanisms. This comprehensive analysis reveals the influence of BC treatment on tight junctions (TJs) and Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) signaling pathway gene expressions. The present study is the first to analyze intestinal and brain samples from BC-treated Ztm mice applying high-throughput RNA sequencing. This study revealed molecular interaction in intestinal barrier function and identified hub genes and their functional pathways and biological processes in response to BC treatment in Ztm mice. Further research is needed to validate these findings and explore their implications for dietary interventions aimed at improving intestinal barrier integrity and function. The MGH Institutional Animal Care and Use Committee authorized the animal study (2013N000013).
Collapse
Affiliation(s)
- Birna Asbjornsdottir
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Unit for Nutrition Research, Landspitali University Hospital, Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavik, Iceland
| | - Snaevar Sigurdsson
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Biomedical Center, University of Iceland, 102 Reykjavik, Iceland
| | - Alba Miranda-Ribera
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
| | - Maria Fiorentino
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
| | - Takumi Konno
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Jinggang Lan
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
| | - Larus S. Gudmundsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, 102 Reykjavik, Iceland
| | - Magnus Gottfredsson
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Department of Scientific Affairs, Landspitali University Hospital, 102 Reykjavik, Iceland
- Department of Infectious Diseases, Landspitali University Hospital, 102 Reykjavik, Iceland
| | - Bertrand Lauth
- School of Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavik, Iceland (M.G.)
- Department of Child and Adolescent Psychiatry, Landspitali University Hospital, 102 Reykjavik, Iceland
| | - Bryndis Eva Birgisdottir
- Unit for Nutrition Research, Landspitali University Hospital, Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavik, Iceland
| | - Alessio Fasano
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; (B.A.); (M.F.); (T.K.); (J.L.)
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02138, USA
| |
Collapse
|
5
|
Egea V, Megens RTA, Santovito D, Wantha S, Brandl R, Siess W, Khani S, Soehnlein O, Bartelt A, Weber C, Ries C. Properties and fate of human mesenchymal stem cells upon miRNA let-7f-promoted recruitment to atherosclerotic plaques. Cardiovasc Res 2023; 119:155-166. [PMID: 35238350 PMCID: PMC10022860 DOI: 10.1093/cvr/cvac022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/28/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS Atherosclerosis is a chronic inflammatory disease of the arteries leading to the formation of atheromatous plaques. Human mesenchymal stem cells (hMSCs) are recruited from the circulation into plaques where in response to their environment they adopt a phenotype with immunomodulatory properties. However, the mechanisms underlying hMSC function in these processes are unclear. Recently, we described that miRNA let-7f controls hMSC invasion guided by inflammatory cytokines and chemokines. Here, we investigated the role of let-7f in hMSC tropism to human atheromas and the effects of the plaque microenvironment on cell fate and release of soluble factors. METHODS AND RESULTS Incubation of hMSCs with LL-37, an antimicrobial peptide abundantly found in plaques, increased biosynthesis of let-7f and N-formyl peptide receptor 2 (FPR2), enabling chemotactic invasion of the cells towards LL-37, as determined by qRT-PCR, flow cytometry, and cell invasion assay analysis. In an Apoe-/- mouse model of atherosclerosis, circulating hMSCs preferentially adhered to athero-prone endothelium. This property was facilitated by elevated levels of let-7f in the hMSCs, as assayed by ex vivo artery perfusion and two-photon laser scanning microscopy. Exposure of hMSCs to homogenized human atheromatous plaque material considerably induced the production of various cytokines, chemokines, matrix metalloproteinases, and tissue inhibitors of metalloproteinases, as studied by PCR array and western blot analysis. Moreover, exposure to human plaque extracts elicited differentiation of hMSCs into cells of the myogenic lineage, suggesting a potentially plaque-stabilizing effect. CONCLUSIONS Our findings indicate that let-7f promotes hMSC tropism towards atheromas through the LL-37/FPR2 axis and demonstrate that hMSCs upon contact with human plaque environment develop a potentially athero-protective signature impacting the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Virginia Egea
- Corresponding authors. Tel: +49-89-4400-55310, E-mail: (C.R.); Tel: +49-89-4400-43902, E-mail: (V.E.)
| | - Remco Theodorus Adrianus Megens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), UoS of Milan, National Research Council (CNR), Milan, Italy
| | - Sarawuth Wantha
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Richard Brandl
- St. Mary’s Square Institute for Vascular Surgery and Phlebology, Munich, Germany
| | - Wolfgang Siess
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Sajjad Khani
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University of Muenster, Muenster, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany
- Department of Molecular Metabolism, Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Ries
- Corresponding authors. Tel: +49-89-4400-55310, E-mail: (C.R.); Tel: +49-89-4400-43902, E-mail: (V.E.)
| |
Collapse
|
6
|
Mokabber H, Vatankhah MA, Najafzadeh N. The regulatory role of microRNAs in the development, cyclic changes, and cell differentiation of the hair follicle. Process Biochem 2022; 114:36-41. [DOI: 10.1016/j.procbio.2022.01.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
7
|
Nath AV, Ajit S, Sekar AJ, P R AK, Muthusamy S. MicroRNA-200c/429 mediated regulation of Zeb1 augments N-Cadherin in mouse cardiac mesenchymal cells. Cell Biol Int 2021; 46:222-233. [PMID: 34747544 DOI: 10.1002/cbin.11724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 11/10/2022]
Abstract
Cardiac mesenchymal cells (CMCs) are a promising cell type that showed therapeutic potential in heart failure models. The analysis of the underlying mechanisms by which the CMCs improve cardiac function is on track. This study aimed to investigate the expression of N-Cadherin, a transmembrane protein that enhances cell adhesion, and recently gained attention for differentiation and augmentation of stem cell function. The mouse CMCs were isolated and analyzed for the mesenchymal markers using flow cytometry. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analysis were used to assess the expression of N-Cadherin along with its counteracting molecule E-Cadherin and their regulator Zeb1 in CMCs and dermal fibroblast. The expression level of miR-200c and miR-429 was analyzed using miRNA assays. Transient transfection of miR-200c followed by qRT-PCR, western blot analysis, and immunostaining was done in CMCs to analyze the expression of Zeb1, N-Cadherin, and E-Cadherin. Flow cytometry analysis showed that CMCs possess mesenchymal markers and absence for hematopoietic and immune cell markers. Increased expression of N-Cadherin and Zeb1 in CMCs was observed in CMCs at both RNA and protein levels compared to fibroblast. We found significant downregulation of miR-200c and miR-429 in CMCs. The ectopic expression of miR-200c in CMCs significantly downregulated Zeb1 and N-Cadherin expression. Our findings suggest that the significant downregulation of miR-200c/429 in CMCs maintains the expression of N-Cadherin, which may be important for its functional integrity.
Collapse
Affiliation(s)
- Asha V Nath
- TIMED, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Shilpa Ajit
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Anupama J Sekar
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Anil Kumar P R
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Senthilkumar Muthusamy
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
8
|
Potter ML, Smith K, Vyavahare S, Kumar S, Periyasamy-Thandavan S, Hamrick M, Isales CM, Hill WD, Fulzele S. Characterization of Differentially Expressed miRNAs by CXCL12/SDF-1 in Human Bone Marrow Stromal Cells. Biomol Concepts 2021; 12:132-143. [PMID: 34648701 DOI: 10.1515/bmc-2021-0015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 01/08/2023] Open
Abstract
Stromal cell-derived factor 1 (SDF-1) is known to influence bone marrow stromal cell (BMSC) migration, osteogenic differentiation, and fracture healing. We hypothesize that SDF-1 mediates some of its effects on BMSCs through epigenetic regulation, specifically via microRNAs (miRNAs). MiRNAs are small non-coding RNAs that target specific mRNA and prevent their translation. We performed global miRNA analysis and determined several miRNAs were differentially expressed in response to SDF-1 treatment. Gene Expression Omnibus (GEO) dataset analysis showed that these miRNAs play an important role in osteogenic differentiation and fracture healing. KEGG and GO analysis indicated that SDF-1 dependent miRNAs changes affect multiple cellular pathways, including fatty acid biosynthesis, thyroid hormone signaling, and mucin-type O-glycan biosynthesis pathways. Furthermore, bioinformatics analysis showed several miRNAs target genes related to stem cell migration and differentiation. This study's findings indicated that SDF-1 induces some of its effects on BMSCs function through miRNA regulation.
Collapse
Affiliation(s)
| | - Kathryn Smith
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA
| | - Sagar Vyavahare
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA
| | - Sandeep Kumar
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA
| | | | - Mark Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA.,Institute of Healthy Aging, Augusta University, Augusta, GA
| | - Carlos M Isales
- Institute of Healthy Aging, Augusta University, Augusta, GA.,Departments of Medicine, Augusta University, Augusta, GA
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403.,Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA.,Institute of Healthy Aging, Augusta University, Augusta, GA.,Departments of Medicine, Augusta University, Augusta, GA.,Department of Orthopedics, Augusta University, Augusta, GA
| |
Collapse
|
9
|
Egea V, Kessenbrock K, Lawson D, Bartelt A, Weber C, Ries C. Let-7f miRNA regulates SDF-1α- and hypoxia-promoted migration of mesenchymal stem cells and attenuates mammary tumor growth upon exosomal release. Cell Death Dis 2021; 12:516. [PMID: 34016957 PMCID: PMC8137693 DOI: 10.1038/s41419-021-03789-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Bone marrow-derived human mesenchymal stem cells (hMSCs) are recruited to damaged or inflamed tissues where they contribute to tissue repair. This multi-step process involves chemokine-directed invasion of hMSCs and on-site release of factors that influence target cells or tumor tissues. However, the underlying molecular mechanisms are largely unclear. Previously, we described that microRNA let-7f controls hMSC differentiation. Here, we investigated the role of let-7f in chemotactic invasion and paracrine anti-tumor effects. Incubation with stromal cell-derived factor-1α (SDF-1α) or inflammatory cytokines upregulated let-7f expression in hMSCs. Transfection of hMSCs with let-7f mimics enhanced CXCR4-dependent invasion by augmentation of pericellular proteolysis and release of matrix metalloproteinase-9. Hypoxia-induced stabilization of the hypoxia-inducible factor 1 alpha in hMSCs promoted cell invasion via let-7f and activation of autophagy. Dependent on its endogenous level, let-7f facilitated hMSC motility and invasion through regulation of the autophagic flux in these cells. In addition, secreted let-7f encapsulated in exosomes was increased upon upregulation of endogenous let-7f by treatment of the cells with SDF-1α, hypoxia, or induction of autophagy. In recipient 4T1 tumor cells, hMSC-derived exosomal let-7f attenuated proliferation and invasion. Moreover, implantation of 3D spheroids composed of hMSCs and 4T1 cells into a breast cancer mouse model demonstrated that hMSCs overexpressing let-7f inhibited tumor growth in vivo. Our findings provide evidence that let-7f is pivotal in the regulation of hMSC invasion in response to inflammation and hypoxia, suggesting that exosomal let-7f exhibits paracrine anti-tumor effects.
Collapse
Affiliation(s)
- Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany.
| | - Kai Kessenbrock
- Department of Anatomy, University of California, San Francisco, CA, USA
| | - Devon Lawson
- Department of Anatomy, University of California, San Francisco, CA, USA
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany.,Department of Molecular Metabolism, 665 Huntington Avenue, Harvard T.H. Chan School of Public Health, 02115, Boston, MA, USA
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany.,Dept. Biochemistry, Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Ries
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany.
| |
Collapse
|
10
|
Xi L, Carroll T, Matos I, Luo JD, Polak L, Pasolli HA, Jaffrey SR, Fuchs E. m6A RNA methylation impacts fate choices during skin morphogenesis. eLife 2020; 9:e56980. [PMID: 32845239 PMCID: PMC7535931 DOI: 10.7554/elife.56980] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/25/2020] [Indexed: 12/30/2022] Open
Abstract
N6-methyladenosine is the most prominent RNA modification in mammals. Here, we study mouse skin embryogenesis to tackle m6A's functions and physiological importance. We first landscape the m6A modifications on skin epithelial progenitor mRNAs. Contrasting with in vivo ribosomal profiling, we unearth a correlation between m6A modification in coding sequences and enhanced translation, particularly of key morphogenetic signaling pathways. Tapping physiological relevance, we show that m6A loss profoundly alters these cues and perturbs cellular fate choices and tissue architecture in all skin lineages. By single-cell transcriptomics and bioinformatics, both signaling and canonical translation pathways show significant downregulation after m6A loss. Interestingly, however, many highly m6A-modified mRNAs are markedly upregulated upon m6A loss, and they encode RNA-methylation, RNA-processing and RNA-metabolism factors. Together, our findings suggest that m6A functions to enhance translation of key morphogenetic regulators, while also destabilizing sentinel mRNAs that are primed to activate rescue pathways when m6A levels drop.
Collapse
Affiliation(s)
- Linghe Xi
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller UniversityNew YorkUnited States
| | - Thomas Carroll
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Irina Matos
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller UniversityNew YorkUnited States
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Lisa Polak
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller UniversityNew YorkUnited States
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Samie R Jaffrey
- Department of Pharmacology, Weill Cornell Medicine, Cornell UniversityNew YorkUnited States
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
11
|
Borgohain MP, Haridhasapavalan KK, Dey C, Adhikari P, Thummer RP. An Insight into DNA-free Reprogramming Approaches to Generate Integration-free Induced Pluripotent Stem Cells for Prospective Biomedical Applications. Stem Cell Rev Rep 2020; 15:286-313. [PMID: 30417242 DOI: 10.1007/s12015-018-9861-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than a decade ago, a pioneering study reported generation of induced Pluripotent Stem Cells (iPSCs) by ectopic expression of a cocktail of reprogramming factors in fibroblasts. This study has revolutionized stem cell research and has garnered immense interest from the scientific community globally. iPSCs hold tremendous potential for understanding human developmental biology, disease modeling, drug screening and discovery, and personalized cell-based therapeutic applications. The seminal study identified Oct4, Sox2, Klf4 and c-Myc as a potent combination of genes to induce reprogramming. Subsequently, various reprogramming factors were identified by numerous groups. Most of these studies have used integrating viral vectors to overexpress reprogramming factors in somatic cells to derive iPSCs. However, these techniques restrict the clinical applicability of these cells as they may alter the genome due to random viral integration resulting in insertional mutagenesis and tumorigenicity. To circumvent this issue, alternative integration-free reprogramming approaches are continuously developed that eliminate the risk of genomic modifications and improve the prospects of iPSCs from lab to clinic. These methods establish that integration of transgenes into the genome is not essential to induce pluripotency in somatic cells. This review provides a comprehensive overview of the most promising DNA-free reprogramming techniques that have the potential to derive integration-free iPSCs without genomic manipulation, such as sendai virus, recombinant proteins, microRNAs, synthetic messenger RNA and small molecules. The understanding of these approaches shall pave a way for the generation of clinical-grade iPSCs. Subsequently, these iPSCs can be differentiated into desired cell type(s) for various biomedical applications.
Collapse
Affiliation(s)
- Manash P Borgohain
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Chandrima Dey
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Poulomi Adhikari
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
12
|
Abazari MF, Zare Karizi S, Kohandani M, Nasiri N, Nejati F, Saburi E, Nikpoor AR, Enderami SE, Soleimanifar F, Mansouri V. MicroRNA
‐2861 and nanofibrous scaffold synergistically promote human induced pluripotent stem cells osteogenic differentiation. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.4946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mohammad Foad Abazari
- Research Center for Clinical VirologyTehran University of Medical Sciences Tehran Iran
| | - Shohreh Zare Karizi
- Department of BiologyVaramin Pishva Branch, Islamic Azad University Pishva, Varamin Iran
| | - Mina Kohandani
- Department of Biology, Faculty of Biological SciencesIslamic Azad University, East Tehran Branch Tehran Iran
| | - Navid Nasiri
- Department of Biology, Central Tehran BranchIslamic Azad University Tehran Iran
| | - Fatemeh Nejati
- Department of Biology, Central Tehran BranchIslamic Azad University Tehran Iran
| | - Ehsan Saburi
- Medical Genetics and Molecular Medicine Department, School of MedicineMashhad University of Medical Sciences Mashhad Iran
| | - Amin Reza Nikpoor
- Molecular Medicine Research CenterHormozgan Health Institute, Hormozgan University of Medical Sciences Bandar Abbas Iran
| | - Seyed Ehsan Enderami
- Diabetes Research Center, Department of Medical BiotechnologySchool of Advanced Technologies in Medicine, Mazandaran university of Medical Sciences Sari Iran
| | - Fatemeh Soleimanifar
- Department of Medical biotechnology, School of MedicineAlborz University of Medical Sciences Karaj Iran
| | - Vahid Mansouri
- Proteomics Research Center, Department of AnatomySchool of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
13
|
Liu G, Li S, Liu H, Zhu Y, Bai L, Sun H, Gao S, Jiang W, Li F. The functions of ocu-miR-205 in regulating hair follicle development in Rex rabbits. BMC DEVELOPMENTAL BIOLOGY 2020; 20:8. [PMID: 32321445 PMCID: PMC7178635 DOI: 10.1186/s12861-020-00213-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 04/13/2020] [Indexed: 12/03/2022]
Abstract
BACKGROUND Hair follicles are an appendage of the vertebrate epithelium in the skin that arise from the embryonic ectoderm and regenerate cyclically during adulthood. Dermal papilla cells (DPCs) are the key dermal component of the hair follicle that directly regulate hair follicle development, growth and regeneration. According to recent studies, miRNAs play an important role in regulating hair follicle morphogenesis and the proliferation, differentiation and apoptosis of hair follicle stem cells. RESULTS The miRNA expression profile of the DPCs from Rex rabbits with different hair densities revealed 240 differentially expressed miRNAs (|log2(HD/LD)| > 1.00 and Q-value≤0.001). Among them, ocu-miR-205-5p was expressed at higher levels in DPCs from rabbits with low hair densities (LD) than in rabbits with high hair densities (HD), and it was expressed at high levels in the skin tissue from Rex rabbits (P < 0.05). Notably, ocu-miR-205 increased cell proliferation and the cell apoptosis rate, altered the progression of the cell cycle (P < 0.05), and modulated the expression of genes involved in the PI3K/Akt, Wnt, Notch and BMP signalling pathways in DPCs and skin tissue from Rex rabbits. It also inhibited the phosphorylation of the CTNNB1 and GSK-3β proteins, decreased the level of the noggin (NOG) protein, and increased the level of phosphorylated Akt (P < 0.05). A significant change in the primary follicle density was not observed (P > 0.05), but the secondary follicle density and total follicle density (P < 0.05) were altered upon interference with ocu-miR-205-5p expression, and the secondary/primary ratio (S/P) in the ocu-miR-205-5p interfered expression group increased 14 days after the injection (P < 0.05). CONCLUSIONS In the present study, ocu-miR-205 promoted the apoptosis of DPCs, altered the expression of genes and proteins involved in the PI3K/Akt, Wnt, Notch and BMP signalling pathways in DPCs and skin from Rex rabbits, promoted the transition of hair follicles from the growth phase to the regression and resting phase, and altered the hair density of Rex rabbits.
Collapse
Affiliation(s)
- Gongyan Liu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, 271018, People's Republic of China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, 271018, People's Republic of China
- Animal Husbandry and Veterinary Institute, Shandong Academy of Agricultural Sciences, Jinan, 251000, People's Republic of China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, 251000, People's Republic of China
| | - Shu Li
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, 271018, People's Republic of China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, 271018, People's Republic of China
| | - Hongli Liu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, 271018, People's Republic of China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, 271018, People's Republic of China
| | - Yanli Zhu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, 271018, People's Republic of China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, 271018, People's Republic of China
| | - Liya Bai
- Animal Husbandry and Veterinary Institute, Shandong Academy of Agricultural Sciences, Jinan, 251000, People's Republic of China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, 251000, People's Republic of China
| | - Haitao Sun
- Animal Husbandry and Veterinary Institute, Shandong Academy of Agricultural Sciences, Jinan, 251000, People's Republic of China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, 251000, People's Republic of China
| | - Shuxia Gao
- Animal Husbandry and Veterinary Institute, Shandong Academy of Agricultural Sciences, Jinan, 251000, People's Republic of China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, 251000, People's Republic of China
| | - Wenxue Jiang
- Animal Husbandry and Veterinary Institute, Shandong Academy of Agricultural Sciences, Jinan, 251000, People's Republic of China
- Shandong Key Laboratory of Animal Disease Control and Breeding, Jinan, 251000, People's Republic of China
| | - Fuchang Li
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, 271018, People's Republic of China.
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, 271018, People's Republic of China.
| |
Collapse
|
14
|
Abstract
Infertility is an important reproductive health problem, and male infertility is especially important in more than half of infertility cases. Due to the importance of genetic factors in this condition, analysis of semen alone is not enough to recognize men with idiopathic infertility. A molecular non-invasive investigation is necessary to gain valuable information. Currently, microRNAs (miRNAs) are being used as non-invasive diagnostic biomarkers. miRNAs, single-stranded non-coding RNA molecules, act as post-transcriptional gene silencing regulators either by inhibition or repression of translation. Changes in the regulation of miRNAs have been investigated in several different types of male infertility, therefore the biological role of miRNA and gene targets has been defined. The purpose of this study was to review recent research on the altered expression of miRNA in semen, sperm, and testicular biopsy samples in infertile males with different types of unexplained infertility. Changes in miRNA regulation were investigated using microarray and the miRNA levels were confirmed by real-time qRT-PCR. This review explains why creating a non-invasive diagnostic method for male infertility is necessary and how changes in miRNA expression can be used as new diagnostic biomarkers in patients with differing spermatogenic and histopathologic injury.
Collapse
|
15
|
Yang R, Liu F, Wang J, Chen X, Xie J, Xiong K. Epidermal stem cells in wound healing and their clinical applications. Stem Cell Res Ther 2019; 10:229. [PMID: 31358069 PMCID: PMC6664527 DOI: 10.1186/s13287-019-1312-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The skin has important barrier, sensory, and immune functions, contributing to the health and integrity of the organism. Extensive skin injuries that threaten the entire organism require immediate and effective treatment. Wound healing is a natural response, but in severe conditions, such as burns and diabetes, this process is insufficient to achieve effective treatment. Epidermal stem cells (EPSCs) are a multipotent cell type and are committed to the formation and differentiation of the functional epidermis. As the contributions of EPSCs in wound healing and tissue regeneration have been increasingly attracting the attention of researchers, a rising number of therapies based on EPSCs are currently under development. In this paper, we review the characteristics of EPSCs and the mechanisms underlying their functions during wound healing. Applications of EPSCs are also discussed to determine the potential and feasibility of using EPSCs clinically in wound healing.
Collapse
Affiliation(s)
- Ronghua Yang
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Fengxia Liu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001, China
| | - Jingru Wang
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 512100, China.
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Morphological Sciences Building, Central South University, 172 Tongzi Po Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
16
|
Developmental Impairments in a Rat Model of Methyl Donor Deficiency: Effects of a Late Maternal Supplementation with Folic Acid. Int J Mol Sci 2019; 20:ijms20040973. [PMID: 30813413 PMCID: PMC6413039 DOI: 10.3390/ijms20040973] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/18/2022] Open
Abstract
Vitamins B9 (folate) and B12 act as methyl donors in the one-carbon metabolism which influences epigenetic mechanisms. We previously showed that an embryofetal deficiency of vitamins B9 and B12 in the rat increased brain expression of let-7a and miR-34a microRNAs involved in the developmental control of gene expression. This was reversed by the maternal supply with folic acid (3 mg/kg/day) during the last third of gestation, resulting in a significant reduction of associated birth defects. Since the postnatal brain is subject to intensive developmental processes, we tested whether further folate supplementation during lactation could bring additional benefits. Vitamin deficiency resulted in weaned pups (21 days) in growth retardation, delayed ossification, brain atrophy and cognitive deficits, along with unchanged brain level of let-7a and decreased expression of miR-34a and miR-23a. Whereas maternal folic acid supplementation helped restore the levels of affected microRNAs, it led to a reduction of structural and functional defects taking place during the perinatal/postnatal periods, such as learning/memory capacities. Our data suggest that a gestational B-vitamin deficiency could affect the temporal control of the microRNA regulation required for normal development. Moreover, they also point out that the continuation of folate supplementation after birth may help to ameliorate neurological symptoms commonly associated with developmental deficiencies in folate and B12.
Collapse
|
17
|
Dietrich C, Singh M, Kumar N, Singh SR. The Emerging Roles of microRNAs in Stem Cell Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:11-26. [PMID: 29754172 DOI: 10.1007/978-3-319-74470-4_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging is the continuous loss of tissue and organ function over time. MicroRNAs (miRNAs) are thought to play a vital role in this process. miRNAs are endogenous small noncoding RNAs that control the expression of target mRNA. They are involved in many biological processes such as developmental timing, differentiation, cell death, stem cell proliferation and differentiation, immune response, aging and cancer. Accumulating studies in recent years suggest that miRNAs play crucial roles in stem cell division and differentiation. In the present chapter, we present a brief overview of these studies and discuss their contributions toward our understanding of the importance of miRNAs in normal and aged stem cell function in various model systems.
Collapse
Affiliation(s)
- Catharine Dietrich
- Stem Cell Regulation and Animal Aging Section, Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Manish Singh
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Nishant Kumar
- Hospitalist Division, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
| | - Shree Ram Singh
- Stem Cell Regulation and Animal Aging Section, Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
18
|
Du KT, Deng JQ, He XG, Liu ZP, Peng C, Zhang MS. MiR-214 Regulates the Human Hair Follicle Stem Cell Proliferation and Differentiation by Targeting EZH2 and Wnt/β-Catenin Signaling Way In Vitro. Tissue Eng Regen Med 2018; 15:341-350. [PMID: 30603559 DOI: 10.1007/s13770-018-0118-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/25/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022] Open
Abstract
miR-214 plays a major role in the self-renewal of skin tissue. However, whether miR-214 regulates the proliferation and differentiation of human hair follicle stem cells (HFSCs) is unknown. Primary HFSCs were isolated from human scalp skin tissue, cultured, and identified using flow cytometry. An miR-214 mimic and inhibitor were constructed for transfection into HFSCs. The MTS and colony formation assays examined cell proliferation. Immunofluorescence detected the localization and expression levels of TCF4, β-catenin, and differentiation markers. Luciferase reporter and TOP/FOP Flash assays investigated whether miR-214 targeted EZH2 and regulated the Wnt/β-catenin signaling pathway. Western blot determined the expression levels of enhancer of zeste homolog 2 (EZH2), Wnt/β-catenin signaling-related proteins, and HFSC differentiation markers in cells subjected to miR-214 transfection. miR-214 expression was remarkably decreased during the proliferation and differentiation of HFSCs into transit-amplifying (TA) cells. Downregulation of miR-214 promotes the proliferation and differentiation of HFSCs. Overexpression of miR-214 led to decreased expression of EZH2, β-catenin, and TCF-4, whereas downregulation of miR-214 resulted in increased expression of EZH2, β-catenin, and TCF-4 as well as TA differentiation markers. Immunofluorescence assay revealed that inhibiting miR-214 triggered the entry of β-catenin and TCF-4 into the nucleus. The luciferase reporter and TOP/FOP Flash assays demonstrated that miR-214 directly targets EZH2 and affects Wnt/β-catenin signaling. The miR-214/EZH2/β-catenin axis could be considered a candidate target in tissue engineering and regenerative medicine for HFSCs.
Collapse
Affiliation(s)
- Ke-Tao Du
- 1Department of Rehabilitation, The First Affiliated Hospital, Jinan University, Guangzhou, 510630 Guangdong China
- 2Department of Rehabilitation, Chenzhou NO.1 People's Hospital, Chenzhou, 423000 Hunan China
| | - Jia-Qin Deng
- 2Department of Rehabilitation, Chenzhou NO.1 People's Hospital, Chenzhou, 423000 Hunan China
| | - Xu-Guang He
- 2Department of Rehabilitation, Chenzhou NO.1 People's Hospital, Chenzhou, 423000 Hunan China
| | - Zhao-Ping Liu
- 2Department of Rehabilitation, Chenzhou NO.1 People's Hospital, Chenzhou, 423000 Hunan China
| | - Cheng Peng
- 3Department of Plastic Surgery, The 3rd Xiangya Hospital, Central South University, Tongzipo Road NO.138, Changsha, 410013 Hunan China
| | - Ming-Sheng Zhang
- 4Department of Rehabilitation Medicine, Guangdong Geriatric Institute, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Zhongshan 2nd Road NO. 106, Guangzhou, 510080 Guangdong China
| |
Collapse
|
19
|
Yang Y, Wang S. RNA Characterization by Solid-State NMR Spectroscopy. Chemistry 2018; 24:8698-8707. [DOI: 10.1002/chem.201705583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Indexed: 02/05/2023]
Affiliation(s)
- Yufei Yang
- College of Chemistry and Molecular Engineering and Beijing NMR Center; Peking University; No.5 Yiheyuan Road, Haidian District Beijing 100871 P. R. China
| | - Shenlin Wang
- College of Chemistry and Molecular Engineering and Beijing NMR Center; Peking University; No.5 Yiheyuan Road, Haidian District Beijing 100871 P. R. China
| |
Collapse
|
20
|
Modulation of miRNAs by Vitamin C in Human Bone Marrow Stromal Cells. Nutrients 2018; 10:nu10020186. [PMID: 29419776 PMCID: PMC5852762 DOI: 10.3390/nu10020186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small (18–25 nucleotides), noncoding RNAs that have been identified as potential regulators of bone marrow stromal cell (BMSC) proliferation, differentiation, and musculoskeletal development. Vitamin C is known to play a vital role in such types of biological processes through various different mechanisms by altering mRNA expression. We hypothesized that vitamin C mediates these biological processes partially through miRNA regulation. We performed global miRNA expression analysis on human BMSCs following vitamin C treatment using microarrays containing human precursor and mature miRNA probes. Bioinformatics analyses were performed on differentially expressed miRNAs to identify novel target genes and signaling pathways. Our bioinformatics analysis suggested that the miRNAs may regulate multiple stem cell-specific signaling pathways such as cell adhesion molecules (CAMs), fatty acid biosynthesis and hormone signaling pathways. Furthermore, our analysis predicted novel stem cell proliferation and differentiation gene targets. The findings of the present study demonstrate that vitamin C can have positive effects on BMSCs in part by regulating miRNA expression.
Collapse
|
21
|
Konicke K, López-Luna A, Muñoz-Carrillo JL, Servín-González LS, Flores-de la Torre A, Olasz E, Lazarova Z. The microRNA landscape of cutaneous squamous cell carcinoma. Drug Discov Today 2018; 23:864-870. [PMID: 29317340 DOI: 10.1016/j.drudis.2018.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 11/19/2017] [Accepted: 01/04/2018] [Indexed: 12/27/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a keratinocyte-derived skin tumor. It is the second-most-common cancer affecting the Caucasian population and is responsible for >20% of all skin-cancer-related deaths. The estimated incidence of non-melanoma skin cancer in the USA is >1000000 cases per year, of which roughly 20-30% are squamous cell carcinoma. To better understand and treat this challenging cancer, current research focuses on development of novel strategies to improve the understanding of tumor biogenesis on an individual basis. microRNAs are becoming important biomarkers in the diagnosis, prognosis and treatment of cSCC. This review describes the current knowledge on miRNA expression in cSCC and its role as a biomarker for personalized medicine.
Collapse
Affiliation(s)
- Kathryn Konicke
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - José Luis Muñoz-Carrillo
- Faculty of Odontology, School of Biomedical Sciences of the Cuauhtémoc University Aguascalientes, Aguascalientes, Mexico.
| | | | | | - Edit Olasz
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zelmira Lazarova
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
22
|
Aval SF, Zarghami N, Alizadeh E, Mohammadi SA. The effect of ketorolac and triamcinolone acetonide on adipogenic and hepatogenic differentiation through miRNAs 16/15/195: Possible clinical application in regenerative medicine. Biomed Pharmacother 2018; 97:675-683. [DOI: 10.1016/j.biopha.2017.10.126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 12/26/2022] Open
|
23
|
Rahmati M, Pennisi CP, Mobasheri A, Mozafari M. Bioengineered Scaffolds for Stem Cell Applications in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:73-89. [DOI: 10.1007/5584_2018_215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Tian Y, Ma X, Lv C, Sheng X, Li X, Zhao R, Song Y, Andl T, Plikus MV, Sun J, Ren F, Shuai J, Lengner CJ, Cui W, Yu Z. Stress responsive miR-31 is a major modulator of mouse intestinal stem cells during regeneration and tumorigenesis. eLife 2017; 6. [PMID: 28870287 PMCID: PMC5584991 DOI: 10.7554/elife.29538] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/07/2017] [Indexed: 12/15/2022] Open
Abstract
Intestinal regeneration and tumorigenesis are believed to be driven by intestinal stem cells (ISCs). Elucidating mechanisms underlying ISC activation during regeneration and tumorigenesis can help uncover the underlying principles of intestinal homeostasis and disease including colorectal cancer. Here we show that miR-31 drives ISC proliferation, and protects ISCs against apoptosis, both during homeostasis and regeneration in response to ionizing radiation injury. Furthermore, miR-31 has oncogenic properties, promoting intestinal tumorigenesis. Mechanistically, miR-31 acts to balance input from Wnt, BMP, TGFβ signals to coordinate control of intestinal homeostasis, regeneration and tumorigenesis. We further find that miR-31 is regulated by the STAT3 signaling pathway in response to radiation injury. These findings identify miR-31 as a critical modulator of ISC biology, and a potential therapeutic target for a broad range of intestinal regenerative disorders and cancers. Cells lining the inner wall of the gut help to absorb nutrients and to protect the body against harmful microbes and substances. Being on the front line of defense means that these cells often sustain injuries. Specialized cells called intestinal stem cells keep the tissues healthy by replacing the damaged and dying cells. The intestinal stem cells can produce copies of themselves and generate precursors of the gut cells. They also have specific mechanism to protect themselves from cell death. These processes are regulated by different signals that are generated by the cell themselves or the neighboring cells. If these processes get out of control, cells can easily be depleted or develop into cancer cells. Until now, it remained unclear how intestinal stem cells can differentiate between and respond to multiple and simultaneous signals. It is known that short RNA molecules called microRNA play an important role in the signaling pathways of damaged cells and during cancer development. In the gut, different microRNAs, including microRNA-31,help to keep the gut lining intact. However, previous research has shown that bowel cancer cells also contain high amounts of microRNA-31. To see whether microRNA-31 plays a role in controlling the signaling systems in intestinal stem cells, Tian, Ma, Lv et al. looked at genetically modified mice that either had too much microRNA-31 or none. Mice with too much microRNA-31 produced more intestinal stem cells and were able to better repair any cell damage. Mice without microRNA-31 gave rise to fewer intestinal stem cellsand had no damage repair, but were able to stop cancer cells in the gut from growing. The results showed that microRNA-31 in intestinal stem cells helps the cells to divide and to protect themselves from cell death. It controlled and balanced the different types of cell signaling by either repressing or activating various signals. When Tian et al. damaged the stem cells using radiation, the cells increased their microRNA-31 levels as a defense mechanism. This helped the cells to survive and to activate repair mechanisms. Furthermore, Tian et al. discovered that microRNA-31 can enhance the growth of tumors. These results indicate that microRNA-31 plays an important role both in repairing gut linings and furthering tumor development. A next step will be to see whether cancer cells use microRNA-31 to protect themselves from chemo- and radiation therapy. This could help scientists find new ways to render cancerous cells more susceptible to existing cancer therapies.
Collapse
Affiliation(s)
- Yuhua Tian
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xianghui Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Cong Lv
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaole Sheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiang Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ran Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yongli Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Thomas Andl
- Vanderbilt University Medical Center, Nashville, United States
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Jinyue Sun
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jianwei Shuai
- Department of Physics and State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, China
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Wei Cui
- Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Zhengquan Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
25
|
Tuning of major signaling networks (TGF-β, Wnt, Notch and Hedgehog) by miRNAs in human stem cells commitment to different lineages: Possible clinical application. Biomed Pharmacother 2017; 91:849-860. [PMID: 28501774 DOI: 10.1016/j.biopha.2017.05.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/29/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023] Open
Abstract
Two distinguishing characteristics of stem cells, their continuous division in the undifferentiated state and growth into any cell types, are orchestrated by a number of cell signaling pathways. These pathways act as a niche factor in controlling variety of stem cells. The core stem cell signaling pathways include Wingless-type (Wnt), Hedgehog (HH), and Notch. Additionally, they critically regulate the self-renewal and survival of cancer stem cells. Conversely, stem cells' main properties, lineage commitment and stemness, are tightly controlled by epigenetic mechanisms such as DNA methylation, histone modifications and non-coding RNA-mediated regulatory events. MicroRNAs (miRNAs) are cellular switches that modulate stem cells outcomes in response to diverse extracellular signals. Numerous scientific evidences implicating miRNAs in major signal transduction pathways highlight new crosstalks of cellular processes. Aberrant signaling pathways and miRNAs levels result in developmental defects and diverse human pathologies. This review discusses the crosstalk between the components of main signaling networks and the miRNA machinery, which plays a role in the context of stem cells development and provides a set of examples to illustrate the extensive relevance of potential novel therapeutic targets.
Collapse
|
26
|
Late Maternal Folate Supplementation Rescues from Methyl Donor Deficiency-Associated Brain Defects by Restoring Let-7 and miR-34 Pathways. Mol Neurobiol 2016; 54:5017-5033. [PMID: 27534418 PMCID: PMC5533871 DOI: 10.1007/s12035-016-0035-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/01/2016] [Indexed: 12/21/2022]
Abstract
The micronutrients folate and vitamin B12 are essential for the proper development of the central nervous system, and their deficiency during pregnancy has been associated with a wide range of disorders. They act as methyl donors in the one-carbon metabolism which critically influences epigenetic mechanisms. In order to depict further underlying mechanisms, we investigated the role of let-7 and miR-34, two microRNAs regulated by methylation, on a rat model of maternal deficiency. In several countries, public health policies recommend periconceptional supplementation with folic acid. However, the question about the duration and periodicity of supplementation remains. We therefore tested maternal supply (3 mg/kg/day) during the last third of gestation from embryonic days (E) 13 to 20. Methyl donor deficiency-related developmental disorders at E20, including cerebellar and interhemispheric suture defects and atrophy of selective cerebral layers, were associated with increased brain expression (by 2.5-fold) of let-7a and miR-34a, with subsequent downregulation of their regulatory targets such as Trim71 and Notch signaling partners, respectively. These processes could be reversed by siRNA strategy in differentiating neuroprogenitors lacking folate, with improvement of their morphological characteristics. While folic acid supplementation helped restoring the levels of let-7a and miR-34a and their downstream targets, it led to a reduction of structural and functional defects taking place during the perinatal period. Our data outline the potential role of let-7 and miR-34 and their related signaling pathways in the developmental defects following gestational methyl donor deficiency and support the likely usefulness of late folate supplementation in at risk women.
Collapse
|
27
|
Bhajun R, Guyon L, Gidrol X. MicroRNA degeneracy and pluripotentiality within a Lavallière-tie architecture confers robustness to gene expression networks. Cell Mol Life Sci 2016; 73:2821-7. [PMID: 27038488 PMCID: PMC4937071 DOI: 10.1007/s00018-016-2186-1] [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: 09/07/2015] [Revised: 02/24/2016] [Accepted: 03/18/2016] [Indexed: 11/03/2022]
Abstract
Modularity, feedback control, functional redundancy and bowtie architecture have been proposed as key factors that confer robustness to complex biological systems. MicroRNAs (miRNAs) are highly conserved but functionally dispensable. These antinomic properties suggest that miRNAs fine-tune gene expression rather than act as genetic switches. We synthesize published and unpublished data and hypothesize that miRNA pluripotentiality acts to buffer gene expression, while miRNA degeneracy tunes the expression of targets, thus providing robustness to gene expression networks. Furthermore, we propose a Lavallière-tie architecture by integrating signal transduction, miRNAs and protein expression data to model complex gene expression networks.
Collapse
Affiliation(s)
- Ricky Bhajun
- CEA, BIG, BGE, 17, rue des Martyrs, 38000, Grenoble, France
- University Grenoble Alpes, BGE, 38000, Grenoble, France
- INSERM, U1038, 38000, Grenoble, France
| | - Laurent Guyon
- CEA, BIG, BGE, 17, rue des Martyrs, 38000, Grenoble, France
- University Grenoble Alpes, BGE, 38000, Grenoble, France
- INSERM, U1038, 38000, Grenoble, France
| | - Xavier Gidrol
- CEA, BIG, BGE, 17, rue des Martyrs, 38000, Grenoble, France.
- University Grenoble Alpes, BGE, 38000, Grenoble, France.
- INSERM, U1038, 38000, Grenoble, France.
| |
Collapse
|
28
|
Sadiq S, Crowley TM, Charchar FJ, Sanigorski A, Lewandowski PA. MicroRNAs in a hypertrophic heart: from foetal life to adulthood. Biol Rev Camb Philos Soc 2016; 92:1314-1331. [DOI: 10.1111/brv.12283] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 04/29/2016] [Accepted: 05/06/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Shahzad Sadiq
- School of Medicine, Faculty of Health; Deakin University; 75 Pigdons Road Waurn Ponds Victoria 3216 Australia
| | - Tamsyn M. Crowley
- School of Medicine, Faculty of Health; Deakin University; 75 Pigdons Road Waurn Ponds Victoria 3216 Australia
| | - Fadi J. Charchar
- School of Health Sciences; Faculty of Science and Technology, Federation University; Ballarat Victoria 3353 Australia
| | - Andrew Sanigorski
- School of Medicine, Faculty of Health; Deakin University; 75 Pigdons Road Waurn Ponds Victoria 3216 Australia
| | - Paul A. Lewandowski
- School of Medicine, Faculty of Health; Deakin University; 75 Pigdons Road Waurn Ponds Victoria 3216 Australia
| |
Collapse
|
29
|
Geisler A, Fechner H. MicroRNA-regulated viral vectors for gene therapy. World J Exp Med 2016; 6:37-54. [PMID: 27226955 PMCID: PMC4873559 DOI: 10.5493/wjem.v6.i2.37] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 03/02/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
Safe and effective gene therapy approaches require targeted tissue-specific transfer of a therapeutic transgene. Besides traditional approaches, such as transcriptional and transductional targeting, microRNA-dependent post-transcriptional suppression of transgene expression has been emerging as powerful new technology to increase the specificity of vector-mediated transgene expression. MicroRNAs are small non-coding RNAs and often expressed in a tissue-, lineage-, activation- or differentiation-specific pattern. They typically regulate gene expression by binding to imperfectly complementary sequences in the 3' untranslated region (UTR) of the mRNA. To control exogenous transgene expression, tandem repeats of artificial microRNA target sites are usually incorporated into the 3' UTR of the transgene expression cassette, leading to subsequent degradation of transgene mRNA in cells expressing the corresponding microRNA. This targeting strategy, first shown for lentiviral vectors in antigen presenting cells, has now been used for tissue-specific expression of vector-encoded therapeutic transgenes, to reduce immune response against the transgene, to control virus tropism for oncolytic virotherapy, to increase safety of live attenuated virus vaccines and to identify and select cell subsets for pluripotent stem cell therapies, respectively. This review provides an introduction into the technical mechanism underlying microRNA-regulation, highlights new developments in this field and gives an overview of applications of microRNA-regulated viral vectors for cardiac, suicide gene cancer and hematopoietic stem cell therapy, as well as for treatment of neurological and eye diseases.
Collapse
|
30
|
Brito BDL, Lourenço SV, Damascena AS, Kowalski LP, Soares FA, Coutinho-Camillo CM. Expression of stem cell-regulating miRNAs in oral cavity and oropharynx squamous cell carcinoma. J Oral Pathol Med 2016; 45:647-654. [PMID: 26841253 DOI: 10.1111/jop.12424] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is the sixth most common tumor worldwide and is histologically heterogeneous. Studies have demonstrated the presence of stem cell markers in HNSCC, and microRNAs (miRNAs) have emerged as powerful regulators of differentiation, controlling the self-renewal of stem cells. miRNAs are non-coding RNA molecules that regulate gene expression post-transcriptionally. Many miRNAs have been described as regulators of stem cells in different types of cancer. METHODS We have analyzed the expression of let-7a, miR-34, miR-125b, miR-138, miR-145, miR-183, miR-200b, miR-203, and miR-205 by real-time RT-PCR (qPCR), in 35 oral cavity and oropharynx squamous cell carcinoma (SCC) samples and 10 non-neoplastic oral mucosa controls, to determine possible associations between the expression of these miRNAs and clinical and pathological features of these tumors. RESULTS We observed downregulation of miR-200b and miR-203 in 60.0% and 71.4% of the samples, respectively. Upregulation of miR-138 and miR-183 was observed in 50.0% of the samples. Downregulation of let-7a was associated with perineural invasion. Upregulation of miR-138, miRNA-145, and miR-205 was associated with advanced tumor stages, vascular invasion, and lymph node metastasis, respectively. CONCLUSIONS Our study provides evidence of the expression of miRNAs associated with stem cell regulation in oral cavity and oropharynx SCC and the association of these miRNAs with clinical and pathological features of these tumors.
Collapse
Affiliation(s)
- Bárbara de Lima Brito
- International Research Center, A.C. Camargo Cancer Center, São Paulo, SP, Brazil.,Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Silvia Vanessa Lourenço
- Department of General Pathology, Dental School, University of São Paulo, São Paulo, SP, Brazil
| | | | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery and Otorhinolaryngology, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Fernando Augusto Soares
- Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil.,Department of General Pathology, Dental School, University of São Paulo, São Paulo, SP, Brazil
| | - Cláudia Malheiros Coutinho-Camillo
- International Research Center, A.C. Camargo Cancer Center, São Paulo, SP, Brazil. .,Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, SP, Brazil.
| |
Collapse
|
31
|
Hijacking the Cellular Mail: Exosome Mediated Differentiation of Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:3808674. [PMID: 26880957 PMCID: PMC4736778 DOI: 10.1155/2016/3808674] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/16/2015] [Accepted: 10/25/2015] [Indexed: 02/06/2023] Open
Abstract
Bone transplantation is one of the most widely performed clinical procedures. Consequently, bone regeneration using mesenchymal stem cells and tissue engineering strategies is one of the most widely researched fields in regenerative medicine. Recent scientific consensus indicates that a biomimetic approach is required to achieve proper regeneration of any tissue. Exosomes are nanovesicles secreted by cells that act as messengers that influence cell fate. Although exosomal function has been studied with respect to cancer and immunology, the role of exosomes as inducers of stem cell differentiation has not been explored. We hypothesized that exosomes can be used as biomimetic tools for regenerative medicine. In this study we have explored the use of cell-generated exosomes as tools to induce lineage specific differentiation of stem cells. Our results indicate that proosteogenic exosomes isolated from cell cultures can induce lineage specific differentiation of naïve MSCs in vitro and in vivo. Additionally, exosomes can also bind to matrix proteins such as type I collagen and fibronectin enabling them to be tethered to biomaterials. Overall, the results from this study show the potential of cell derived exosomes in bone regenerative medicine and opens up new avenues for future research.
Collapse
|
32
|
Epigenetic Regulation of Epidermal Stem Cell Biomarkers and Their Role in Wound Healing. Int J Mol Sci 2015; 17:ijms17010016. [PMID: 26712738 PMCID: PMC4730263 DOI: 10.3390/ijms17010016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/11/2015] [Accepted: 12/16/2015] [Indexed: 12/11/2022] Open
Abstract
As an actively renewable tissue, changes in skin architecture are subjected to the regulation of stem cells that maintain the population of cells responsible for the formation of epidermal layers. Stems cells retain their self-renewal property and express biomarkers that are unique to this population. However, differential regulation of the biomarkers can initiate the pathway of terminal cell differentiation. Although, pockets of non-clarity in stem cell maintenance and differentiation in skin still exist, the influence of epigenetics in epidermal stem cell functions and differentiation in skin homeostasis and wound healing is clearly evident. The focus of this review is to discuss the epigenetic regulation of confirmed and probable epidermal stem cell biomarkers in epidermal stratification of normal skin and in diseased states. The role of epigenetics in wound healing, especially in diseased states of diabetes and cancer, will also be conveyed.
Collapse
|
33
|
Knudsen LA, Petersen N, Schwartz TW, Egerod KL. The MicroRNA Repertoire in Enteroendocrine Cells: Identification of miR-375 as a Potential Regulator of the Enteroendocrine Lineage. Endocrinology 2015; 156:3971-83. [PMID: 26322371 DOI: 10.1210/en.2015-1088] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Micro-RNAs (miRNAs) are crucial for many biological processes, but their role in the enteroendocrine development and differentiation has been neglected due to the elusive nature of the enteroendocrine cells. However, transgenic mice expressing fluorescent reporter proteins under the control of promoters for Cck, Gpr41, and Lgr5, ie, two different enteroendocrine markers and a marker for the stem cells, now enables identification and FACS purification of enteroendocrine cells at different stages of their differentiation along the crypt-villus axis. Surprisingly few of the 746 analyzed miRNAs differed in their expression pattern between enteroendocrine and nonenteroendocrine cells of the gut mucosa and between enteroendocrine cells of the crypt versus the villus. Thus, only let-7g-3p, miR-7b-5p (miR-7b), and miR-375-3p (miR-375) were up-regulated in the enteroendocrine cells of both the crypt and villus compared with nonenteroendocrine cells, and in situ hybridization confirmed colocalization of miR-375 with the enteroendocrine cells. Finally, functional assays using miR-375 inhibitor and mimetic in organoid cultures revealed miR-375 as a potential regulator of the enteroendocrine lineage. Overexpression of miR-375 inhibited enteroendocrine lineage development, whereas inhibition of miR-375 stimulated the development of enteroendocrine cells in vitro. Thus, through an unbiased expression screening of all miRNA, we find very few miRNAs that are differentially expressed in the gastrointestinal mucosa. Of these, miR-375 is found to be both highly expressed and enriched in the enteroendocrine cells. Additionally, miR-375 appears to negatively regulate the development of enteroendocrine cells. Consequently, miR-375 emerges as a potential target to modulate the function of the enteroendocrine system.
Collapse
Affiliation(s)
- Lina A Knudsen
- Novo Nordisk Foundation Center for Basic Metabolic Research (L.A.K., N.P., T.W.S., K.L.E.) and Laboratory for Molecular Pharmacology (L.A.K., N.P., T.W.S., K.L.E.), Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Natalia Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research (L.A.K., N.P., T.W.S., K.L.E.) and Laboratory for Molecular Pharmacology (L.A.K., N.P., T.W.S., K.L.E.), Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research (L.A.K., N.P., T.W.S., K.L.E.) and Laboratory for Molecular Pharmacology (L.A.K., N.P., T.W.S., K.L.E.), Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research (L.A.K., N.P., T.W.S., K.L.E.) and Laboratory for Molecular Pharmacology (L.A.K., N.P., T.W.S., K.L.E.), Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| |
Collapse
|
34
|
Jia KT, Zhang J, Jia P, Zeng L, Jin Y, Yuan Y, Chen J, Hong Y, Yi M. Identification of MicroRNAs in Zebrafish Spermatozoa. Zebrafish 2015; 12:387-97. [PMID: 26418264 DOI: 10.1089/zeb.2015.1115] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) participate in almost all biological processes. Plenty of evidences show that some testis- or spermatozoa-specific miRNAs play crucial roles in the process of gonad and germ cell development. In this study, the spermatozoa miRNA profiles were investigated through a combination of illumina deep sequencing and bioinformatics analysis in zebrafish. Deep sequencing of small RNAs yielded 11,820,680 clean reads. By mapping to the zebrafish genome, we identified 400 novel and 204 known miRNAs that could be grouped into 104 families. Furthermore, we selected the six highest expressions of known miRNAs to detect their expression patterns in different tissues by stem-loop quantitative real-time polymerase chain reaction. We found that among the six miRNAs, dre-miR-202-5p displayed specific and high expression in zebrafish spermatozoa and testis. Fluorescence in situ hybridization analysis indicated that dre-miR-202-5p was predominantly expressed in all kind of germ cells at different spermatogenetic stages, including spermatogonia and spermatozoa, but barely expressed in the germ cells in the ovary. This sex-biased expression pattern suggests that dre-miR-202-5p might be related to spermatogenesis and the functioning of spermatozoa. The identification of miRNAs in zebrafish spermatozoa and germ cells offers new insights into the spermatogenesis and spermatozoa in the teleost and other vertebrates.
Collapse
Affiliation(s)
- Kun-Tong Jia
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China .,3 South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University , Guangzhou, China
| | - Jing Zhang
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China .,3 South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University , Guangzhou, China
| | - Peng Jia
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China .,3 South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University , Guangzhou, China
| | - Lin Zeng
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China .,3 South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University , Guangzhou, China
| | - Yilin Jin
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China .,3 South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University , Guangzhou, China
| | - Yongming Yuan
- 4 Department of Biological Sciences, National University of Singapore , Singapore, Singapore
| | - Jieying Chen
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China
| | - Yunhan Hong
- 4 Department of Biological Sciences, National University of Singapore , Singapore, Singapore
| | - Meisheng Yi
- 1 School of Marine Sciences, Sun Yat-sen University , Guangzhou, China .,2 Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University , Guangzhou, China .,3 South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-sen University , Guangzhou, China
| |
Collapse
|
35
|
Deppe J, Steinritz D, Santovito D, Egea V, Schmidt A, Weber C, Ries C. Upregulation of miR-203 and miR-210 affect growth and differentiation of keratinocytes after exposure to sulfur mustard in normoxia and hypoxia. Toxicol Lett 2015; 244:81-87. [PMID: 26383628 DOI: 10.1016/j.toxlet.2015.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/08/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
Abstract
Exposure of the skin to sulfur mustard (SM) results in long-term complications such as impaired tissue regeneration. Previous own studies in normal human epidermal keratinocytes (NHEK) treated with SM demonstrated reduced proliferation, premature differentiation and a restricted functionality of hypoxia-mediated signaling in the cells. Here, we investigated the involvement of microRNAs, miR-203 and miR-210, in these mechanisms. SM significantly upregulated the expression of miR-203 in NHEK when cultivated under normoxic and hypoxic conditions. SM had no effect on miR-210 under normoxia. However, miR-210 levels were greatly increased in NHEK when grown in hypoxia and further elevated upon exposure of the cells to SM. In normoxia and hypoxia, inhibition of miR-203 by transfection of NHEK with complementary oligonucleotides, anti-miR-203, attenuated the SM-induced impairment of metabolic activity and proliferation, and counteracted SM-promoted keratin-1 expression in these cells. Consistent ameliorating effects on dysregulated metabolic activity, proliferation and keratin-1 expression in SM-treated NHEK were obtained upon inhibition of miR-210 in these cells grown in hypoxia. Our findings provide evidence that miR-203 and miR-210 are key regulators in normal and SM-impaired keratinocyte functionality, and suggest potential usefulness of inhibitors against miR-203 and miR-210 for target-directed therapeutical intervention to improve re-epithelialization of SM-injured skin.
Collapse
Affiliation(s)
- Janina Deppe
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany; European Center of Excellence on Atherosclerosis, Hypertension and Dyslipidemia, G. d'Annunzio University, 66100 Chieti, Italy
| | - Virginia Egea
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Christian Ries
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
36
|
Guibinga GH. MicroRNAs: tools of mechanistic insights and biological therapeutics discovery for the rare neurogenetic syndrome Lesch-Nyhan disease (LND). ADVANCES IN GENETICS 2015; 90:103-131. [PMID: 26296934 DOI: 10.1016/bs.adgen.2015.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
MicroRNAs (miRNAs) are small regulatory RNAs that modulate the translation of mRNA. They have emerged over the past few years as indispensable entities in the transcriptional regulation of genes. Their discovery has added additional layers of complexity to regulatory networks that control cellular homeostasis. Also, their dysregulated pattern of expression is now well demonstrated in myriad diseases and pathogenic processes. In the current review, we highlight the role of miRNAs in Lesch-Nyhan disease (LND), a rare neurogenetic syndrome caused by mutations in the purine metabolic gene encoding the hypoxanthine-guanine phosphoribosyltransferase (HPRT) enzyme. We describe how experimental and biocomputational approaches have helped to unravel genetic and signaling pathways that provide mechanistic understanding of some of the molecular and cellular basis of this ill-defined neurogenetic disorder. Through miRNA-based target predictions, we have identified signaling pathways that may be of significance in guiding biological therapeutic discovery for this incurable neurological disorder. We also propose a model to explain how a gene such as HPRT, mostly known for its housekeeping metabolic functions, can have pleiotropic effects on disparate genes and signal transduction pathways. Our hypothetical model suggests that HPRT mRNA transcripts may be acting as competitive endogenous RNAs (ceRNAs) intertwined in multiregulatory cross talk between key neural transcripts and miRNAs. Overall, this approach of using miRNA-based genomic approaches to elucidate the molecular and cellular basis of LND and guide biological target identification might be applicable to other ill-defined rare inborn-error metabolic diseases.
Collapse
Affiliation(s)
- Ghiabe-Henri Guibinga
- Division of Genetics, Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
37
|
Xu P, Li Y, Yang S, Yang H, Tang J, Li M. Micro-ribonucleic acid 143 (MiR-143) inhibits oral squamous cell carcinoma (OSCC) cell migration and invasion by downregulation of phospho-c-Met through targeting CD44 v3. Oral Surg Oral Med Oral Pathol Oral Radiol 2015; 120:43-51. [DOI: 10.1016/j.oooo.2015.02.486] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/11/2015] [Accepted: 02/20/2015] [Indexed: 11/28/2022]
|
38
|
Update on the Pathogenic Implications and Clinical Potential of microRNAs in Cardiac Disease. BIOMED RESEARCH INTERNATIONAL 2015. [PMID: 26221581 PMCID: PMC4499420 DOI: 10.1155/2015/105620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
miRNAs, a unique class of endogenous noncoding RNAs, are highly conserved across species, repress gene translation upon binding to mRNA, and thereby influence many biological processes. As such, they have been recently recognized as regulators of virtually all aspects of cardiac biology, from the development and cell lineage specification of different cell populations within the heart to the survival of cardiomyocytes under stress conditions. Various miRNAs have been recently established as powerful mediators of distinctive aspects in many cardiac disorders. For instance, acute myocardial infarction induces cardiac tissue necrosis and apoptosis but also initiates a pathological remodelling response of the left ventricle that includes hypertrophic growth of cardiomyocytes and fibrotic deposition of extracellular matrix components. In this regard, recent findings place various miRNAs as unquestionable contributing factors in the pathogenesis of cardiac disorders, thus begging the question of whether miRNA modulation could become a novel strategy for clinical intervention. In the present review, we aim to expose the latest mechanistic concepts regarding miRNA function within the context of CVD and analyse the reported roles of specific miRNAs in the different stages of left ventricular remodelling as well as their potential use as a new class of disease-modifying clinical options.
Collapse
|
39
|
Fuchs RT, Sun Z, Zhuang F, Robb GB. Bias in ligation-based small RNA sequencing library construction is determined by adaptor and RNA structure. PLoS One 2015; 10:e0126049. [PMID: 25942392 PMCID: PMC4420488 DOI: 10.1371/journal.pone.0126049] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 03/28/2015] [Indexed: 01/01/2023] Open
Abstract
High-throughput sequencing (HTS) has become a powerful tool for the detection of and sequence characterization of microRNAs (miRNA) and other small RNAs (sRNA). Unfortunately, the use of HTS data to determine the relative quantity of different miRNAs in a sample has been shown to be inconsistent with quantitative PCR and Northern Blot results. Several recent studies have concluded that the major contributor to this inconsistency is bias introduced during the construction of sRNA libraries for HTS and that the bias is primarily derived from the adaptor ligation steps, specifically where single stranded adaptors are sequentially ligated to the 3’ and 5’-end of sRNAs using T4 RNA ligases. In this study we investigated the effects of ligation bias by using a pool of randomized ligation substrates, defined mixtures of miRNA sequences and several combinations of adaptors in HTS library construction. We show that like the 3’ adaptor ligation step, the 5’ adaptor ligation is also biased, not because of primary sequence, but instead due to secondary structures of the two ligation substrates. We find that multiple secondary structural factors influence final representation in HTS results. Our results provide insight about the nature of ligation bias and allowed us to design adaptors that reduce ligation bias and produce HTS results that more accurately reflect the actual concentrations of miRNAs in the defined starting material.
Collapse
Affiliation(s)
- Ryan T. Fuchs
- RNA Research Division, New England Biolabs Incorporated, Ipswich, Massachusetts, United States of America
| | - Zhiyi Sun
- RNA Research Division, New England Biolabs Incorporated, Ipswich, Massachusetts, United States of America
| | - Fanglei Zhuang
- RNA Research Division, New England Biolabs Incorporated, Ipswich, Massachusetts, United States of America
| | - G. Brett Robb
- RNA Research Division, New England Biolabs Incorporated, Ipswich, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
40
|
Ahmed MI, Alam M, Emelianov VU, Poterlowicz K, Patel A, Sharov AA, Mardaryev AN, Botchkareva NV. MicroRNA-214 controls skin and hair follicle development by modulating the activity of the Wnt pathway. ACTA ACUST UNITED AC 2015; 207:549-67. [PMID: 25422376 PMCID: PMC4242830 DOI: 10.1083/jcb.201404001] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
miRNA-214 regulates hair follicle development and cycling by targeting β-catenin and thereby modulating Wnt pathway transduction. Skin development is governed by complex programs of gene activation and silencing, including microRNA-dependent modulation of gene expression. Here, we show that miR-214 regulates skin morphogenesis and hair follicle (HF) cycling by targeting β-catenin, a key component of the Wnt signaling pathway. miR-214 exhibits differential expression patterns in the skin epithelium, and its inducible overexpression in keratinocytes inhibited proliferation, which resulted in formation of fewer HFs with decreased hair bulb size and thinner hair production. The inhibitory effects of miR-214 on HF development and cycling were associated with altered activities of multiple signaling pathways, including decreased expression of key Wnt signaling mediators β-catenin and Lef-1, and were rescued by treatment with pharmacological Wnt activators. Finally, we identify β-catenin as one of the conserved miR-214 targets in keratinocytes. These data provide an important foundation for further analyses of miR-214 as a key regulator of Wnt pathway activity and stem cell functions during normal tissue homeostasis, regeneration, and aging.
Collapse
Affiliation(s)
- Mohammed I Ahmed
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford BD7 1DP, England, UK
| | - Majid Alam
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford BD7 1DP, England, UK
| | | | - Krzysztof Poterlowicz
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford BD7 1DP, England, UK
| | - Ankit Patel
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford BD7 1DP, England, UK
| | - Andrey A Sharov
- Department of Dermatology, Boston University, Boston, MA 02118
| | - Andrei N Mardaryev
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford BD7 1DP, England, UK
| | - Natalia V Botchkareva
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford BD7 1DP, England, UK
| |
Collapse
|
41
|
Bhajun R, Guyon L, Pitaval A, Sulpice E, Combe S, Obeid P, Haguet V, Ghorbel I, Lajaunie C, Gidrol X. A statistically inferred microRNA network identifies breast cancer target miR-940 as an actin cytoskeleton regulator. Sci Rep 2015; 5:8336. [PMID: 25673565 PMCID: PMC5389139 DOI: 10.1038/srep08336] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/14/2015] [Indexed: 01/21/2023] Open
Abstract
MiRNAs are key regulators of gene expression. By binding to many genes, they create a complex network of gene co-regulation. Here, using a network-based approach, we identified miRNA hub groups by their close connections and common targets. In one cluster containing three miRNAs, miR-612, miR-661 and miR-940, the annotated functions of the co-regulated genes suggested a role in small GTPase signalling. Although the three members of this cluster targeted the same subset of predicted genes, we showed that their overexpression impacted cell fates differently. miR-661 demonstrated enhanced phosphorylation of myosin II and an increase in cell invasion, indicating a possible oncogenic miRNA. On the contrary, miR-612 and miR-940 inhibit phosphorylation of myosin II and cell invasion. Finally, expression profiling in human breast tissues showed that miR-940 was consistently downregulated in breast cancer tissues
Collapse
Affiliation(s)
- Ricky Bhajun
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Laurent Guyon
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Amandine Pitaval
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Eric Sulpice
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Stéphanie Combe
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Patricia Obeid
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Vincent Haguet
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Itebeddine Ghorbel
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| | - Christian Lajaunie
- 1] Center for Computational Biology - CBIO, Mines ParisTech, F-77300 Fontainebleau, France [2] Institut Curie, F-75248 Paris, France [3] INSERM, U900, F-75248 Paris, France
| | - Xavier Gidrol
- 1] Univ. Grenoble Alpes, iRTSV-BGE, F-38000 Grenoble, France [2] CEA, iRTSV-BGE, F-38000 Grenoble, France [3] INSERM, BGE, F-38000 Grenoble, France
| |
Collapse
|
42
|
Atkinson SP, Lako M, Armstrong L. Potential for pharmacological manipulation of human embryonic stem cells. Br J Pharmacol 2014; 169:269-89. [PMID: 22515554 DOI: 10.1111/j.1476-5381.2012.01978.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The therapeutic potential of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) is vast, allowing disease modelling, drug discovery and testing and perhaps most importantly regenerative therapies. However, problems abound; techniques for cultivating self-renewing hESCs tend to give a heterogeneous population of self-renewing and partially differentiated cells and general include animal-derived products that can be cost-prohibitive for large-scale production, and effective lineage-specific differentiation protocols also still remain relatively undefined and are inefficient at producing large amounts of cells for therapeutic use. Furthermore, the mechanisms and signalling pathways that mediate pluripotency and differentiation are still to be fully appreciated. However, over the recent years, the development/discovery of a range of effective small molecule inhibitors/activators has had a huge impact in hESC biology. Large-scale screening techniques, coupled with greater knowledge of the pathways involved, have generated pharmacological agents that can boost hESC pluripotency/self-renewal and survival and has greatly increased the efficiency of various differentiation protocols, while also aiding the delineation of several important signalling pathways. Within this review, we hope to describe the current uses of small molecule inhibitors/activators in hESC biology and their potential uses in the future.
Collapse
|
43
|
Marimuthu A, Huang TC, Selvan LDN, Renuse S, Nirujogi RS, Kumar P, Pinto SM, Rajagopalan S, Pandey A, Harsha H, Chatterjee A. Identification of targets of miR-200b by a SILAC-based quantitative proteomic approach. EUPA OPEN PROTEOMICS 2014. [DOI: 10.1016/j.euprot.2014.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
44
|
miR-375 induces human decidua basalis-derived stromal cells to become insulin-producing cells. Cell Mol Biol Lett 2014; 19:483-99. [PMID: 25169436 PMCID: PMC6275735 DOI: 10.2478/s11658-014-0207-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 08/19/2014] [Indexed: 02/07/2023] Open
Abstract
This paper focuses on the development of renewable sources of isletreplacement tissue for the treatment of type I diabetes mellitus. Placental tissue-derived mesenchymal stem cells (MSCs) are a promising source for regenerative medicine due to their plasticity and easy availability. They have the potential to differentiate into insulin-producing cells. miR-375 is a micro RNA that is expressed in the pancreas and involved in islet development. Human placental decidua basalis MSCs (PDB-MSCs) were cultured from full-term human placenta. The immunophenotype of the isolated cells was checked for CD90, CD105, CD44, CD133 and CD34 markers. The MSCs (P3) were chemically transfected with hsa-miR-375. Total RNA was extracted 4 and 6 days after transfection. The expressions of insulin, NGN3, GLUT2, PAX4, PAX6, KIR6.2, NKX6.1, PDX1, and glucagon genes were evaluated using real-time qPCR. On day 6, we tested the potency of the clusters in response to the high glucose challenge and assessed the presence of insulin and NGN3 proteins via immunocytochemistry. Flow cytometry analysis confirmed that more than 90% of the cells were positive for CD90, CD105 and CD44 and negative for CD133 and CD34. Morphological changes were followed from day 2. Cell clusters formed during day 6. Insulin-producing clusters showed a deep red color with DTZ. The expression of pancreatic-specific transcription factors increased remarkably during the four days after transfection and significantly increased on day 7. The clusters were positive for insulin and NGN3 proteins, and C-peptide and insulin secretion increased in response to changes in the glucose concentration (2.8 mM and 16.7 mM). In conclusion, the MSCs could be programmed into functional insulin-producing cells by transfection of miR-375.
Collapse
|
45
|
Hu JKH, Mushegyan V, Klein OD. On the cutting edge of organ renewal: Identification, regulation, and evolution of incisor stem cells. Genesis 2014; 52:79-92. [PMID: 24307456 PMCID: PMC4252016 DOI: 10.1002/dvg.22732] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 11/22/2013] [Accepted: 11/25/2013] [Indexed: 12/14/2022]
Abstract
The rodent incisor is one of a number of organs that grow continuously throughout the life of an animal. Continuous growth of the incisor arose as an evolutionary adaptation to compensate for abrasion at the distal end of the tooth. The sustained turnover of cells that deposit the mineralized dental tissues is made possible by epithelial and mesenchymal stem cells residing at the proximal end of the incisor. A complex network of signaling pathways and transcription factors regulates the formation, maintenance, and differentiation of these stem cells during development and throughout adulthood. Research over the past 15 years has led to significant progress in our understanding of this network, which includes FGF, BMP, Notch, and Hh signaling, as well as cell adhesion molecules and micro-RNAs. This review surveys key historical experiments that laid the foundation of the field and discusses more recent findings that definitively identified the stem cell population, elucidated the regulatory network, and demonstrated possible genetic mechanisms for the evolution of continuously growing teeth.
Collapse
Affiliation(s)
- Jimmy Kuang-Hsien Hu
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vagan Mushegyan
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
46
|
MicroRNA-128 regulates the differentiation of rat bone mesenchymal stem cells into neuron-like cells by Wnt signaling. Mol Cell Biochem 2013; 387:151-8. [DOI: 10.1007/s11010-013-1880-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/18/2013] [Indexed: 01/15/2023]
|
47
|
Xu T, Li L, Huang C, Li X, Peng Y, Li J. MicroRNA-323-3p with clinical potential in rheumatoid arthritis, Alzheimer's disease and ectopic pregnancy. Expert Opin Ther Targets 2013; 18:153-8. [PMID: 24283221 DOI: 10.1517/14728222.2014.855201] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION MicroRNAs (miRNAs) are a group of noncoding RNAs,∼ 20 - 22 nucleotides in length, that repress target gene expression through mRNA degradation and translation inhibition. The gene encoding miR-323-3p, which is a biomarker in immune and inflammatory responses, exists in a miRNA cluster in chromosomal region 14q32.31. It has been shown that miR-323-3p associates with the pathogenesis of several diseases, such as rheumatoid arthritis, Alzheimer's disease and ectopic pregnancy. AREAS COVERED This review provides a current view on the association of miR-323-3p with several human diseases and is focused on the recent studies of miR-323-3p regulation, discussing its potential as an epigenetic biomarker and therapeutic target for these diseases. In particular, the mechanisms of miR-323-3p in these diseases and how miR-323-3p is regulated are also discussed. EXPERT OPINION Although the exact role of miR-323-3p in these diseases has not been fully elucidated, targeting miR-323-3p may serve as a promising therapy strategy.
Collapse
Affiliation(s)
- Tao Xu
- Anhui Medical University, School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products , Anhui Province, 81 Meishan Road, Hefei 230032 , China +86 551 65161001 ; +86 551 65161001 ; ,
| | | | | | | | | | | |
Collapse
|
48
|
Shao Y, Zhang SQ, Quan F, Zhang PF, Wu SL. MicroRNA-145 inhibits the proliferation, migration and invasion of the human TCA8113 oral cancer line. Oncol Lett 2013; 6:1636-1640. [PMID: 24273601 PMCID: PMC3835314 DOI: 10.3892/ol.2013.1621] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/25/2013] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to investigate the effect of microRNA (miR)-145 on the proliferation, migration and invasion of the human oral cancer line, TCA8113. Expression levels of miR-145 in TCA8113 cells were detected by quantitative PCR. miR-145 was transfected into human TCA8113 oral cancer cells and the proliferation, migration and invasion abilities of treated TCA8113 cells were detected by proliferation, migration and invasion assays, respectively. The expression levels of miR-145 in TCA8113 cells were significantly lower than those in human normal oral keratinocytes (P<0.05). Cellular proliferation, migration and invasion abilities in the miR-145 transfection group were significantly lower than those in the control group (all P<0.05). High miR-145 expression was found to negatively regulate the proliferation, migration and invasion of TCA8113 cells. Results of the present study indicate that the expression of miR-145 may be associated with the genesis and development of human oral cancer.
Collapse
Affiliation(s)
- Yuan Shao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | | | | | | | | |
Collapse
|
49
|
Wang D, Zhang Z, O’Loughlin E, Wang L, Fan X, Lai EC, Yi R. MicroRNA-205 controls neonatal expansion of skin stem cells by modulating the PI(3)K pathway. Nat Cell Biol 2013; 15:1153-63. [PMID: 23974039 PMCID: PMC3789848 DOI: 10.1038/ncb2827] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/16/2013] [Indexed: 12/17/2022]
Abstract
Skin stem cells (SCs) are specified and rapidly expanded to fuel body growth during early development. However, the molecular mechanisms that govern the amplification of skin SCs remain unclear. Here we report an essential role for miR-205, one of the most highly expressed microRNAs in skin SCs, in promoting neonatal expansion of these cells. Unlike most mammalian miRNAs, genetic deletion of miR-205 causes neonatal lethality with severely compromised epidermal and hair follicle growth. In the miR-205 knockout skin SCs, phospho-Akt is significantly downregulated, and the SCs prematurely exit the cell cycle. In the hair follicle, this accelerates the transition of the neonatal SCs towards quiescence. We identify multiple miR-205-targeted negative regulators of PI(3)K signalling that mediate the repression of phospho-Akt and restrict the proliferation of SCs. Our findings reveal an essential role for miR-205 in maintaining the expansion of skin SCs by antagonizing negative regulators of PI(3)K signalling.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309 USA
| | - Zhaojie Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309 USA
| | - Evan O’Loughlin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309 USA
| | - Li Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309 USA
| | - Xiying Fan
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309 USA
| | - Eric C. Lai
- Sloan-Kettering Institute, 1017 Rockefeller Research Labs, 1275 York Avenue Box 252, New York, New York 10065 USA
| | - Rui Yi
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309 USA
| |
Collapse
|
50
|
Kuo YC, Li YSJ, Zhou J, Shih YRV, Miller M, Broide D, Lee OKS, Chien S. Human mesenchymal stem cells suppress the stretch-induced inflammatory miR-155 and cytokines in bronchial epithelial cells. PLoS One 2013; 8:e71342. [PMID: 23967196 PMCID: PMC3742760 DOI: 10.1371/journal.pone.0071342] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 06/30/2013] [Indexed: 12/31/2022] Open
Abstract
Current research in pulmonary pathology has focused on inflammatory reactions initiated by immunological responses to allergens and irritants. In addition to these biochemical stimuli, physical forces also play an important role in regulating the structure, function, and metabolism of the lung. Hyperstretch of lung tissues can contribute to the inflammatory responses in asthma, but the mechanisms of mechanically induced inflammation in the lung remain unclear. Our results demonstrate that excessive stretch increased the secretion of inflammatory cytokines by human bronchial epithelial cells (hBECs), including IL-8. This increase of IL-8 secretion was due to an elevated microRNA-155 (miR-155) expression, which caused the suppression of Src homology 2 domain–containing inositol 5-phosphatase 1 (SHIP1) production and the subsequent activation of JNK signaling. In vivo studies in our asthmatic mouse model also showed such changes in miR-155, IL-8, and SHIP1 expressions that reflect inflammatory responses. Co-culture with human mesenchymal stem cells (hMSCs) reversed the stretch-induced hBEC inflammatory responses as a result of IL-10 secretion by hMSCs to down-regulate miR-155 expression in hBECs. In summary, we have demonstrated that mechanical stretch modulates the homeostasis of the hBEC secretome involving miR-155 and that hMSCs can be used as a potential therapeutic approach to reverse bronchial epithelial inflammation in asthma.
Collapse
Affiliation(s)
- Yi-Chun Kuo
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Bioengineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Yi-Shuan Julie Li
- Department of Bioengineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jing Zhou
- Department of Bioengineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Yu-Ru Vernon Shih
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - David Broide
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (OK-SL); (SC)
| | - Shu Chien
- Department of Bioengineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail: (OK-SL); (SC)
| |
Collapse
|