51
|
Cordeiro-Spinetti E, Rothbart SB. Lysine methylation signaling in skeletal muscle biology: from myogenesis to clinical insights. Biochem J 2023; 480:1969-1986. [PMID: 38054592 DOI: 10.1042/bcj20230223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Lysine methylation signaling is well studied for its key roles in the regulation of transcription states through modifications on histone proteins. While histone lysine methylation has been extensively studied, recent discoveries of lysine methylation on thousands of non-histone proteins has broadened our appreciation for this small chemical modification in the regulation of protein function. In this review, we highlight the significance of histone and non-histone lysine methylation signaling in skeletal muscle biology, spanning development, maintenance, regeneration, and disease progression. Furthermore, we discuss potential future implications for its roles in skeletal muscle biology as well as clinical applications for the treatment of skeletal muscle-related diseases.
Collapse
Affiliation(s)
| | - Scott B Rothbart
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan 49503, U.S.A
| |
Collapse
|
52
|
Han X, Wang H, Du F, Zeng X, Guo C. Nrf2 for a key member of redox regulation: A novel insight against myocardial ischemia and reperfusion injuries. Biomed Pharmacother 2023; 168:115855. [PMID: 37939614 DOI: 10.1016/j.biopha.2023.115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023] Open
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2), a nuclear transcription factor, modulates genes responsible for antioxidant responses against toxic and oxidative stress to maintain redox homeostasis and participates in varieties of cellular processes such as metabolism and inflammation during myocardial ischemia and reperfusion injuries (MIRI). The accumulation of reactive oxygen species (ROS) from damaged mitochondria, xanthine oxidase, NADPH oxidases, and inflammation contributes to depraved myocardial ischemia and reperfusion injuries. Considering that Nrf2 played crucial roles in antagonizing oxidative stress, it is reasonable to delve into the up or down-regulated molecular mechanisms of Nrf2 in the progression of MIRI to provide the possibility of new therapeutic medicine targeting Nrf2 in cardiovascular diseases. This review systematically describes the generation of ROS, the regulatory metabolisms of Nrf2 as well as several natural or synthetic compounds activating Nrf2 during MIRI, which might provide novel insights for the anti-oxidative stress and original ideas targeting Nrf2 for the prevention and treatment in cardiovascular diseases.
Collapse
Affiliation(s)
- Xuejie Han
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China
| | - Fenghe Du
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, PR China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China.
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China.
| |
Collapse
|
53
|
Yu M, Thorner K, Parameswaran S, Wei W, Yu C, Lin X, Kopan R, Hass MR. The unique function of Runx1 in skeletal muscle differentiation and regeneration is mediated by an ETS interaction domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568117. [PMID: 38045385 PMCID: PMC10690193 DOI: 10.1101/2023.11.21.568117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The conserved Runt-related (RUNX) transcription factor family are well-known master regulators of developmental and regenerative processes. Runx1 and Runx2 are both expressed in satellite cells (SC) and skeletal myotubes. Conditional deletion of Runx1 in adult SC negatively impacted self-renewal and impaired skeletal muscle maintenance. Runx1- deficient SC retain Runx2 expression but cannot support muscle regeneration in response to injury. To determine the unique molecular functions of Runx1 that cannot be compensated by Runx2 we deleted Runx1 in C2C12 that retain Runx2 expression and established that myoblasts differentiation was blocked in vitro due in part to ectopic expression of Mef2c, a target repressed by Runx1 . Structure-function analysis demonstrated that the Ets-interacting MID/EID region of Runx1, absent from Runx2, is critical to regulating myoblasts proliferation, differentiation, and fusion. Analysis of in-house and published ChIP-seq datasets from Runx1 (T-cells, muscle) versus Runx2 (preosteoblasts) dependent tissue identified enrichment for a Ets:Runx composite site in Runx1 -dependent tissues. Comparing ATACseq datasets from WT and Runx1KO C2C12 cells showed that the Ets:Runx composite motif was enriched in peaks open exclusively in WT cells compared to peaks unique to Runx1KO cells. Thus, engagement of a set of targets by the RUNX1/ETS complex define the non-redundant functions of Runx1 .
Collapse
|
54
|
Brunet T. Cell contractility in early animal evolution. Curr Biol 2023; 33:R966-R985. [PMID: 37751712 DOI: 10.1016/j.cub.2023.07.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Tissue deformation mediated by collective cell contractility is a signature characteristic of animals. In most animals, fast and reversible contractions of muscle cells mediate behavior, while slow and irreversible contractions of epithelial or mesenchymal cells play a key role in morphogenesis. Animal tissue contractility relies on the activity of the actin/myosin II complex (together referred to as 'actomyosin'), an ancient and versatile molecular machinery that performs a broad range of functions in development and physiology. This review synthesizes emerging insights from morphological and molecular studies into the evolutionary history of animal contractile tissue. The most ancient functions of actomyosin are cell crawling and cytokinesis, which are found in a wide variety of unicellular eukaryotes and in individual metazoan cells. Another contractile functional module, apical constriction, is universal in metazoans and shared with choanoflagellates, their closest known living relatives. The evolution of animal contractile tissue involved two key innovations: firstly, the ability to coordinate and integrate actomyosin assembly across multiple cells, notably to generate supracellular cables, which ensure tissue integrity but also allow coordinated morphogenesis and movements at the organism scale; and secondly, the evolution of dedicated contractile cell types for adult movement, belonging to two broad categories respectively defined by the expression of the fast (striated-type) and slow (smooth/non-muscle-type) myosin II paralogs. Both contractile cell types ancestrally resembled generic contractile epithelial or mesenchymal cells and might have played a versatile role in both behavior and morphogenesis. Modern animal contractile cells span a continuum between unspecialized contractile epithelia (which underlie behavior in modern placozoans), epithelia with supracellular actomyosin cables (found in modern sponges), epitheliomuscular tissues (with a concentration of actomyosin cables in basal processes, for example in sea anemones), and specialized muscle tissue that has lost most or all epithelial properties (as in ctenophores, jellyfish and bilaterians). Recent studies in a broad range of metazoans have begun to reveal the molecular basis of these transitions, powered by the elaboration of the contractile apparatus and the evolution of 'core regulatory complexes' of transcription factors specifying contractile cell identity.
Collapse
Affiliation(s)
- Thibaut Brunet
- Institut Pasteur, Université Paris-Cité, CNRS UMR3691, Evolutionary Cell Biology and Evolution of Morphogenesis Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France.
| |
Collapse
|
55
|
Qiu Y, Li Z, Walther D, Köhler C. Updated Phylogeny and Protein Structure Predictions Revise the Hypothesis on the Origin of MADS-box Transcription Factors in Land Plants. Mol Biol Evol 2023; 40:msad194. [PMID: 37652031 PMCID: PMC10484287 DOI: 10.1093/molbev/msad194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023] Open
Abstract
MADS-box transcription factors (TFs), among the first TFs extensively studied, exhibit a wide distribution across eukaryotes and play diverse functional roles. Varying by domain architecture, MADS-box TFs in land plants are categorized into Type I (M-type) and Type II (MIKC-type). Type I and II genes have been considered orthologous to the SRF and MEF2 genes in animals, respectively, presumably originating from a duplication before the divergence of eukaryotes. Here, we exploited the increasing availability of eukaryotic MADS-box sequences and reassessed their evolution. While supporting the ancient duplication giving rise to SRF- and MEF2-types, we found that Type I and II genes originated from the MEF2-type genes through another duplication in the most recent common ancestor (MRCA) of land plants. Protein structures predicted by AlphaFold2 and OmegaFold support our phylogenetic analyses, with plant Type I and II TFs resembling the MEF2-type structure, rather than SRFs. We hypothesize that the ancestral SRF-type TFs were lost in the MRCA of Archaeplastida (the kingdom Plantae sensu lato). The retained MEF2-type TFs acquired a Keratin-like domain and became MIKC-type before the divergence of Streptophyta. Subsequently in the MRCA of land plants, M-type TFs evolved from a duplicated MIKC-type precursor through loss of the Keratin-like domain, leading to the Type I clade. Both Type I and II TFs expanded and functionally differentiated in concert with the increasing complexity of land plant body architecture. The recruitment of these originally stress-responsive TFs into developmental programs, including those underlying reproduction, may have facilitated the adaptation to the terrestrial environment.
Collapse
Affiliation(s)
- Yichun Qiu
- Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- Swedish University of Agricultural Sciences & Linnean Center for Plant Biology, Uppsala BioCenter, Uppsala, Sweden
| | - Zhen Li
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, VIB, Ghent, Belgium
| | - Dirk Walther
- Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Claudia Köhler
- Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- Swedish University of Agricultural Sciences & Linnean Center for Plant Biology, Uppsala BioCenter, Uppsala, Sweden
| |
Collapse
|
56
|
Cai C, Wan P, Wang H, Cai X, Wang J, Chai Z, Wang J, Wang H, Zhang M, Yang N, Wu Z, Zhu J, Yang X, Li Y, Yue B, Dang R, Zhong J. Transcriptional and open chromatin analysis of bovine skeletal muscle development by single-cell sequencing. Cell Prolif 2023; 56:e13430. [PMID: 36855961 PMCID: PMC10472525 DOI: 10.1111/cpr.13430] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Skeletal muscle is a complex heterogeneous tissue and characterizing its cellular heterogeneity and transcriptional and epigenetic signatures are important for understanding the details of its ontogeny. In our study, we applied scRNA-seq and scATAC-seq to investigate the cell types, molecular features, transcriptional and epigenetic regulation, and patterns of developing bovine skeletal muscle from gestational, lactational and adult stages. Detailed molecular analyses were used to dissect cellular heterogeneity, and we deduced the differentiation trajectory of myogenic cells and uncovered their dynamic gene expression profiles. SCENIC analysis was performed to demonstrate key regulons during cell fate decisions. We explored the future expression states of these heterogeneous cells by RNA velocity analysis and found extensive networks of intercellular communication using the toolkit CellChat. Moreover, the transcriptomic and chromatin accessibility modalities were confirmed to be highly concordant, and integrative analysis of chromatin accessibility and gene expression revealed key transcriptional regulators acting during myogenesis. In bovine skeletal muscle, by scRNA-seq and scATAC-seq analysis, different cell types such as adipocytes, endothelial cells, fibroblasts, lymphocytes, monocytes, pericyte cells and eight skeletal myogenic subpopulations were identified at the three developmental stages. The pseudotime trajectory exhibited a distinct sequential ordering for these myogenic subpopulations and eight distinct gene clusters were observed according to their expression pattern. Moreover, specifically expressed TFs (such as MSC, MYF5, MYOD1, FOXP3, ESRRA, BACH1, SIX2 and ATF4) associated with muscle development were predicted, and likely future transcriptional states of individual cells and the developmental dynamics of differentiation among neighbouring cells were predicted. CellChat analysis on the scRNA-seq data set then classified many ligand-receptor pairs among these cell clusters, which were further categorized into significant signalling pathways, including BMP, IGF, WNT, MSTN, ANGPTL, TGFB, TNF, VEGF and FGF. Finally, scRNA-seq and scATAC-seq results were successfully integrated to reveal a series of specifically expressed TFs that are likely to be candidates for the promotion of cell fate transition during bovine skeletal muscle development. Overall, our results outline a single-cell dynamic chromatin/transcriptional landscape for normal bovine skeletal muscle development; these provide an important resource for understanding the structure and function of mammalian skeletal muscle, which will promote research into its biology.
Collapse
Affiliation(s)
- Cuicui Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Peng Wan
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Hui Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Xin Cai
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Jiabo Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Zhixin Chai
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Jikun Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Haibo Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Ming Zhang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Nan Yang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Zhijuan Wu
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Xueyao Yang
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Yulian Li
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Binglin Yue
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Ruihua Dang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Jincheng Zhong
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| |
Collapse
|
57
|
List EO, Duran-Ortiz S, Kulkarni P, Davis E, Mora-Criollo P, Berryman DE, Kopchick JJ. Growth hormone receptor gene disruption. VITAMINS AND HORMONES 2023; 123:109-149. [PMID: 37717983 PMCID: PMC11462719 DOI: 10.1016/bs.vh.2022.12.004] [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] [Indexed: 09/19/2023]
Abstract
Much of our understanding of growth hormone's (GH)'s numerous activities stems from studies utilizing GH receptor (GHR) knockout mice. More recently, the role of GH action has been examined by creating mice with tissue-specific or temporal GHR disruption. To date, 37 distinct GHR knockout mouse lines have been created. Targeted tissues include fat, liver, muscle, heart, bone, brain, macrophage, intestine, hematopoietic stem cells, pancreatic β cells, and inducible multi-tissue "global" disruption at various ages. In this chapter, a summary of each mouse line is provided with background information on the generation of the mouse line as well as important physiological outcomes resulting from GHR gene disruption. Collectively, these mouse lines provide unique insights into GH action and have resulted in the development of new hypotheses about the functions ascribed to GH action in particular tissues.
Collapse
Affiliation(s)
- Edward O List
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Silvana Duran-Ortiz
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Prateek Kulkarni
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Emily Davis
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Patricia Mora-Criollo
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Darlene E Berryman
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - John J Kopchick
- The Edison Biotechnology Institute, and the Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States.
| |
Collapse
|
58
|
Maisuria R, Norton A, Shao C, Bradley EW, Mansky K. Conditional Loss of MEF2C Expression in Osteoclasts Leads to a Sex-Specific Osteopenic Phenotype. Int J Mol Sci 2023; 24:12686. [PMID: 37628864 PMCID: PMC10454686 DOI: 10.3390/ijms241612686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Myocyte enhancement factor 2C (MEF2C) is a transcription factor studied in the development of skeletal and smooth muscles. Bone resorption studies have exhibited that the reduced expression of MEF2C contributes to osteopetrosis and the dysregulation of pathological bone remodeling. Our current study aims to determine how MEF2C contributes to osteoclast differentiation and to analyze the skeletal phenotype of Mef2c-cKO mice (Cfms-cre; Mef2cfl/fl). qRT-PCR and Western blot demonstrated that Mef2c expression is highest during the early days of osteoclast differentiation. Osteoclast genes, including c-Fos, c-Jun, Dc-stamp, Cathepsin K, and Nfatc1, had a significant reduction in expression, along with a reduction in osteoclast size. Despite reduced CTX activity, female Mef2c cKO mice were osteopenic, with decreased bone formation as determined via a P1NP ELISA, and a reduced number of osteoblasts. There was no difference between male WT and Mef2c-cKO mice. Our results suggest that Mef2c is critical for osteoclastogenesis, and that its dysregulation leads to a sex-specific osteopenic phenotype.
Collapse
Affiliation(s)
- Ravi Maisuria
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| | - Andrew Norton
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| | - Cynthia Shao
- College of Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Elizabeth W. Bradley
- Department of Orthopedics, School of Medicine and Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Kim Mansky
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| |
Collapse
|
59
|
Le NT. The significance of ERK5 catalytic-independent functions in disease pathways. Front Cell Dev Biol 2023; 11:1235217. [PMID: 37601096 PMCID: PMC10436230 DOI: 10.3389/fcell.2023.1235217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5), also known as BMK1 or MAPK7, represents a recent addition to the classical mitogen-activated protein kinase (MAPK) family. This family includes well-known members such as ERK1/2, c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK), as well as atypical MAPKs such as ERK3, ERK4, ERK7 (ERK8), and Nemo-like kinase (NLK). Comprehensive reviews available elsewhere provide detailed insights into ERK5, which interested readers can refer to for in-depth knowledge (Nithianandarajah-Jones et al., 2012; Monti et al., Cancers (Basel), 2022, 14). The primary aim of this review is to emphasize the essential characteristics of ERK5 and shed light on the intricate nature of its activation, with particular attention to the catalytic-independent functions in disease pathways.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
60
|
de Mattos K, Dumas FO, Campolina-Silva GH, Belleannée C, Viger RS, Tremblay JJ. ERK5 Cooperates With MEF2C to Regulate Nr4a1 Transcription in MA-10 and MLTC-1 Leydig Cells. Endocrinology 2023; 164:bqad120. [PMID: 37539861 PMCID: PMC10435423 DOI: 10.1210/endocr/bqad120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/30/2023] [Accepted: 08/02/2023] [Indexed: 08/05/2023]
Abstract
Leydig cells produce hormones required for the development and maintenance of sex characteristics and fertility in males. MEF2 transcription factors are important regulators of Leydig cell gene expression and steroidogenesis. ERK5 is an atypical member of the MAP kinase family that modulates transcription factor activity, either by direct phosphorylation or by acting as a transcriptional coactivator. While MEF2 and ERK5 are known to cooperate transcriptionally, the presence and role of ERK5 in Leydig cells remained unknown. Our goal was to determine whether ERK5 is present in Leydig cells and whether it cooperates with MEF2 to regulate gene expression. We found that ERK5 is present in Leydig cells in testicular tissue and immortalized cell lines. ERK5 knockdown in human chorionic gonadotrophin-treated MA-10 Leydig cells reduced steroidogenesis and decreased Star and Nr4a1 expression. Luciferase assays using a synthetic reporter plasmid containing 3 MEF2 elements revealed that ERK5 enhances MEF2-dependent promoter activation. Although ERK5 did not cooperate with MEF2 on the Star promoter in Leydig cell lines, we found that ERK5 and MEF2C do cooperate on the Nr4a1 promoter, which contains 2 adjacent MEF2 elements. Mutation of each MEF2 element in a short version of the Nr4a1 promoter significantly decreased the ERK5/MEF2C cooperation, indicating that both MEF2 elements need to be intact. The ERK5/MEF2C cooperation did not require phosphorylation of MEF2C on Ser387. Taken together, our data identify ERK5 as a new regulator of MEF2 activity in Leydig cells and provide potential new insights into mechanisms that regulate Leydig cell gene expression and function.
Collapse
Affiliation(s)
- Karine de Mattos
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Félix-Olivier Dumas
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Gabriel Henrique Campolina-Silva
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Clémence Belleannée
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| | - Robert S Viger
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| | - Jacques J Tremblay
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| |
Collapse
|
61
|
Sun J, Ruan Y, Xu J, Shi P, Xu H. Effect of Bovine MEF2A Gene Expression on Proliferation and Apoptosis of Myoblast Cells. Genes (Basel) 2023; 14:1498. [PMID: 37510401 PMCID: PMC10379155 DOI: 10.3390/genes14071498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Myocyte enhancer factor 2A (MEF2A) is a member of the myocyte enhancer factor 2 family. MEF2A is widely distributed in various tissues and organs and participates in various physiological processes. This study aimed to investigate the effect of MEF2A expression on the proliferation and apoptosis of bovine myoblasts. CCK8, ELISA, cell cycle, and apoptosis analyses were conducted to assess cell status. In addition, the mRNA expression levels of genes associated with bovine myoblast proliferation and apoptosis were evaluated using RT-qPCR. The results showed that the upregulation of MEF2A mRNA promoted the proliferation rate of myoblasts, shortened the cycle process, and increased the anti-apoptotic rate. Furthermore, the RT-qPCR results showed that the upregulation of MEF2A mRNA significantly increased the cell proliferation factors MyoD1 and IGF1, cell cycle factors CDK2 and CCNA2, and the apoptotic factors Bcl2 and BAD (p < 0.01). These results show that the MEF2A gene can positively regulate myoblast proliferation and anti-apoptosis, providing a basis for the analysis of the regulatory mechanism of the MEF2A gene on bovine growth and development.
Collapse
Affiliation(s)
- Jinkui Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jiali Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Pengfei Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
62
|
Sato K, Miyauchi Y, Xu X, Kon R, Ikarashi N, Chiba Y, Hosoe T, Sakai H. Platinum-based anticancer drugs-induced downregulation of myosin heavy chain isoforms in skeletal muscle of mouse. J Pharmacol Sci 2023; 152:167-177. [PMID: 37257944 DOI: 10.1016/j.jphs.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023] Open
Abstract
Cisplatin, a platinum-based anticancer drug used frequently in cancer treatment, causes skeletal muscle atrophy. It was predicted that the proteolytic pathway is enhanced as the mechanism of this atrophy. Therefore, we investigated whether a platinum-based anticancer drug affects the expression of the major proteins of skeletal muscle, myosin heavy chain (MyHC). Mice were injected with cisplatin or oxaliplatin for four consecutive days. C2C12 myotubes were treated using cisplatin and oxaliplatin. Administration of platinum-based anticancer drug reduced quadriceps mass and muscle strength compared to the control group. Protein levels of all MyHC isoforms were reduced in the platinum-based anticancer drug groups. However, only Myh2 (MyHC-IIa) gene expression in skeletal muscle of mice treated with platinum-based anticancer drugs was found to be reduced. Treatment of C2C12 myotubes with platinum-based anticancer drugs reduced the protein levels of all MyHCs, and treatment with the proteasome inhibitor MG-132 restored this reduction. The expression of Mef2c, which was predicted to act upstream of Myh2, was reduced in the skeletal muscle of mice treated systemically with platinum-based anticancer drug. Degradation of skeletal muscle MyHCs by proteasomes may be a factor that plays an important role in muscle mass loss in platinum-based anticancer drug-induced muscle atrophy.
Collapse
Affiliation(s)
- Ken Sato
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Yu Miyauchi
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Xinran Xu
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Risako Kon
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Nobutomo Ikarashi
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Tomoo Hosoe
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan; Department of Bioregulatory Science, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan
| | - Hiroyasu Sakai
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 1428501, Japan.
| |
Collapse
|
63
|
Cai C, Yue Y, Yue B. Single-cell RNA sequencing in skeletal muscle developmental biology. Biomed Pharmacother 2023; 162:114631. [PMID: 37003036 DOI: 10.1016/j.biopha.2023.114631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/01/2023] Open
Abstract
Skeletal muscle is the most extensive tissue in mammals, and they perform several functions; it is derived from paraxial mesodermal somites and undergoes hyperplasia and hypertrophy to form multinucleated, contractile, and functional muscle fibers. Skeletal muscle is a complex heterogeneous tissue composed of various cell types that establish communication strategies to exchange biological information; therefore, characterizing the cellular heterogeneity and transcriptional signatures of skeletal muscle is central to understanding its ontogeny's details. Studies of skeletal myogenesis have focused primarily on myogenic cells' proliferation, differentiation, migration, and fusion and ignored the intricate network of cells with specific biological functions. The rapid development of single-cell sequencing technology has recently enabled the exploration of skeletal muscle cell types and molecular events during development. This review summarizes the progress in single-cell RNA sequencing and its applications in skeletal myogenesis, which will provide insights into skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Cuicui Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China; Guyuan Branch, Ningxia Academy of Agriculture and Forestry Sciences, Guyuan 7560000, China
| | - Yuan Yue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China.
| |
Collapse
|
64
|
Pasqualucci L. The germinal center in the pathogenesis of B cell lymphomas. Hematol Oncol 2023; 41 Suppl 1:62-69. [PMID: 37294970 DOI: 10.1002/hon.3141] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
The adaptive immune system has evolved to allow effective responses against a virtually unlimited number of invading pathogens. This process requires the transient formation of germinal centers (GC), a dynamic environment that ensures the generation and selection of B cells capable to produce antibodies with high antigen affinity, or to maintain the memory of that antigen for life. However, this comes at a cost, as the unique events accompanying the GC reaction pose a significant risk to the genome of B cells, which must endure elevated levels of replication stress, while proliferating at high rates and undergoing DNA breaks introduced by somatic hypermutation and class switch recombination. Indeed, the genetic/epigenetic disruption of programs implicated in normal GC biology has emerged as a hallmark of most B cell lymphomas. This improved understanding provides a conceptual framework for the identification of cellular pathways that could be exploited for precision medicine approaches.
Collapse
Affiliation(s)
- Laura Pasqualucci
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, USA
| |
Collapse
|
65
|
Qu R, Zhou M, Qiu Y, Peng Y, Yin X, Liu B, Bi H, Gao Y, Guo D. Glucocorticoids improve the balance of M1/M2 macrophage polarization in experimental autoimmune uveitis through the P38MAPK-MEF2C axis. Int Immunopharmacol 2023; 120:110392. [PMID: 37262960 DOI: 10.1016/j.intimp.2023.110392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Uveitis is a common ocular disease that can induce serious complications and sequelae. It is one of the major causes of blindness. Currently, mounting evidence suggests that glucocorticoids (GCs) can suppress ocular inflammation and promote the healing of damaged ocular tissues, but the underlying mechanism remains unclear. The present study aimed to elucidate the mechanism by which GCs modulate the homeostasis of M1/M2 macrophage polarization in experimental autoimmune uveitis (EAU) through the p38MAPK-MEF2C axis. Female Lewis rats were randomly divided into four groups: a normal control (NC) group, an EAU group, an EAU + glucocorticoid (EAU + GC) group, and an EAU + p38MAPK inhibitor (EAU + SB) group. The EAU model was induced in EAU, EAU + GC, and EAU + SB groups, followed by the treatments of normal saline, GC (predisione), and SB203580, respectively. The findings demonstrated that the rats in GC and SB groups had much less ocular inflammation, and the clinical and pathological scores decreased. Further research revealed that GC and SB treatment could inhibit iNOS and CD86 expression while promoting Arg-1 and CD206 secretion in IRBP-induced uveitis rats. Moreover, we found that the role of GC was similar to the results of SB203580, but the role of GC was masked by the C16-PAF (a p38MAPK activator) treatment. Molecular docking and western blot results confirmed that GC's therapeutic action against EAU is mediated via the p38MAPK-MEF2C axis. It regulates macrophage polarization by encouraging M1 to M2 transition and releasing anti-inflammatory factors.
Collapse
Affiliation(s)
- Ruyi Qu
- Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Mengxian Zhou
- Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Yan Qiu
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Yuan Peng
- Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Xuewei Yin
- Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Bin Liu
- Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Yan'e Gao
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan 250002, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan 250002, China.
| |
Collapse
|
66
|
Vishal K, Barajas Alonso E, DeAguero AA, Waters JA, Chechenova MB, Cripps RM. Phosphorylation of the Myogenic Factor Myocyte Enhancer Factor-2 Impacts Myogenesis In Vivo. Mol Cell Biol 2023; 43:241-253. [PMID: 37184381 PMCID: PMC10251773 DOI: 10.1080/10985549.2023.2198167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 05/16/2023] Open
Abstract
Activity of the myogenic regulatory protein myocyte enhancer factor-2 (MEF2) is modulated by post-translational modification. We investigated the in vivo phosphorylation of Drosophila MEF2, and identified serine 98 (S98) as a phosphorylated residue. Phospho-mimetic (S98E) and phospho-null (S98A) isoforms of MEF2 did not differ from wild-type in their activity in vitro, so we used CRISPR/Cas9 to generate an S98A allele of the endogenous gene. In mutant larvae we observed phenotypes characteristic of reduced MEF2 function, including reduced body wall muscle size and reduced expression of myofibrillar protein genes; conversely,S98A homozygotes showed enhanced MEF2 function through muscle differentiation within the adult myoblasts associated with the wing imaginal disc. In adults, S98A homozygotes were viable with normal mobility, yet showed patterning defects in muscles that were enhanced when the S98A allele was combined with a Mef2 null allele. Overall our data indicate that blocking MEF2 S98 phosphorylation in myoblasts enhances its myogenic capability, whereas blocking S98 phosphorylation in differentiating muscles attenuates MEF2 function. Our studies are among the first to assess the functional significance of MEF2 phosphorylation sites in the intact animal, and suggest that the same modification can have profoundly different effects upon MEF2 function depending upon the developmental context.
Collapse
Affiliation(s)
- Kumar Vishal
- Department of Biology, San Diego State University, San Diego, California, USA
| | | | - Ashley A. DeAguero
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Jennifer A. Waters
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Maria B. Chechenova
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Richard M. Cripps
- Department of Biology, San Diego State University, San Diego, California, USA
| |
Collapse
|
67
|
Chen X, Pacis A, Aracena KA, Gona S, Kwan T, Groza C, Lin YL, Sindeaux R, Yotova V, Pramatarova A, Simon MM, Pastinen T, Barreiro LB, Bourque G. Transposable elements are associated with the variable response to influenza infection. CELL GENOMICS 2023; 3:100292. [PMID: 37228757 PMCID: PMC10203045 DOI: 10.1016/j.xgen.2023.100292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 05/27/2023]
Abstract
Influenza A virus (IAV) infections are frequent every year and result in a range of disease severity. Here, we wanted to explore the potential contribution of transposable elements (TEs) to the variable human immune response. Transcriptome profiling in monocyte-derived macrophages from 39 individuals following IAV infection revealed significant inter-individual variation in viral load post-infection. Using transposase-accessible chromatin using sequencing (ATAC-seq), we identified a set of TE families with either enhanced or reduced accessibility upon infection. Of the enhanced families, 15 showed high variability between individuals and had distinct epigenetic profiles. Motif analysis showed an association with known immune regulators (e.g., BATFs, FOSs/JUNs, IRFs, STATs, NFkBs, NFYs, and RELs) in stably enriched families and with other factors in variable families, including KRAB-ZNFs. We showed that TEs and host factors regulating TEs were predictive of viral load post-infection. Our findings shed light on the role TEs and KRAB-ZNFs may play in inter-individual variation in immunity.
Collapse
Affiliation(s)
- Xun Chen
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - Alain Pacis
- Canadian Center for Computational Genomics, McGill University, Montréal, QC H3A 0G1, Canada
| | | | - Saideep Gona
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Tony Kwan
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC H3A 0G1, Canada
| | - Cristian Groza
- Quantitative Life Science, McGill University, Montréal, QC H3A 1E3, Canada
| | - Yen Lung Lin
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Renata Sindeaux
- Centre de Recherche, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Vania Yotova
- Centre de Recherche, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Albena Pramatarova
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC H3A 0G1, Canada
| | - Marie-Michelle Simon
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC H3A 0G1, Canada
| | - Tomi Pastinen
- Genomic Medicine Center, Children’s Mercy Hospital and Research Institute, Kansas City, MO 64108, USA
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| | - Luis B. Barreiro
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Guillaume Bourque
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan
- Canadian Center for Computational Genomics, McGill University, Montréal, QC H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC H3A 0G1, Canada
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
68
|
Wood S, Ishida K, Hagerty JR, Karahodza A, Dennis JN, Jolly ER. Characterization of Schistosome Sox Genes and Identification of a Flatworm Class of Sox Regulators. Pathogens 2023; 12:690. [PMID: 37242360 PMCID: PMC10222431 DOI: 10.3390/pathogens12050690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Schistosome helminths infect over 200 million people across 78 countries and are responsible for nearly 300,000 deaths annually. However, our understanding of basic genetic pathways crucial for schistosome development is limited. The sex determining region Y-box 2 (Sox2) protein is a Sox B type transcriptional activator that is expressed prior to blastulation in mammals and is necessary for embryogenesis. Sox expression is associated with pluripotency and stem cells, neuronal differentiation, gut development, and cancer. Schistosomes express a Sox-like gene expressed in the schistosomula after infecting a mammalian host when schistosomes have about 900 cells. Here, we characterized and named this Sox-like gene SmSOXS1. SmSoxS1 protein is a developmentally regulated activator that localizes to the anterior and posterior ends of the schistosomula and binds to Sox-specific DNA elements. In addition to SmSoxS1, we have also identified an additional six Sox genes in schistosomes, two Sox B, one SoxC, and three Sox genes that may establish a flatworm-specific class of Sox genes with planarians. These data identify novel Sox genes in schistosomes to expand the potential functional roles for Sox2 and may provide interesting insights into early multicellular development of flatworms.
Collapse
Affiliation(s)
- Stephanie Wood
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA; (S.W.); (K.I.); (J.R.H.)
| | - Kenji Ishida
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA; (S.W.); (K.I.); (J.R.H.)
| | - James R. Hagerty
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA; (S.W.); (K.I.); (J.R.H.)
| | - Anida Karahodza
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA; (S.W.); (K.I.); (J.R.H.)
| | - Janay N. Dennis
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA; (S.W.); (K.I.); (J.R.H.)
| | - Emmitt R. Jolly
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA; (S.W.); (K.I.); (J.R.H.)
- Center for Global Health and Disease, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
69
|
Lim WF, Rinaldi C. RNA Transcript Diversity in Neuromuscular Research. J Neuromuscul Dis 2023:JND221601. [PMID: 37182892 DOI: 10.3233/jnd-221601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Three decades since the Human Genome Project began, scientists have now identified more then 25,000 protein coding genes in the human genome. The vast majority of the protein coding genes (> 90%) are multi-exonic, with the coding DNA being interrupted by intronic sequences, which are removed from the pre-mRNA transcripts before being translated into proteins, a process called splicing maturation. Variations in this process, i.e. by exon skipping, intron retention, alternative 5' splice site (5'ss), 3' splice site (3'ss), or polyadenylation usage, lead to remarkable transcriptome and proteome diversity in human tissues. Given its critical biological importance, alternative splicing is tightly regulated in a tissue- and developmental stage-specific manner. The central nervous system and skeletal muscle are amongst the tissues with the highest number of differentially expressed alternative exons, revealing a remarkable degree of transcriptome complexity. It is therefore not surprising that splicing mis-regulation is causally associated with a myriad of neuromuscular diseases, including but not limited to amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), Duchenne muscular dystrophy (DMD), and myotonic dystrophy type 1 and 2 (DM1, DM2). A gene's transcript diversity has since become an integral and an important consideration for drug design, development and therapy. In this review, we will discuss transcript diversity in the context of neuromuscular diseases and current approaches to address splicing mis-regulation.
Collapse
Affiliation(s)
- Wooi Fang Lim
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Carlo Rinaldi
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| |
Collapse
|
70
|
de Araujo Tavares ME, Cupertino RB, Bandeira CE, da Silva BS, Vitola ES, Salgado CAI, Dos Santos Soares R, Picon FA, Rohde LA, Rovaris DL, Grevet EH, Bau CHD. Refining patterns of MEF2C effects in white matter microstructure and psychiatric features. J Neural Transm (Vienna) 2023; 130:697-706. [PMID: 37002331 DOI: 10.1007/s00702-023-02626-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
Several GWAS reported Myocyte Enhancer Factor 2 C (MEF2C) gene associations with white matter microstructure and psychiatric disorders, and MEF2C involvement in pathways related to neuronal development suggests a common biological factor underlying these phenotypes. We aim to refine the MEF2C effects in the brain relying on an integrated analysis of white matter and psychiatric phenotypes in an extensively characterized sample. This study included 870 Brazilian adults (47% from an attention-deficit/hyperactivity disorder outpatient clinic) assessed through standardized psychiatric interviews, 139 of which underwent a magnetic resonance imaging scan. We evaluated variants in the MEF2C region using two approaches: 1) a gene-wide analysis, which uses the sum of polymorphism effects, and 2) SNP analyses, restricted to the independent variants within the gene. The outcomes included psychiatric phenotypes and fractional anisotropy for brain images. Results: The gene-wide analyses pointed to a nominal association between MEF2C and the Temporal Portion of the Superior Longitudinal Fasciculus (SLFTEMP). The SNP analysis identified four independent variants significantly associated with SLFTEMP and one (rs4218438) with Substance Use Disorder. Our findings showing specific associations of MEF2C variants with temporal-frontal circuitry components may help to elucidate how the MEF2C gene underlies a broad range of psychiatric phenotypes since these regions are relevant to executive and cognitive functions.
Collapse
Affiliation(s)
- Maria Eduarda de Araujo Tavares
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | - Cibele Edom Bandeira
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Bruna Santos da Silva
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Eduardo Schneider Vitola
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carlos Alberto Iglesias Salgado
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Robson Dos Santos Soares
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Felipe Almeida Picon
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Luis Augusto Rohde
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- National Institute of Developmental Psychiatry, São Paulo, Brazil
| | - Diego Luiz Rovaris
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
| | - Eugenio Horacio Grevet
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Claiton Henrique Dotto Bau
- Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul (UFRGS), Avenida Bento Gonçalves, 9500, Porto Alegre, RS, 91501-970, Brazil.
- Adulthood ADHD Outpatient Program (ProDAH), Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| |
Collapse
|
71
|
Bahrami F, Fathi M, Ahmadvand H, Pajohi N. Endurance training changes the expression of miR-1 and miR-133 and predicted genes in slow and fast twitch muscles. Arch Gerontol Geriatr 2023; 108:104929. [PMID: 36645970 DOI: 10.1016/j.archger.2023.104929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
PURPOSE OF THE RESEARCH Endurance training can modify signaling and gene expression pathways that play a pivotal role in determining the phenotype of the fibers. The present study aimed to investigate the effects of endurance training on the expression of some myomiRs and related genes in slow and fast twitch muscles. METHODS Twenty healthy male adult Wistar rats (281 ± 14 g) were randomized to either control (n = 10) or treated (n = 10). The treated group performed an endurance program for eight weeks (running on a treadmill for eight weeks, 50 min, 23 m/min). After the end of the training protocol, the slow (soleus) and fast (EDL) twitch muscles were removed to assess the miR-1, miR-133 expression, and hdac4, mef2c genes, and protein by real-time PCR and western blot, respectively. RESULTS The soleus muscle miR-1 expression and mef2c gene in the treated group were significantly lower compared control (p = 0.0001). In contrast, miR-133 and hdac4 gene expression of the soleus muscle of the treated group increased significantly (p = 0001), and the EDL miR-133 and mef2c expression of the treated group increased in the compared control group (p = 0.0001). The EDL MEF2c protein expression in the treated group significantly decreased compared to the control group, although the expression of EDL HDAC4 protein significantly increased (p = 0.0001). CONCLUSIONS Endurance training changes the expression of the miR-1, miR-133, and their predicted genes in slow and fast twitch muscles. Also, the rate of HDAC4 and MEF2c protein synthesis, which are upstream and downstream of these myomiRs, was affected by endurance training.
Collapse
Affiliation(s)
- Farid Bahrami
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khorramabad, Iran
| | - Mohammad Fathi
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khorramabad, Iran.
| | - Hassan Ahmadvand
- Faculty of Medical Sciences, Lorestan University of Medical Sciences, Khorammabad, Iran
| | - Naser Pajohi
- Faculty of Medical Sciences, Lorestan University of Medical Sciences, Khorammabad, Iran
| |
Collapse
|
72
|
Kim YJ, Oh J, Jung S, Kim CJ, Choi J, Jeon YK, Kim HJ, Kim JW, Suh CH, Lee Y, Im SH, Crotty S, Choi YS. The transcription factor Mef2d regulates B:T synapse-dependent GC-T FH differentiation and IL-21-mediated humoral immunity. Sci Immunol 2023; 8:eadf2248. [PMID: 36961907 PMCID: PMC10311795 DOI: 10.1126/sciimmunol.adf2248] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/15/2023] [Indexed: 03/26/2023]
Abstract
Communication between CD4 T cells and cognate B cells is key for the former to fully mature into germinal center-T follicular helper (GC-TFH) cells and for the latter to mount a CD4 T cell-dependent humoral immune response. Although this interaction occurs in a B:T synapse-dependent manner, how CD4 T cells transcriptionally regulate B:T synapse formation remains largely unknown. Here, we report that Mef2d, an isoform of the myocyte enhancer factor 2 (Mef2) transcription factor family, is a critical regulator of this process. In CD4 T cells, Mef2d negatively regulates expression of Sh2d1a, which encodes SLAM-associated protein (SAP), a critical regulator of B:T synapses. We found that Mef2d regulates Sh2d1a expression via DNA binding-dependent transcriptional repression, inhibiting SAP-dependent B:T synapse formation and preventing antigen-specific CD4 T cells from differentiating into GC-TFH cells. Mef2d also impeded IL-21 production by CD4 T cells, an important B cell help signaling molecule, via direct repression of the Il21 gene. In contrast, CD4 T cell-specific disruption of Mef2d led to a substantial increase in GC-TFH differentiation in response to protein immunization, concurrent with enhanced SAP expression. MEF2D mRNA expression inversely correlates with human systemic lupus erythematosus (SLE) patient autoimmune parameters, including circulating TFH-like cell frequencies, autoantibodies, and SLEDAI scores. These findings highlight Mef2d as a pivotal rheostat in CD4 T cells for controlling GC formation and antibody production by B cells.
Collapse
Affiliation(s)
- Ye-Ji Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jeein Oh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Soohan Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Johng Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Jinyong Choi
- Department of Microbiology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jik Kim
- Department of Otorhinolaryngology, Seoul National University Hospital, Seoul, Korea
| | - Ji-Won Kim
- Department of Rheumatology, Ajou University School of Medicine, Gyeonggi-do, Korea
| | - Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Gyeonggi-do, Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Korea
- ImmunoBiome Inc., Pohang, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Korea
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- University of California San Diego, Department of Medicine, Division of Infectious Diseases and Global Public Health, La Jolla, CA, USA
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul, Korea
- Transplantation Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
73
|
Missinato MA, Murphy S, Lynott M, Yu MS, Kervadec A, Chang YL, Kannan S, Loreti M, Lee C, Amatya P, Tanaka H, Huang CT, Puri PL, Kwon C, Adams PD, Qian L, Sacco A, Andersen P, Colas AR. Conserved transcription factors promote cell fate stability and restrict reprogramming potential in differentiated cells. Nat Commun 2023; 14:1709. [PMID: 36973293 PMCID: PMC10043290 DOI: 10.1038/s41467-023-37256-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
Defining the mechanisms safeguarding cell fate identity in differentiated cells is crucial to improve 1) - our understanding of how differentiation is maintained in healthy tissues or altered in a disease state, and 2) - our ability to use cell fate reprogramming for regenerative purposes. Here, using a genome-wide transcription factor screen followed by validation steps in a variety of reprogramming assays (cardiac, neural and iPSC in fibroblasts and endothelial cells), we identified a set of four transcription factors (ATF7IP, JUNB, SP7, and ZNF207 [AJSZ]) that robustly opposes cell fate reprogramming in both lineage and cell type independent manners. Mechanistically, our integrated multi-omics approach (ChIP, ATAC and RNA-seq) revealed that AJSZ oppose cell fate reprogramming by 1) - maintaining chromatin enriched for reprogramming TF motifs in a closed state and 2) - downregulating genes required for reprogramming. Finally, KD of AJSZ in combination with MGT overexpression, significantly reduced scar size and improved heart function by 50%, as compared to MGT alone post-myocardial infarction. Collectively, our study suggests that inhibition of barrier to reprogramming mechanisms represents a promising therapeutic avenue to improve adult organ function post-injury.
Collapse
Affiliation(s)
- Maria A Missinato
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Michaela Lynott
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Michael S Yu
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Anaïs Kervadec
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Yu-Ling Chang
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mafalda Loreti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Christopher Lee
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Prashila Amatya
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Hiroshi Tanaka
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Chun-Teng Huang
- Viral Vector Core Facility Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Peter D Adams
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Alexandre R Colas
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
74
|
Odaira K, Yasuda T, Okada K, Shimooka T, Kojima Y, Noura M, Tamura S, Kurahashi S, Iwamoto E, Sanada M, Matsumura I, Miyazaki Y, Kojima T, Kiyoi H, Tsuzuki S, Hayakawa F. Functional inhibition of MEF2 by C/EBP is a possible mechanism of leukemia development by CEBP-IGH fusion gene. Cancer Sci 2023; 114:781-792. [PMID: 36341510 PMCID: PMC9986073 DOI: 10.1111/cas.15641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
CEBPA-IGH, a fusion gene of the immunoglobulin heavy-chain locus (IGH) and the CCAAT enhancer-binding protein α (C/EBPα) gene, is recurrently found in B-ALL cases and causes aberrant expression of C/EBPα, a master regulator of granulocyte differentiation, in B cells. Forced expression of C/EBPα in B cells was reported to cause loss of B-cell identity due to the inhibition of Pax5, a master regulator of B-cell differentiation; however, it is not known whether the same mechanism is applicable for B-ALL development by CEBPA-IGH. It is known that a full-length isoform of C/EBPα, p42, promotes myeloid differentiation, whereas its N-terminal truncated isoform, p30, inhibits myeloid differentiation through the inhibition of p42; however, the differential role between p42 and p30 in ALL development has not been clarified. In the present study, we examined the effect of the expression of p42 and p30 in B cells by performing RNA-seq of mRNA from LCL stably transfected with p42 or p30. Unexpectedly, suppression of PAX5 target genes was barely observed. Instead, both isoforms suppressed the target genes of MEF2 family members (MEF2s), other regulators of B-cell differentiation. Similarly, MEF2s target genes rather than PAX5 target genes were suppressed in CEBP-IGH-positive ALL (n = 8) compared with other B-ALL (n = 315). Furthermore, binding of both isoforms to MEF2s target genes and the reduction of surrounding histone acetylation were observed in ChIP-qPCR. Our data suggest that the inhibition of MEF2s by C/EBPα plays a role in the development of CEBPA-IGH-positive ALL and that both isoforms work co-operatively to achieve it.
Collapse
Affiliation(s)
- Koya Odaira
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiko Yasuda
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Kentaro Okada
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Shimooka
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukino Kojima
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mina Noura
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shogo Tamura
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Kurahashi
- Division of Hematology and Oncology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Eisuke Iwamoto
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Masashi Sanada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tetsuhito Kojima
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Aichi Health Promotion Foundation, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Fumihiko Hayakawa
- Department of Integrated Health Sciences, Division of Cellular and Genetic Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
75
|
The Role of MEF2 Transcription Factor Family in Neuronal Survival and Degeneration. Int J Mol Sci 2023; 24:ijms24043120. [PMID: 36834528 PMCID: PMC9963821 DOI: 10.3390/ijms24043120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
The family of myocyte enhancer factor 2 (MEF2) transcription factors comprises four highly conserved members that play an important role in the nervous system. They appear in precisely defined time frames in the developing brain to turn on and turn off genes affecting growth, pruning and survival of neurons. MEF2s are known to dictate neuronal development, synaptic plasticity and restrict the number of synapses in the hippocampus, thus affecting learning and memory formation. In primary neurons, negative regulation of MEF2 activity by external stimuli or stress conditions is known to induce apoptosis, albeit the pro or antiapoptotic action of MEF2 depends on the neuronal maturation stage. By contrast, enhancement of MEF2 transcriptional activity protects neurons from apoptotic death both in vitro and in preclinical models of neurodegenerative diseases. A growing body of evidence places this transcription factor in the center of many neuropathologies associated with age-dependent neuronal dysfunctions or gradual but irreversible neuron loss. In this work, we discuss how the altered function of MEF2s during development and in adulthood affecting neuronal survival may be linked to neuropsychiatric disorders.
Collapse
|
76
|
Bomkamp C, Musgrove L, Marques DMC, Fernando GF, Ferreira FC, Specht EA. Differentiation and Maturation of Muscle and Fat Cells in Cultivated Seafood: Lessons from Developmental Biology. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1-29. [PMID: 36374393 PMCID: PMC9931865 DOI: 10.1007/s10126-022-10174-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cultivated meat, also known as cultured or cell-based meat, is meat produced directly from cultured animal cells rather than from a whole animal. Cultivated meat and seafood have been proposed as a means of mitigating the substantial harms associated with current production methods, including damage to the environment, antibiotic resistance, food security challenges, poor animal welfare, and-in the case of seafood-overfishing and ecological damage associated with fishing and aquaculture. Because biomedical tissue engineering research, from which cultivated meat draws a great deal of inspiration, has thus far been conducted almost exclusively in mammals, cultivated seafood suffers from a lack of established protocols for producing complex tissues in vitro. At the same time, fish such as the zebrafish Danio rerio have been widely used as model organisms in developmental biology. Therefore, many of the mechanisms and signaling pathways involved in the formation of muscle, fat, and other relevant tissue are relatively well understood for this species. The same processes are understood to a lesser degree in aquatic invertebrates. This review discusses the differentiation and maturation of meat-relevant cell types in aquatic species and makes recommendations for future research aimed at recapitulating these processes to produce cultivated fish and shellfish.
Collapse
Affiliation(s)
- Claire Bomkamp
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Lisa Musgrove
- University of the Sunshine Coast, Sippy Downs, Queensland Australia
| | - Diana M. C. Marques
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gonçalo F. Fernando
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Frederico C. Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Elizabeth A. Specht
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| |
Collapse
|
77
|
Samak M, Kues A, Kaltenborn D, Klösener L, Mietsch M, Germena G, Hinkel R. Dysregulation of Krüppel-like Factor 2 and Myocyte Enhancer Factor 2D Drive Cardiac Microvascular Inflammation and Dysfunction in Diabetes. Int J Mol Sci 2023; 24:2482. [PMID: 36768805 PMCID: PMC9916909 DOI: 10.3390/ijms24032482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Cardiovascular complications are the main cause of morbidity and mortality from diabetes. Herein, vascular inflammation is a major pathological manifestation. We previously characterized the cardiac microvascular inflammatory phenotype in diabetic patients and highlighted micro-RNA 92a (miR-92a) as a driver of endothelial dysfunction. In this article, we further dissect the molecular underlying of these findings by addressing anti-inflammatory Krüppel-like factors 2 and 4 (KLF2 and KLF4). We show that KLF2 dysregulation in diabetes correlates with greater monocyte adhesion as well as migratory defects in cardiac microvascular endothelial cells. We also describe, for the first time, a role for myocyte enhancer factor 2D (MEF2D) in cardiac microvascular dysfunction in diabetes. We show that both KLFs 2 and 4, as well as MEF2D, are dysregulated in human and porcine models of diabetes. Furthermore, we prove a direct interaction between miR-92a and all three targets. Altogether, our data strongly qualify miR-92a as a potential therapeutic target for diabetes-associated cardiovascular disease.
Collapse
Affiliation(s)
- Mostafa Samak
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Andreas Kues
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
| | - Diana Kaltenborn
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
| | - Lina Klösener
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine, 30173 Hannover, Germany
| | - Matthias Mietsch
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Giulia Germena
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine, 30173 Hannover, Germany
| |
Collapse
|
78
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
79
|
Shi P, Ruan Y, Liu W, Sun J, Xu J, Xu H. Analysis of Promoter Methylation of the Bovine FOXO1 Gene and Its Effect on Proliferation and Differentiation of Myoblasts. Animals (Basel) 2023; 13:ani13020319. [PMID: 36670858 PMCID: PMC9854826 DOI: 10.3390/ani13020319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/01/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
This study aimed to explore the regulatory role of FOXO1 promoter methylation on its transcriptional level and unravel the effect of FOXO1 on the proliferation and differentiation of bovine myoblasts. Bisulfite sequencing polymerase chain reaction (BSP) and real-time quantitative PCR were performed to determine the methylation status and transcript levels of the FOXO1 promoter region at different growth stages. BSP results showed that the methylation level in the calf bovine (CB) group was significantly higher than that in the adult bovine (AB) group (p < 0.05). On the other hand, qRT-PCR results indicated that the mRNA expression level in the AB group was significantly higher than that in the CB group (p < 0.05), suggesting a significant decrease in gene expression at high levels of DNA methylation. CCK-8 and flow cytometry were applied to determine the effect of silencing the FOXO1 gene on the proliferation of bovine myoblasts. Furthermore, qRT-PCR and Western blot were conducted to analyze the expression of genes associated with the proliferation and differentiation of bovine myoblasts. Results from CCK-8 revealed that the short hairpin FOXO1 (shFOXO1) group significantly promoted the proliferation of myoblasts compared to the short-hairpin negative control (shNC) group (p < 0.05). Flow cytometry results showed a significant decrease in the number of the G1 phase cells (p < 0.05) and a significant increase in the number of the S phase cells (p < 0.05) in the shFOXO1 group compared to the shNC group. In addition, the expression of key genes for myoblast proliferation (CDK2, PCNA, and CCND1) and differentiation (MYOG, MYOD, and MYHC) was significantly increased at both mRNA and protein levels (p < 0.05). In summary, this study has demonstrated that FOXO1 transcription is regulated by methylation in the promoter region and that silencing FOXO1 promotes the proliferation and differentiation of bovine myoblasts. Overall, our findings lay the foundation for further studies on the regulatory role of epigenetics in the development of bovine myoblasts.
Collapse
Affiliation(s)
- Pengfei Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Wenjiao Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jinkui Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jiali Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
- Correspondence:
| |
Collapse
|
80
|
Lee SM, Lee JW, Kim I, Woo DC, Pack CG, Sung YH, Baek IJ, Jung CH, Kim YH, Ha CH. Angiogenic adipokine C1q-TNF-related protein 9 ameliorates myocardial infarction via histone deacetylase 7-mediated MEF2 activation. SCIENCE ADVANCES 2022; 8:eabq0898. [PMID: 36459558 PMCID: PMC10936044 DOI: 10.1126/sciadv.abq0898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 10/18/2022] [Indexed: 06/17/2023]
Abstract
C1q/tumor necrosis factor-related protein 9 (CTRP9) is an adipokine and has high potential as a therapeutic target. However, the role of CTRP9 in cardiovascular disease pathogenesis remains unclear. We found CTRP9 to induce HDAC7 and p38 MAPK phosphorylation via tight regulation of AMPK in vascular endothelial cells, leading to angiogenesis through increased MEF2 activity. The expression of CTRP9 and atheroprotective MEF2 was decreased in plaque tissue of atherosclerotic patients and the ventricle of post-infarction mice. CTRP9 treatment inhibited the formation of atherosclerotic plaques in ApoE KO and CTRP9 KO mice. In addition, CTRP9 induced significant ischemic injury prevention in the post-MI mice. Clinically, serum CTRP9 levels were reduced in patients with MI compared with healthy controls. In summary, CTRP9 induces a vasoprotective response via the AMPK/HDAC7/p38 MAPK pathway in vascular endothelial cells, whereas its absence can contribute to atherosclerosis and MI. Hence, CTRP9 may represent a valuable therapeutic target and biomarker in cardiovascular diseases.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inki Kim
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Young Hoon Sung
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - In-Jeoung Baek
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chang Hee Jung
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Hak Kim
- Cardiology Division, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Hoon Ha
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
81
|
Shao X, Gong W, Wang Q, Wang P, Shi T, Mahmut A, Qin J, Yao Y, Yan W, Chen D, Chen X, Jiang Q, Guo B. Atrophic skeletal muscle fibre-derived small extracellular vesicle miR-690 inhibits satellite cell differentiation during ageing. J Cachexia Sarcopenia Muscle 2022; 13:3163-3180. [PMID: 36237168 PMCID: PMC9745557 DOI: 10.1002/jcsm.13106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sarcopenia is a common and progressive skeletal muscle disorder characterized by atrophic muscle fibres and contractile dysfunction. Accumulating evidence shows that the number and function of satellite cells (SCs) decline and become impaired during ageing, which may contribute to impaired regenerative capacity. A series of myokines/small extracellular vesicles (sEVs) released from muscle fibres regulate metabolism in muscle and extramuscular tissues in an autocrine/paracrine/endocrine manner during muscle atrophy. It is still unclear whether myokines/sEVs derived from muscle fibres can affect satellite cell function during ageing. METHODS Aged mice were used to investigate changes in the myogenic capacity of SCs during ageing-induced muscle atrophy. The effects of atrophic myotube-derived sEVs on satellite cell differentiation were investigated by biochemical methods and immunofluorescence staining. Small RNA sequencing was performed to identify differentially expressed sEV microRNAs (miRNAs) between the control myotubes and atrophic myotubes. The target genes of the miRNA were predicted by bioinformatics analysis and verified by luciferase activity assays. The effects of identified miRNA on the myogenic capacity of SCs in vivo were investigated by intramuscular injection of adeno-associated virus (AAV) to overexpress or silence miRNA in skeletal muscle. RESULTS Our study showed that the myogenic capacity of SCs was significantly decreased (50%, n = 6, P < 0.001) in the tibialis anterior muscle of aged mice. We showed that atrophic myotube-derived sEVs inhibited satellite cell differentiation in vitro (n = 3, P < 0.001) and in vivo (35%, n = 6, P < 0.05). We also found that miR-690 was the most highly enriched miRNA among all the screened sEV miRNAs in atrophic myotubes [Log2 (Fold Change) = 7, P < 0.001], which was verified in the atrophic muscle of aged mice (threefold, n = 6, P < 0.001) and aged men with mean age of 71 ± 5.27 years (2.8-fold, n = 10, P < 0.001). MiR-690 can inhibit myogenic capacity of SCs by targeting myocyte enhancer factor 2, including Mef2a, Mef2c and Mef2d, in vitro (n = 3, P < 0.05) and in vivo (n = 6, P < 0.05). Specific silencing of miR-690 in the muscle can promote satellite cell differentiation (n = 6, P < 0.001) and alleviate muscle atrophy in aged mice (n = 6, P < 0.001). CONCLUSIONS Our study demonstrated that atrophic muscle fibre-derived sEV miR-690 may inhibit satellite cell differentiation by targeting myocyte enhancer factor 2 during ageing.
Collapse
Affiliation(s)
- Xiaoyan Shao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Wang Gong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qianjin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Pu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Abdurahman Mahmut
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jianghui Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yao Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenjin Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Dongyang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
82
|
Liu M, Liu Y, Li X, Pei M, Han M, Qi F. Dexmedetomidine inhibits abnormal muscle hypertrophy of myofascial trigger points via TNF-α/ NF-κB signaling pathway in rats. Front Pharmacol 2022; 13:1031804. [PMID: 36408215 PMCID: PMC9669483 DOI: 10.3389/fphar.2022.1031804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Myofascial pain syndrome (MPS) is a chronic pain disorder with inflammation-related primarily characterized by the presence of myofascial trigger points (MTrPs). Myocyte enhancer factor 2C (MEF2C) is involved in the occurrence of a variety of skeletal muscle diseases. However, it is not yet clear if MEF2C is involved in MTrPs. The purpose of this study was to investigate whether MEF2C was involved in the inflammatory pathogenesis of MTrPs. In the present study, we used RNA sequencing (RNA-seq) to compare the differential expression of myocyte enhancer factor 2C (MEF2C) in healthy participants and MTrPs participants. The widely used rat MTrPs model was established to research the upstream and downstream regulatory mechanism of MEF2C and found that MEF2C was significantly increased in patients with MTrPs. Dexmedetomidine (Dex) was injected intramuscularly in the MTrPs animal to assess its effects on MEF2C. The expression of MEF2C protein and mRNA in skeletal muscle of rats in the MTrPs group were up-regulated. In addition, the expression of TNF- α, p-P65, MLCK, and Myocilin (MyoC) was up-regulated and the mechanical pain threshold was decreased. Peripheral TNF- α injection significantly decreased the mechanical pain threshold and increased the expression of p-P65, MLCK, MEF2C, and MyoC in healthy rats. Maslinic acid increased the mechanical pain threshold and inhibited the expression of p-P65, MLCK, MEF2C, and MyoC. In addition, peripheral injection of DEX in MTrPs rats also inhibited the expression of TNF- α, p-P65, MLCK, MEF2C, and MyoC. These results suggest that MEF2C is involved in the inflammatory pathogenesis of MTrPs and DEX serves as a potential therapeutic strategy for the treatment of MPS.
Collapse
Affiliation(s)
- Mingjian Liu
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yu Liu
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xuan Li
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Miao Pei
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
- Department of Anesthesiology Clinic, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, China
| | - Mei Han
- Department of the Quality Management, The Second Hospital of Shandong University, Jinan, China
- *Correspondence: Mei Han, ; Feng Qi,
| | - Feng Qi
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Mei Han, ; Feng Qi,
| |
Collapse
|
83
|
Korlimarla A, Ps H, Prabhu J, Ragulan C, Patil Y, Vp S, Desai K, Mathews A, Appachu S, Diwakar RB, Bs S, Melcher A, Cheang M, Sadanandam A. Comprehensive characterization of immune landscape of Indian and Western triple negative breast cancers. Transl Oncol 2022; 25:101511. [PMID: 35964339 PMCID: PMC9386467 DOI: 10.1016/j.tranon.2022.101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 11/01/2022] Open
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is a heterogeneous disease with a significant challenge to effectively manage in the clinic worldwide. Immunotherapy may be beneficial to TNBC patients if responders can be effectively identified. Here we sought to elucidate the immune landscape of TNBCs by stratifying patients into immune-specific subtypes (immunotypes) to decipher the molecular and cellular presentations and signaling events of this heterogeneous disease and associating them with their clinical outcomes and potential treatment options. EXPERIMENTAL DESIGN We profiled 730 immune genes in 88 retrospective Indian TNBC samples using the NanoString platform, established immunotypes using non-negative matrix factorization-based machine learning approach, and validated them using Western TNBCs (n=422; public datasets). Immunotype-specific gene signatures were associated with clinicopathological features, immune cell types, biological pathways, acute/chronic inflammatory responses, and immunogenic cell death processes. Responses to different immunotherapies associated with TNBC immunotypes were assessed using cross-cancer comparison to melanoma (n=504). Tumor-infiltrating lymphocytes (TILs) and pan-macrophage spatial marker expression were evaluated. RESULTS We identified three robust transcriptome-based immunotypes in both Indian and Western TNBCs in similar proportions. Immunotype-1 tumors, mainly representing well-known claudin-low and immunomodulatory subgroups, harbored dense TIL infiltrates and T-helper-1 (Th1) response profiles associated with smaller tumors, pre-menopausal status, and a better prognosis. They displayed a cascade of events, including acute inflammation, damage-associated molecular patterns, T-cell receptor-related and chemokine-specific signaling, antigen presentation, and viral-mimicry pathways. On the other hand, immunotype-2 was enriched for Th2/Th17 responses, CD4+ regulatory cells, basal-like/mesenchymal immunotypes, and an intermediate prognosis. In contrast to the two T-cell enriched immunotypes, immunotype-3 patients expressed innate immune genes/proteins, including those representing myeloid infiltrations (validated by spatial immunohistochemistry), and had poor survival. Remarkably, a cross-cancer comparison analysis revealed the association of immunotype-1 with responses to anti-PD-L1 and MAGEA3 immunotherapies. CONCLUSION Overall, the TNBC immunotypes identified in TNBCs reveal different prognoses, immune infiltrations, signaling, acute/chronic inflammation leading to immunogenic cell death of cancer cells, and potentially distinct responses to immunotherapies. The overlap in immune characteristics in Indian and Western TNBCs suggests similar efficiency of immunotherapy in both populations if strategies to select patients according to immunotypes can be further optimized and implemented.
Collapse
Affiliation(s)
- Aruna Korlimarla
- St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India; Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Hari Ps
- St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India; Sri Shankara Cancer Hospital and Research Centre, Bangalore, India; Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Jyoti Prabhu
- St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Chanthirika Ragulan
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Yatish Patil
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Snijesh Vp
- St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Krisha Desai
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Aju Mathews
- MOSC Medical College, Kolenchery, Kerala, India
| | - Sandhya Appachu
- Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Ravi B Diwakar
- Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Srinath Bs
- Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Alan Melcher
- Centre for Translational Immunotherapy, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Maggie Cheang
- Clinical Trials and Statistical Unit, The Institute of Cancer Research, London, UK
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK; Centre for Translational Immunotherapy, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK; Centre for Global Oncology, Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK.
| |
Collapse
|
84
|
Yenamandra AK, Smith RB, Senaratne TN, Kang SHL, Fink JM, Corboy G, Hodge CA, Lu X, Mathew S, Crocker S, Fang M. Evidence-based review of genomic aberrations in diffuse large B cell lymphoma, not otherwise specified (DLBCL, NOS): Report from the cancer genomics consortium lymphoma working group. Cancer Genet 2022; 268-269:1-21. [PMID: 35970109 DOI: 10.1016/j.cancergen.2022.07.006] [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: 05/02/2022] [Revised: 06/26/2022] [Accepted: 07/31/2022] [Indexed: 01/25/2023]
Abstract
Diffuse large B cell lymphoma, not otherwise specified (DLBCL, NOS) is the most common type of non-Hodgkin lymphoma (NHL). The 2016 World Health Organization (WHO) classification defined DLBCL, NOS and its subtypes based on clinical findings, morphology, immunophenotype, and genetics. However, even within the WHO subtypes, it is clear that additional clinical and genetic heterogeneity exists. Significant efforts have been focused on utilizing advanced genomic technologies to further subclassify DLBCL, NOS into clinically relevant subtypes. These efforts have led to the implementation of novel algorithms to support optimal risk-oriented therapy and improvement in the overall survival of DLBCL patients. We gathered an international group of experts to review the current literature on DLBCL, NOS, with respect to genomic aberrations and the role they may play in the diagnosis, prognosis and therapeutic decisions. We comprehensively surveyed clinical laboratory directors/professionals about their genetic testing practices for DLBCL, NOS. The survey results indicated that a variety of diagnostic approaches were being utilized and that there was an overwhelming interest in further standardization of routine genetic testing along with the incorporation of new genetic testing modalities to help guide a precision medicine approach. Additionally, we present a comprehensive literature summary on the most clinically relevant genomic aberrations in DLBCL, NOS. Based upon the survey results and literature review, we propose a standardized, tiered testing approach which will help laboratories optimize genomic testing in order to provide the maximum information to guide patient care.
Collapse
Affiliation(s)
- Ashwini K Yenamandra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37215, United States.
| | | | - T Niroshi Senaratne
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Sung-Hae L Kang
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - James M Fink
- Department of Pathology and Laboratory Medicine, Hennepin Healthcare, Minneapolis, MN, United States
| | - Gregory Corboy
- Haematology, Pathology Queensland, Herston, Queensland, Australia; Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand; School of Clinical Sciences, Monash University, Clayton, Vic, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, Vic, Australia
| | - Casey A Hodge
- Department of Pathology and Immunology, Barnes Jewish Hospital, St. Louis, MO, United States
| | - Xinyan Lu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Susan Mathew
- Department of Pathology, Weill Cornell Medicine, New York, NY, United States
| | - Susan Crocker
- Department of Pathology and Molecular Medicine, Kingston Health Sciences Centre, Queen's University, Kingston, ON, Canada
| | - Min Fang
- Fred Hutchinson Cancer Center and University of Washington, Seattle, WA, United States
| |
Collapse
|
85
|
Pingul BY, Huang H, Chen Q, Alikarami F, Zhang Z, Qi J, Bernt KM, Berger SL, Cao Z, Shi J. Dissection of the MEF2D-IRF8 transcriptional circuit dependency in acute myeloid leukemia. iScience 2022; 25:105139. [PMID: 36193052 PMCID: PMC9526175 DOI: 10.1016/j.isci.2022.105139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 08/05/2022] [Accepted: 09/10/2022] [Indexed: 11/26/2022] Open
Abstract
Transcriptional dysregulation is a prominent feature in leukemia. Here, we systematically surveyed transcription factor (TF) vulnerabilities in leukemia and uncovered TF clusters that exhibit context-specific vulnerabilities within and between different subtypes of leukemia. Among these TF clusters, we demonstrated that acute myeloid leukemia (AML) with high IRF8 expression was addicted to MEF2D. MEF2D and IRF8 form an autoregulatory loop via direct binding to mutual enhancer elements. One important function of this circuit in AML is to sustain PU.1/MEIS1 co-regulated transcriptional outputs via stabilizing PU.1’s chromatin occupancy. We illustrated that AML could acquire dependency on this circuit through various oncogenic mechanisms that results in the activation of their enhancers. In addition to forming a circuit, MEF2D and IRF8 can also separately regulate gene expression, and dual perturbation of these two TFs leads to a more robust inhibition of AML proliferation. Collectively, our results revealed a TF circuit essential for AML survival. MEF2D is a context-specific vulnerability in IRF8hi AML MEF2D and IRF8 form a transcriptional circuit via binding to each other’s enhancers MEF2D-IRF8 circuit supports PU.1’s chromatin occupancy and transcriptional output MEF2D and IRF8 can regulate separate gene expression programs alongside the circuit
Collapse
|
86
|
Bailon-Zambrano R, Sucharov J, Mumme-Monheit A, Murry M, Stenzel A, Pulvino AT, Mitchell JM, Colborn KL, Nichols JT. Variable paralog expression underlies phenotype variation. eLife 2022; 11:e79247. [PMID: 36134886 PMCID: PMC9555865 DOI: 10.7554/elife.79247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Human faces are variable; we look different from one another. Craniofacial disorders further increase facial variation. To understand craniofacial variation and how it can be buffered, we analyzed the zebrafish mef2ca mutant. When this transcription factor encoding gene is mutated, zebrafish develop dramatically variable craniofacial phenotypes. Years of selective breeding for low and high penetrance of mutant phenotypes produced strains that are either resilient or sensitive to the mef2ca mutation. Here, we compared gene expression between these strains, which revealed that selective breeding enriched for high and low mef2ca paralog expression in the low- and high-penetrance strains, respectively. We found that mef2ca paralog expression is variable in unselected wild-type zebrafish, motivating the hypothesis that heritable variation in paralog expression underlies mutant phenotype severity and variation. In support, mutagenizing the mef2ca paralogs, mef2aa, mef2b, mef2cb, and mef2d demonstrated modular buffering by paralogs. Specifically, some paralogs buffer severity while others buffer variability. We present a novel, mechanistic model for phenotypic variation where variable, vestigial paralog expression buffers development. These studies are a major step forward in understanding the mechanisms of facial variation, including how some genetically resilient individuals can overcome a deleterious mutation.
Collapse
Affiliation(s)
- Raisa Bailon-Zambrano
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Juliana Sucharov
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Abigail Mumme-Monheit
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Matthew Murry
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Amanda Stenzel
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Anthony T Pulvino
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Jennyfer M Mitchell
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kathryn L Colborn
- Department of Surgery, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - James T Nichols
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
87
|
Liu C, Leng J, Li Y, Ge T, Li J, Chen Y, Guo C, Qi J. A spatiotemporal atlas of organogenesis in the development of orchid flowers. Nucleic Acids Res 2022; 50:9724-9737. [PMID: 36095130 PMCID: PMC9508851 DOI: 10.1093/nar/gkac773] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/19/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022] Open
Abstract
Development of floral organs exhibits complex molecular mechanisms involving the co-regulation of many genes specialized and precisely functioning in various tissues and developing stages. Advance in spatial transcriptome technologies allows for quantitative measurement of spatially localized gene abundance making it possible to bridge complex scenario of flower organogenesis with genome-wide molecular phenotypes. Here, we apply the 10× Visium technology in the study of the formation of floral organs through development in an orchid plant, Phalaenopsis Big Chili. Cell-types of early floral development including inflorescence meristems, primordia of floral organs and identity determined tissues, are recognized based on spatial expression distribution of thousands of genes in high resolution. In addition, meristematic cells on the basal position of floral organs are found to continuously function in multiple developmental stages after organ initiation. Particularly, the development of anther, which primordium starts from a single spot to multiple differentiated cell-types in later stages including pollinium and other vegetative tissues, is revealed by well-known MADS-box genes and many other downstream regulators. The spatial transcriptome analyses provide comprehensive information of gene activity for understanding the molecular architecture of flower organogenesis and for future genomic and genetic studies of specific cell-types.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Leng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yonglong Li
- Jiangxi Provincial Key Laboratory for Bamboo Germplasm Resources and Utilization, Forestry College, Jiangxi Agricultural University, Nanchang, China
| | - Tingting Ge
- Jiangxi Provincial Key Laboratory for Bamboo Germplasm Resources and Utilization, Forestry College, Jiangxi Agricultural University, Nanchang, China
| | - Jinglong Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yamao Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Chunce Guo
- Jiangxi Provincial Key Laboratory for Bamboo Germplasm Resources and Utilization, Forestry College, Jiangxi Agricultural University, Nanchang, China
| | - Ji Qi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
88
|
Wendt KD, Brown J, Lungova V, Mohad V, Kendziorski C, Thibeault SL. Transcriptome Dynamics in the Developing Larynx, Trachea, and Esophagus. Front Cell Dev Biol 2022; 10:942622. [PMID: 35938172 PMCID: PMC9353518 DOI: 10.3389/fcell.2022.942622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/03/2022] [Indexed: 11/18/2022] Open
Abstract
The larynx, trachea, and esophagus share origin and proximity during embryonic development. Clinical and experimental evidence support the existence of neurophysiological, structural, and functional interdependencies before birth. This investigation provides the first comprehensive transcriptional profile of all three organs during embryonic organogenesis, where differential gene expression gradually assembles the identity and complexity of these proximal organs from a shared origin in the anterior foregut. By applying bulk RNA sequencing and gene network analysis of differentially expressed genes (DEGs) within and across developing embryonic mouse larynx, esophagus, and trachea, we identified co-expressed modules of genes enriched for key biological processes. Organ-specific temporal patterns of gene activity corresponding to gene modules within and across shared tissues during embryonic development (E10.5-E18.5) are described, and the laryngeal transcriptome during vocal fold development and maturation from birth to adulthood is characterized in the context of laryngeal organogenesis. The findings of this study provide new insights into interrelated gene sets governing the organogenesis of this tripartite organ system within the aerodigestive tract. They are relevant to multiple families of disorders defined by cardiocraniofacial syndromes.
Collapse
Affiliation(s)
- Kristy D. Wendt
- Department of Surgery, Division of Otolaryngology, Head, and Neck Surgery, University of Wisconsin, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Jared Brown
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, United States
| | - Vlasta Lungova
- Department of Surgery, Division of Otolaryngology, Head, and Neck Surgery, University of Wisconsin, Madison, WI, United States
| | - Vidisha Mohad
- Department of Surgery, Division of Otolaryngology, Head, and Neck Surgery, University of Wisconsin, Madison, WI, United States
| | - Christina Kendziorski
- Department of Biostatistics and Medical Information, University of Wisconsin-Madison, Madison, WI, United States
| | - Susan L. Thibeault
- Department of Surgery, Division of Otolaryngology, Head, and Neck Surgery, University of Wisconsin, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
- *Correspondence: Susan L. Thibeault,
| |
Collapse
|
89
|
Bu L, Cripps RM. Promoter architecture of Drosophila genes regulated by Myocyte enhancer factor-2. PLoS One 2022; 17:e0271554. [PMID: 35862472 PMCID: PMC9302807 DOI: 10.1371/journal.pone.0271554] [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: 12/23/2021] [Accepted: 07/01/2022] [Indexed: 11/18/2022] Open
Abstract
To gain understanding into the mechanisms of transcriptional activation of muscle genes, we sought to determine if genes targeted by the myogenic transcription factor Myocyte enhancer factor-2 (MEF2) were enriched for specific core promoter elements. We identified 330 known MEF2 target promoters in Drosophila, and analyzed them for for the presence and location of 17 known consensus promoter sequences. As a control, we also searched all Drosophila RNA polymerase II-dependent promoters for the same sequences. We found that promoter motifs were readily detected in the MEF2 target dataset, and that many of them were slightly enriched in frequency compared to the control dataset. A prominent sequence over-represented in the MEF2 target genes was NDM2, that appeared in over 50% of MEF2 target genes and was 2.5-fold over-represented in MEF2 targets compared to background. To test the functional significance of NDM2, we identified two promoters containing a single copy of NDM2 plus an upstream MEF2 site, and tested the activity of these promoters in vivo. Both the sticks and stones and Kahuli fragments showed strong skeletal myoblast-specific expression of a lacZ reporter in embryos. However, the timing and level of reporter expression was unaffected when the NDM2 site in either element was mutated. These studies identify variations in promoter architecture for a set of regulated genes compared to all RNA polymerase II-dependent genes, and underline the potential redundancy in the activities of some core promoter elements.
Collapse
Affiliation(s)
- Lijing Bu
- Department of Biology and Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States of America
| | - Richard M. Cripps
- Department of Biology, San Diego State University, San Diego, CA, United States of America
| |
Collapse
|
90
|
Fung CW, Zhou S, Zhu H, Wei X, Wu Z, Wu AR. Cell fate determining molecular switches and signaling pathways in Pax7-expressing somitic mesoderm. Cell Discov 2022; 8:61. [PMID: 35764624 PMCID: PMC9240041 DOI: 10.1038/s41421-022-00407-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/28/2022] [Indexed: 11/09/2022] Open
Abstract
During development, different cell types originate from a common progenitor at well-defined time points. Previous lineage-tracing of Pax7+ progenitors from the somitic mesoderm has established its developmental trajectory towards the dermis, brown adipocytes, and skeletal muscle in the dorsal trunk; yet the molecular switches and mechanisms guiding the differentiation into different lineages remain unknown. We performed lineage-tracing of Pax7-expressing cells in mouse embryos at E9.5 and profiled the transcriptomes of Pax7-progenies on E12.5, E14.5, and E16.5 at single-cell level. Analysis of single-cell transcriptomic data at multiple time points showed temporal-specific differentiation events toward muscle, dermis, and brown adipocyte, identified marker genes for putative progenitors and revealed transcription factors that could drive lineage-specific differentiation. We then utilized a combination of surface markers identified in the single-cell data, Pdgfra, Thy1, and Cd36, to enrich brown adipocytes, dermal fibroblasts, and progenitors specific for these two cell types at E14.5 and E16.5. These enriched cell populations were then used for further culture and functional assays in vitro, in which Wnt5a and Rgcc are shown to be important factors that could alter lineage decisions during embryogenesis. Notably, we found a bipotent progenitor population at E14.5, having lineage potentials towards both dermal fibroblasts and brown adipocytes. They were termed eFAPs (embryonic fibro/adipogenic progenitors) as they functionally resemble adult fibro/adipogenic progenitors. Overall, this study provides further understanding of the Pax7 lineage during embryonic development using a combination of lineage tracing with temporally sampled single-cell transcriptomics.
Collapse
|
91
|
Abdallah HY, Hassan R, Fareed A, Abdelgawad M, Mostafa SA, Mohammed EAM. Identification of a circulating microRNAs biomarker panel for non-invasive diagnosis of coronary artery disease: case-control study. BMC Cardiovasc Disord 2022; 22:286. [PMID: 35751015 PMCID: PMC9233383 DOI: 10.1186/s12872-022-02711-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/09/2022] [Indexed: 12/07/2022] Open
Abstract
Background Circulating microRNAs (miRNAs) are considered a hot spot of research that can be employed for monitoring and/or diagnostic purposes in coronary artery disease (CAD). Since different disease features might be reflected on altered profiles or plasma miRNAs concentrations, a combination of miRNAs can provide more reliable non-invasive biomarkers for CAD. Subjects and methods We investigated a panel of 14-miRNAs selected using bioinformatics databases and current literature searching for miRNAs involved in CAD using quantitative real-time PCR technique in 73 CAD patients compared to 73 controls followed by function and pathway enrichment analysis for the 14-miRNAs. Results Our results revealed three out of the 14 circulating miRNAs understudy; miRNAs miR133a, miR155 and miR208a were downregulated. While 11 miRNAs were up-regulated in a descending order from highest fold change to lowest: miR-182, miR-145, miR-21, miR-126, miR-200b, miR-146A, miR-205, miR-135b, miR-196b, miR-140b and, miR-223. The ROC curve analysis indicated that miR-145, miR-182, miR-133a and, miR-205 were excellent biomarkers with the highest AUCs as biomarkers in CAD. All miRNAs under study except miR-208 revealed a statistically significant relation with dyslipidemia. MiR-126 and miR-155 showed significance with BMI grade, while only miR-133a showed significance with the obese patients in general. MiR-135b and miR-140b showed a significant correlation with the Wall Motion Severity Index. Pathway enrichment analysis for the miRNAS understudy revealed pathways relevant to the fatty acid biosynthesis, ECM-receptor interaction, proteoglycans in cancer, and adherens junction. Conclusion The results of this study identified a differentially expressed circulating miRNAs signature that can discriminate CAD patients from normal subjects. These results provide new insights into the significant role of miRNAs expression associated with CAD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02711-9.
Collapse
Affiliation(s)
- Hoda Y Abdallah
- Medical Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt. .,Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Ranya Hassan
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Ahmed Fareed
- Department of Cardiology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Mai Abdelgawad
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Sally Abdallah Mostafa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Abdel-Moemen Mohammed
- Medical Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.,Center of Excellence in Molecular & Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
92
|
Lu Z, Mao W, Yang H, Santiago-O'Farrill JM, Rask PJ, Mondal J, Chen H, Ivan C, Liu X, Liu CG, Xi Y, Masuda K, Carrami EM, Chen M, Tang Y, Pang L, Lakomy DS, Calin GA, Liang H, Ahmed AA, Vankayalapati H, Bast RC. SIK2 inhibition enhances PARP inhibitor activity synergistically in ovarian and triple-negative breast cancers. J Clin Invest 2022; 132:146471. [PMID: 35642638 PMCID: PMC9151707 DOI: 10.1172/jci146471] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/19/2022] [Indexed: 12/21/2022] Open
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARP inhibitors) have had an increasing role in the treatment of ovarian and breast cancers. PARP inhibitors are selectively active in cells with homologous recombination DNA repair deficiency caused by mutations in BRCA1/2 and other DNA repair pathway genes. Cancers with homologous recombination DNA repair proficiency respond poorly to PARP inhibitors. Cancers that initially respond to PARP inhibitors eventually develop drug resistance. We have identified salt-inducible kinase 2 (SIK2) inhibitors, ARN3236 and ARN3261, which decreased DNA double-strand break (DSB) repair functions and produced synthetic lethality with multiple PARP inhibitors in both homologous recombination DNA repair deficiency and proficiency cancer cells. SIK2 is required for centrosome splitting and PI3K activation and regulates cancer cell proliferation, metastasis, and sensitivity to chemotherapy. Here, we showed that SIK2 inhibitors sensitized ovarian and triple-negative breast cancer (TNBC) cells and xenografts to PARP inhibitors. SIK2 inhibitors decreased PARP enzyme activity and phosphorylation of class-IIa histone deacetylases (HDAC4/5/7). Furthermore, SIK2 inhibitors abolished class-IIa HDAC4/5/7-associated transcriptional activity of myocyte enhancer factor-2D (MEF2D), decreasing MEF2D binding to regulatory regions with high chromatin accessibility in FANCD2, EXO1, and XRCC4 genes, resulting in repression of their functions in the DNA DSB repair pathway. The combination of PARP inhibitors and SIK2 inhibitors provides a therapeutic strategy to enhance PARP inhibitor sensitivity for ovarian cancer and TNBC.
Collapse
Affiliation(s)
- Zhen Lu
- Department of Experimental Therapeutics
| | | | | | | | | | | | - Hu Chen
- Department of Bioinformatics & Computational Biology, and
| | - Cristina Ivan
- Department of Experimental Therapeutics.,Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Yuanxin Xi
- Department of Bioinformatics & Computational Biology, and
| | - Kenta Masuda
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA.,Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, United Kingdom
| | - Eli M Carrami
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA.,Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, United Kingdom
| | - Meng Chen
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yitao Tang
- Department of Bioinformatics & Computational Biology, and.,The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Lan Pang
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - George A Calin
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Han Liang
- Department of Bioinformatics & Computational Biology, and.,Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ahmed A Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, United Kingdom.,Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom
| | | | | |
Collapse
|
93
|
Zhou Z, Li K, Liu J, Zhang H, Fan Y, Chen Y, Han H, Yang J, Liu Y. Expression Profile Analysis to Identify Circular RNA Expression Signatures in Muscle Development of Wu'an Goat Longissimus Dorsi Tissues. Front Vet Sci 2022; 9:833946. [PMID: 35518637 PMCID: PMC9062782 DOI: 10.3389/fvets.2022.833946] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
The growth and development of skeletal muscle is a physiological process regulated by a variety of genes and signaling pathways. As a posttranscriptional regulatory factor, circRNA plays a certain regulatory role in the development of animal skeletal muscle in the form of a miRNA sponge. However, the role of circRNAs in muscle development and growth in goats is still unclear. In our study, apparent differences in muscle fibers in Wu'an goats of different ages was firstly detected by hematoxylin-eosin (HE) staining, the circRNA expression profiles of longissimus dorsi muscles from 1-month-old (mon1) and 9-month-old (mon9) goats were screened by RNA-seq and verified by RT-qPCR. The host genes of differentially expressed (DE) circRNAs were predicted, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses (KEGG) of host genes with DE circRNAs were performed to explore the functions of circRNAs. The circRNA-miRNA-mRNA networks were then constructed using Cytoscape software. Ten significantly differentially expressed circRNAs were also verified in the mon1 and mon9 groups by RT-qPCR. Luciferase Reporter Assay was used to verify the binding site between circRNA and its targeted miRNA. The results showed that a total of 686 DE circRNAs were identified between the mon9 and mon1 groups, of which 357 were upregulated and 329 were downregulated. Subsequently, the 467 host genes of DE circRNAs were predicted using Find_circ and CIRI software. The circRNA-miRNA-mRNA network contained 201 circRNAs, 85 miRNAs, and 581 mRNAs; the host mRNAs were associated with "muscle fiber development" and "AMPK signaling pathway" and were enriched in the FoxO signaling pathway. Competing endogenous RNA (ceRNA) network analysis showed that novel_circ_0005314, novel_circ_0005319, novel_circ_0009256, novel_circ_0009845, novel_circ_0005934 and novel_circ_0000134 may play important roles in skeletal muscle growth and development between the mon9 and mon1 groups. Luciferase Reporter Assay confirmed the combination between novel_circ_0005319 and chi-miR-199a-5p, novel_circ_0005934 and chi-miR-450-3p and novel_circ_0000134 and chi-miR-655. Our results provide specific information related to goat muscle development and a reference for the goat circRNA profile.
Collapse
Affiliation(s)
- Zuyang Zhou
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Kunyu Li
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Jiannan Liu
- School of Landscape and Ecological Engineering, Hebei University of Engineering, Handan, China
| | - Hui Zhang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yekai Fan
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yulin Chen
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Haiyin Han
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Junqi Yang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yufang Liu
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
94
|
Yoshida M, Yokoi N, Takahashi H, Hatano N, Hayami T, Ogawa W, Seino S. O-GlcNAcylation of myocyte-specific enhancer factor 2D negatively regulates insulin secretion from pancreatic β-cells. Biochem Biophys Res Commun 2022; 605:90-96. [DOI: 10.1016/j.bbrc.2022.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/07/2022] [Indexed: 11/02/2022]
|
95
|
Mubeen H, Farooq M, Rehman AU, Zubair M, Haque A. Gene expression and transcriptional regulation driven by transcription factors involved in congenital heart defects. Ir J Med Sci 2022; 192:595-604. [PMID: 35441975 DOI: 10.1007/s11845-022-02974-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/24/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Congenital heart disease (CHD) is one of the most important birth defects caused by more than one mutated gene. Mutations in the genes could cause different types of congenital heart defects including atrial septal defect (ASD), tetralogy of Fallot (TOF), and ventricular septal defect (VSD). OBJECTIVES Cardiac transcription factors are key players for heart development and are actively involved in controlling stress regulation of the heart. Transcription factors are sequence-specific DNA binding proteins that control the process of transcription and work in a synergistic manner. We aim to characterize core cardiac transcription factors including NKX2-5, TBX, SRF, GATA4, and MEF2, which encode homeobox and MADS domain and play a crucial role in heart development. METHODS In this study, we have explored the important transcription factors involved in cardiac development and genes controlling the expression and regulation process by using the bioinformatics approach. RESULTS We have predicted the orthologs and homologs based on their evolutionary history, conserved protein domains, functional sites, and 3D structures for better understanding and presentation of factors responsible for causing CHD. Results showed the importance of these transcription factors for normal heart functioning and development. CONCLUSION Understanding the molecular pathways and genetic basis of CHD will help to open a new door for the treatment of patients with cardiac defects.
Collapse
Affiliation(s)
- Hira Mubeen
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Farooq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan. .,Department of Bioinformatics, Institute of Biochemistry, Biotechnology & Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | | | - Muhammad Zubair
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Asma Haque
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
96
|
Fu X, Li S, Jia M, Xu B, Yang L, Ma R, Cheng H, Yang W, Hu P. Myogenesis controlled by a long non-coding RNA 1700113A16RIK and post-transcriptional regulation. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:13. [PMID: 35366685 PMCID: PMC8977255 DOI: 10.1186/s13619-022-00114-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/22/2022] [Indexed: 01/05/2023]
Abstract
Long non-coding (lnc) RNA plays important roles in many cellular processes. The function of the vast majority of lncRNAs remains unknown. Here we identified that lncRNA-1700113A16RIK existed in skeletal muscle stem cells (MuSCs) and was significantly elevated during MuSC differentiation. Knockdown of 1700113A16RIK inhibits the differentiation of muscle stem cells. In contrast, overexpression of 1700113A16RIK promotes the differentiation of muscle stem cells. Further study shows the muscle specific transcription factor Myogenin (MyoG) positively regulates the expression of 1700113A16RIK by binding to the promoter region of 1700113A16RIK. Mechanistically, 1700113A16RIK may regulate the expression of myogenic genes by directly binding to 3'UTR of an important myogenic transcription factor MEF2D, which in turn promotes the translation of MEF2D. Taken together, our results defined 1700113A16RIK as a positive regulator of MuSC differentiation and elucidated a mechanism as to how 1700113A16RIK regulated MuSC differentiation.
Collapse
Affiliation(s)
- Xin Fu
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Sheng Li
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Minzhi Jia
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Bo Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lele Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ruimiao Ma
- Guangzhou Laboratory, Guangzhou, 510700, Guangdong, China
| | - Hong Cheng
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenjun Yang
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China.
| | - Ping Hu
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China. .,Guangzhou Laboratory, Guangzhou, 510700, Guangdong, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
97
|
Zhao X, Di Q, Liu H, Quan J, Ling J, Zhao Z, Xiao Y, Wu H, Wu Z, Song W, An H, Chen W. MEF2C promotes M1 macrophage polarization and Th1 responses. Cell Mol Immunol 2022; 19:540-553. [PMID: 35194174 PMCID: PMC8975968 DOI: 10.1038/s41423-022-00841-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
The polarization of macrophages to the M1 or M2 phenotype has a pivotal role in inflammation and host defense; however, the underlying molecular mechanism remains unclear. Here, we show that myocyte enhancer factor 2 C (MEF2C) is essential for regulating M1 macrophage polarization in response to infection and inflammation. Global gene expression analysis demonstrated that MEF2C deficiency in macrophages downregulated the expression of M1 phenotypic markers and upregulated the expression of M2 phenotypic markers. MEF2C significantly promoted the expression of interleukin-12 p35 subunit (Il12a) and interleukin-12 p40 subunit (Il12b). Myeloid-specific Mef2c-knockout mice showed reduced IL-12 production and impaired Th1 responses, which led to susceptibility to Listeria monocytogenes infection and protected against DSS-induced IBD in vivo. Mechanistically, we showed that MEF2C directly activated the transcription of Il12a and Il12b. These findings reveal a new function of MEF2C in macrophage polarization and Th1 responses and identify MEF2C as a potential target for therapeutic intervention in inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Xibao Zhao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Qianqian Di
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Han Liu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jiazheng Quan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Jing Ling
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zizhao Zhao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yue Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Han Wu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Zherui Wu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Wengang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong, 250014, China
| | - Huazhang An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong, 250014, China.
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China.
| |
Collapse
|
98
|
Loumaye A, Lause P, Zhong X, Zimmers TA, Bindels LB, Thissen JP. Activin A Causes Muscle Atrophy through MEF2C-Dependent Impaired Myogenesis. Cells 2022; 11:cells11071119. [PMID: 35406681 PMCID: PMC8997966 DOI: 10.3390/cells11071119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/05/2022] Open
Abstract
Activin A (ActA) is considered to play a major role in cancer-induced cachexia (CC). Indeed, circulating ActA levels are elevated and predict survival in patients with CC. However, the mechanisms by which ActA mediates CC development and in particular skeletal muscle (SM) atrophy in humans are not yet fully understood. In this work, we aimed to investigate the effects of ActA on human SM and in mouse models of CC. We used a model of human muscle cells in culture to explore how ActA acts towards human SM. In this model, recombinant ActA induced myotube atrophy associated with the decline of MyHC-β/slow, the main myosin isoform in human muscle cells studied. Moreover, ActA inhibited the expression and activity of MEF2C, the transcription factor regulating MYH7, the gene which codes for MyHC-β/slow. This decrease in MEF2C was involved in the decline of MyHC-β/slow expression, since inhibition of MEF2C by a siRNA leads to the decrease in MyHC-β/slow expression. The relevance of this ActA/MEF2C pathway in vivo was supported by the parallel decline of MEF2C expression and SM mass, which are both blunted by ActA inhibition, in animal models of CC. In this work, we showed that ActA is a potent negative regulator of SM mass by inhibiting MyHC-β/slow synthesis through downregulation of MEF2C. This observation highlights a novel interaction between ActA signaling and MEF2C transcriptional activity which contributes to SM atrophy in CC models.
Collapse
Affiliation(s)
- Audrey Loumaye
- Pole of Endocrinology, Diabetology and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (P.L.); (J.-P.T.)
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Correspondence: ; Tel.: +32-2-764-6001
| | - Pascale Lause
- Pole of Endocrinology, Diabetology and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (P.L.); (J.-P.T.)
| | - Xiaoling Zhong
- Department of Surgery, Indiana University School of Medicine, Indiana University Simon and Bren Comprehensive Cancer Center, Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA; (X.Z.); (T.A.Z.)
- Research Service, Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indiana University Simon and Bren Comprehensive Cancer Center, Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA; (X.Z.); (T.A.Z.)
- Research Service, Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Jean-Paul Thissen
- Pole of Endocrinology, Diabetology and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (P.L.); (J.-P.T.)
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
99
|
Wagner M, Bračun S, Duenser A, Sturmbauer C, Gessl W, Ahi EP. Expression variations in ectodysplasin-A gene (eda) may contribute to morphological divergence of scales in haplochromine cichlids. BMC Ecol Evol 2022; 22:28. [PMID: 35272610 PMCID: PMC8908630 DOI: 10.1186/s12862-022-01984-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Elasmoid scales are one of the most common dermal appendages and can be found in almost all species of bony fish differing greatly in their shape. Whilst the genetic underpinnings behind elasmoid scale development have been investigated, not much is known about the mechanisms involved in moulding of scales. To investigate the links between gene expression differences and morphological divergence, we inferred shape variation of scales from two different areas of the body (anterior and posterior) stemming from ten haplochromine cichlid species from different origins (Lake Tanganyika, Lake Malawi, Lake Victoria and riverine). Additionally, we investigated transcriptional differences of a set of genes known to be involved in scale development and morphogenesis in fish. RESULTS We found that scales from the anterior and posterior part of the body strongly differ in their overall shape, and a separate look on scales from each body part revealed similar trajectories of shape differences considering the lake origin of single investigated species. Above all, nine as well as 11 out of 16 target genes showed expression differences between the lakes for the anterior and posterior dataset, respectively. Whereas in posterior scales four genes (dlx5, eda, rankl and shh) revealed significant correlations between expression and morphological differentiation, in anterior scales only one gene (eda) showed such a correlation. Furthermore, eda displayed the most significant expression difference between species of Lake Tanganyika and species of the other two younger lakes. Finally, we found genetic differences in downstream regions of eda gene (e.g., in the eda-tnfsf13b inter-genic region) that are associated with observed expression differences. This is reminiscent of a genetic difference in the eda-tnfsf13b inter-genic region which leads to gain or loss of armour plates in stickleback. CONCLUSION These findings provide evidence for cross-species transcriptional differences of an important morphogenetic factor, eda, which is involved in formation of ectodermal appendages. These expression differences appeared to be associated with morphological differences observed in the scales of haplochromine cichlids indicating potential role of eda mediated signal in divergent scale morphogenesis in fish.
Collapse
Affiliation(s)
- Maximilian Wagner
- Institute of Biology, University of Graz, Universitätsplatz 2, 8010, Graz, Austria.,Department of Biology, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Sandra Bračun
- Institute of Biology, University of Graz, Universitätsplatz 2, 8010, Graz, Austria
| | - Anna Duenser
- Institute of Biology, University of Graz, Universitätsplatz 2, 8010, Graz, Austria
| | - Christian Sturmbauer
- Institute of Biology, University of Graz, Universitätsplatz 2, 8010, Graz, Austria.
| | - Wolfgang Gessl
- Institute of Biology, University of Graz, Universitätsplatz 2, 8010, Graz, Austria
| | - Ehsan Pashay Ahi
- Institute of Biology, University of Graz, Universitätsplatz 2, 8010, Graz, Austria. .,Organismal and Evolutionary Biology Research Programme, University of Helsinki, Viikinkaari 9, 00014, Helsinki, Finland.
| |
Collapse
|
100
|
Copola AGL, Dos Santos ÍGD, Coutinho LL, Del-Bem LEV, de Almeida Campos-Junior PH, da Conceição IMCA, Nogueira JM, do Carmo Costa A, Silva GAB, Jorge EC. Transcriptomic characterization of the molecular mechanisms induced by RGMa during skeletal muscle nuclei accretion and hypertrophy. BMC Genomics 2022; 23:188. [PMID: 35255809 PMCID: PMC8902710 DOI: 10.1186/s12864-022-08396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 02/15/2022] [Indexed: 12/02/2022] Open
Abstract
Background The repulsive guidance molecule a (RGMa) is a GPI-anchor axon guidance molecule first found to play important roles during neuronal development. RGMa expression patterns and signaling pathways via Neogenin and/or as BMP coreceptors indicated that this axon guidance molecule could also be working in other processes and diseases, including during myogenesis. Previous works from our research group have consistently shown that RGMa is expressed in skeletal muscle cells and that its overexpression induces both nuclei accretion and hypertrophy in muscle cell lineages. However, the cellular components and molecular mechanisms induced by RGMa during the differentiation of skeletal muscle cells are poorly understood. In this work, the global transcription expression profile of RGMa-treated C2C12 myoblasts during the differentiation stage, obtained by RNA-seq, were reported. Results RGMa treatment could modulate the expression pattern of 2,195 transcripts in C2C12 skeletal muscle, with 943 upregulated and 1,252 downregulated. Among them, RGMa interfered with the expression of several RNA types, including categories related to the regulation of RNA splicing and degradation. The data also suggested that nuclei accretion induced by RGMa could be due to their capacity to induce the expression of transcripts related to ‘adherens junsctions’ and ‘extracellular-cell adhesion’, while RGMa effects on muscle hypertrophy might be due to (i) the activation of the mTOR-Akt independent axis and (ii) the regulation of the expression of transcripts related to atrophy. Finally, RGMa induced the expression of transcripts that encode skeletal muscle structural proteins, especially from sarcolemma and also those associated with striated muscle cell differentiation. Conclusions These results provide comprehensive knowledge of skeletal muscle transcript changes and pathways in response to RGMa. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08396-w.
Collapse
Affiliation(s)
- Aline Gonçalves Lio Copola
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Íria Gabriela Dias Dos Santos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Luiz Lehmann Coutinho
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, Brasil
| | - Luiz Eduardo Vieira Del-Bem
- Departamento de Botânica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | | | | | - Júlia Meireles Nogueira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Alinne do Carmo Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Gerluza Aparecida Borges Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Erika Cristina Jorge
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil.
| |
Collapse
|