1
|
Chambers TL, Dimet‐Wiley A, Keeble AR, Haghani A, Lo W, Kang G, Brooke R, Horvath S, Fry CS, Watowich SJ, Wen Y, Murach KA. Methylome-proteome integration after late-life voluntary exercise training reveals regulation and target information for improved skeletal muscle health. J Physiol 2025; 603:211-237. [PMID: 39058663 PMCID: PMC11702923 DOI: 10.1113/jp286681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Exercise is a potent stimulus for combatting skeletal muscle ageing. To study the effects of exercise on muscle in a preclinical setting, we developed a combined endurance-resistance training stimulus for mice called progressive weighted wheel running (PoWeR). PoWeR improves molecular, biochemical, cellular and functional characteristics of skeletal muscle and promotes aspects of partial epigenetic reprogramming when performed late in life (22-24 months of age). In this investigation, we leveraged pan-mammalian DNA methylome arrays and tandem mass-spectrometry proteomics in skeletal muscle to provide detailed information on late-life PoWeR adaptations in female mice relative to age-matched sedentary controls (n = 7-10 per group). Differential CpG methylation at conserved promoter sites was related to transcriptional regulation genes as well as Nr4a3, Hes1 and Hox genes after PoWeR. Using a holistic method of -omics integration called binding and expression target analysis (BETA), methylome changes were associated with upregulated proteins related to global and mitochondrial translation after PoWeR (P = 0.03). Specifically, BETA implicated methylation control of ribosomal, mitoribosomal, and mitochondrial complex I protein abundance after training. DNA methylation may also influence LACTB, MIB1 and UBR4 protein induction with exercise - all are mechanistically linked to muscle health. Computational cistrome analysis predicted several transcription factors including MYC as regulators of the exercise trained methylome-proteome landscape, corroborating prior late-life PoWeR transcriptome data. Correlating the proteome to muscle mass and fatigue resistance revealed positive relationships with VPS13A and NPL levels, respectively. Our findings expose differential epigenetic and proteomic adaptations associated with translational regulation after PoWeR that could influence skeletal muscle mass and function in aged mice. KEY POINTS: Late-life combined endurance-resistance exercise training from 22-24 months of age in mice is shown to improve molecular, biochemical, cellular and in vivo functional characteristics of skeletal muscle and promote aspects of partial epigenetic reprogramming and epigenetic age mitigation. Integration of DNA CpG 36k methylation arrays using conserved sites (which also contain methylation ageing clock sites) with exploratory proteomics in skeletal muscle extends our prior work and reveals coordinated and widespread regulation of ribosomal, translation initiation, mitochondrial ribosomal (mitoribosomal) and complex I proteins after combined voluntary exercise training in a sizeable cohort of female mice (n = 7-10 per group and analysis). Multi-omics integration predicted epigenetic regulation of serine β-lactamase-like protein (LACTB - linked to tumour resistance in muscle), mind bomb 1 (MIB1 - linked to satellite cell and type 2 fibre maintenance) and ubiquitin protein ligase E3 component N-recognin 4 (UBR4 - linked to muscle protein quality control) after training. Computational cistrome analysis identified MYC as a regulator of the late-life training proteome, in agreement with prior transcriptional analyses. Vacuolar protein sorting 13 homolog A (VPS13A) was positively correlated to muscle mass, and the glycoprotein/glycolipid associated sialylation enzyme N-acetylneuraminate pyruvate lyase (NPL) was associated to in vivo muscle fatigue resistance.
Collapse
Affiliation(s)
- Toby L. Chambers
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and RecreationUniversity of ArkansasFayettevilleARUSA
| | | | - Alexander R. Keeble
- University of Kentucky Center for Muscle BiologyLexingtonKYUSA
- Department of Athletic Training and Clinical NutritionUniversity of KentuckyLexingtonKYUSA
| | - Amin Haghani
- Department of Human GeneticsUniversity of California Los AngelesLos AngelesCAUSA
- Altos LabsSan DiegoCAUSA
| | - Wen‐Juo Lo
- Department of Educational Statistics and Research MethodsUniversity of ArkansasFayettevilleARUSA
| | - Gyumin Kang
- University of Kentucky Center for Muscle BiologyLexingtonKYUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKYUSA
- Division of Biomedical Informatics, Department of Internal MedicineUniversity of KentuckyLexingtonKYUSA
| | - Robert Brooke
- Epigenetic Clock Development FoundationLos AngelesCAUSA
| | - Steve Horvath
- Department of Human GeneticsUniversity of California Los AngelesLos AngelesCAUSA
- Altos LabsSan DiegoCAUSA
- Epigenetic Clock Development FoundationLos AngelesCAUSA
| | - Christopher S. Fry
- University of Kentucky Center for Muscle BiologyLexingtonKYUSA
- Department of Athletic Training and Clinical NutritionUniversity of KentuckyLexingtonKYUSA
| | - Stanley J. Watowich
- Ridgeline TherapeuticsHoustonTXUSA
- Department of Biochemistry and Molecular BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Yuan Wen
- University of Kentucky Center for Muscle BiologyLexingtonKYUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKYUSA
- Division of Biomedical Informatics, Department of Internal MedicineUniversity of KentuckyLexingtonKYUSA
| | - Kevin A. Murach
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and RecreationUniversity of ArkansasFayettevilleARUSA
| |
Collapse
|
2
|
Edman S, Jones Iii RG, Jannig PR, Fernandez-Gonzalo R, Norrbom J, Thomas NT, Khadgi S, Koopmans PJ, Morena F, Chambers TL, Peterson CS, Scott LN, Greene NP, Figueiredo VC, Fry CS, Zhengye L, Lanner JT, Wen Y, Alkner B, Murach KA, von Walden F. The 24-hour molecular landscape after exercise in humans reveals MYC is sufficient for muscle growth. EMBO Rep 2024; 25:5810-5837. [PMID: 39482487 PMCID: PMC11624283 DOI: 10.1038/s44319-024-00299-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
A detailed understanding of molecular responses to a hypertrophic stimulus in skeletal muscle leads to therapeutic advances aimed at promoting muscle mass. To decode the molecular factors regulating skeletal muscle mass, we utilized a 24-h time course of human muscle biopsies after a bout of resistance exercise. Our findings indicate: (1) the DNA methylome response at 30 min corresponds to upregulated genes at 3 h, (2) a burst of translation- and transcription-initiation factor-coding transcripts occurs between 3 and 8 h, (3) changes to global protein-coding gene expression peaks at 8 h, (4) ribosome-related genes dominate the mRNA landscape between 8 and 24 h, (5) methylation-regulated MYC is a highly influential transcription factor throughout recovery. To test whether MYC is sufficient for hypertrophy, we periodically pulse MYC in skeletal muscle over 4 weeks. Transient MYC increases muscle mass and fiber size in the soleus of adult mice. We present a temporally resolved resource for understanding molecular adaptations to resistance exercise in muscle ( http://data.myoanalytics.com ) and suggest that controlled MYC doses influence the exercise-related hypertrophic transcriptional landscape.
Collapse
Affiliation(s)
- Sebastian Edman
- Division of Pediatric Neurology, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Ronald G Jones Iii
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Paulo R Jannig
- Division of Pediatric Neurology, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Rodrigo Fernandez-Gonzalo
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Unit of Clinical Physiology, Karolinska University Hospital, Huddinge, Sweden
| | - Jessica Norrbom
- Molecular Exercise Physiology Group, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Sabin Khadgi
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Pieter J Koopmans
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, AR, USA
| | - Francielly Morena
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Toby L Chambers
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Calvin S Peterson
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Logan N Scott
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Nicholas P Greene
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Vandre C Figueiredo
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Biological Sciences, Oakland University, Rochester Hills, MI, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Liu Zhengye
- Molecular Muscle Physiology & Pathophysiology Group, Department of Physiology & Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Johanna T Lanner
- Molecular Muscle Physiology & Pathophysiology Group, Department of Physiology & Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Yuan Wen
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Björn Alkner
- Department of Orthopaedic Surgery, Region Jönköping County, Eksjö, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Kevin A Murach
- Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA.
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, AR, USA.
| | - Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
3
|
Corsaro L, Sacco D, Corbetta C, Gentilini D, Faversani A, Ferrara F, Costantino L. A new approach to study stochastic epigenetic mutations in sperm methylome of Vietnam war veterans directly exposed to Agent Orange. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae020. [PMID: 39664489 PMCID: PMC11631699 DOI: 10.1093/eep/dvae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Among the various environmental pollutants, dioxin, a highly toxic and widely used compound, is associated with numerous adverse health effects, including a potentially toxic multigenerational effect. Understanding the mechanisms by which dioxin exposure can affect sperm epigenetics is critical to comprehending the potential consequences for offspring health and development. This study investigates the possible association between weighted epimutations, hypothesized as markers of epigenetic drift, and dioxin exposure in sperm tissues. We used a public online methylation dataset consisting of 37 participants: 26 Vietnam veterans exposed to Agent Orange, an herbicide contaminated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and 11 individuals not directly exposed to TCDD but whose serum dioxin levels are equivalent to the background. In our study, conducted at the gene level, 437 epimutated genes were identified as significantly associated with each single-digit increase in serum dioxin levels. We found no significant association between the rise in total epimutation load and serum dioxin levels. The pathway analysis performed on the genes reveals biological processes mainly related to changes in embryonic morphology, development, and reproduction. Results from our current study suggest the importance of further investigations on the consequences of dioxin exposure in humans with specific reference to germinal tissue and related heredity.
Collapse
Affiliation(s)
- Luigi Corsaro
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
- Department of Brain and Behavioural Sciences, Università di Pavia, Pavia, PV 27100, Italy
| | - Davide Sacco
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
- Department of Brain and Behavioural Sciences, Università di Pavia, Pavia, PV 27100, Italy
| | - Carlo Corbetta
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| | - Davide Gentilini
- Department of Brain and Behavioural Sciences, Università di Pavia, Pavia, PV 27100, Italy
| | - Alice Faversani
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| | - Lucy Costantino
- Medical Genetics Laboratory, Centro Diagnostico Italiano, Milan, MI 20147, Italy
| |
Collapse
|
4
|
Okino R, Goda Y, Ono Y. The Hox-based positional memory in muscle stem cells. J Biochem 2024; 176:277-283. [PMID: 39194026 DOI: 10.1093/jb/mvae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
The skeletal muscle is a contractile tissue distributed throughout the body with various anatomical sizes, shapes and functions. In pathological conditions, such as muscular dystrophy, age-related sarcopenia and cancer cachexia, skeletal muscles are not uniformly affected throughout the body. This region-specific vulnerability cannot be fully explained by known physiological classifications, including muscle fiber types. Accumulating evidence indicates that the expression patterns of topographic homeobox (Hox) genes provide a molecular signature of positional memory, reflecting the anatomical locations and embryonic history of muscles and their associated muscle stem cells in adult mice and humans. Hox-based positional memory is not merely a remnant of embryonic development but is expected to be an intrinsic determinant controlling muscle function because recent studies have shown that aberrant Hox genes affect muscle stem cells. In this review, we discuss the concept of Hox-based positional memory, which may offer a new perspective on the region-specific pathophysiology of muscle disorders.
Collapse
Affiliation(s)
- Ryosuke Okino
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yuki Goda
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yusuke Ono
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811, Japan
| |
Collapse
|
5
|
Chen S, Jiang X, Xia L, Chen Z, Zhou K, Yan J, Li P. The identification, adaptive evolutionary analyses and mRNA expression levels of homeobox (hox) genes in the Chinese mitten crab Eriocheir sinensis. BMC Genomics 2023; 24:436. [PMID: 37537567 PMCID: PMC10401747 DOI: 10.1186/s12864-023-09489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/28/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Arthropods are the largest group in the animal kingdom and are morphologically characterized by heterorhythmic segments. Brachyuran decapod crustaceans undergo brachyurization metamorphosis in the early developmental process, characterized by a reduced abdomen that is folded beneath the cephalothorax and inserted between the pereiopods or in a special cavity. As the main cause of major alterations in the evolution of animal body plans, Hox genes encode transcription factors and are involved in bilaterian anterior-posterior axis patterning. RESULTS We found eight Hox genes (labial, proboscipedia, Deformed, zerknüllt, Sex combs reduced, Antennapedia, Ultrabithorax, fushi tarazu, abdominal-A and Abdominal-B) in Eriocheir sinensis. The phylogenetic topology of 13 arthropod Hox genes was closely related to traditional taxonomic groupings. Genome collinearity analysis was performed using genomic data and chromosomal location data of E. sinensis and Portunus trituratus. We found that their chromosomes were highly collinear, and there was a corresponding collinear relationship between the three Hox genes (lab, ftz and Abd-B). The mRNA expression levels of Scr and Antp fluctuated significantly in different developmental stages of E. sinensis, especially in the brachyurization stages. Evolutionary analysis indicated the presence of positively selected sites in Ubx. CONCLUSIONS In this study, we used genome-wide analysis to identify and analyze all members of the Hox genes in E. sinensis. Our data will contribute to a better understanding of Hox genes in E. sinensis and provide useful molecular evolutionary information for further investigation on their roles in the brachyurization of crabs.
Collapse
Affiliation(s)
- Shasha Chen
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Xianfeng Jiang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Longjie Xia
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Zhiyi Chen
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Kaiya Zhou
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Jie Yan
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China.
| | - Peng Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
6
|
Castilla-Vallmanya L, Centeno-Pla M, Serrano M, Franco-Valls H, Martínez-Cabrera R, Prat-Planas A, Rojano E, Ranea JAG, Seoane P, Oliva C, Paredes-Fuentes AJ, Marfany G, Artuch R, Grinberg D, Rabionet R, Balcells S, Urreizti R. Advancing in Schaaf-Yang syndrome pathophysiology: from bedside to subcellular analyses of truncated MAGEL2. J Med Genet 2023; 60:406-415. [PMID: 36243518 PMCID: PMC10086475 DOI: 10.1136/jmg-2022-108690] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Schaaf-Yang syndrome (SYS) is caused by truncating mutations in MAGEL2, mapping to the Prader-Willi region (15q11-q13), with an observed phenotype partially overlapping that of Prader-Willi syndrome. MAGEL2 plays a role in retrograde transport and protein recycling regulation. Our aim is to contribute to the characterisation of SYS pathophysiology at clinical, genetic and molecular levels. METHODS We performed an extensive phenotypic and mutational revision of previously reported patients with SYS. We analysed the secretion levels of amyloid-β 1-40 peptide (Aβ1-40) and performed targeted metabolomic and transcriptomic profiles in fibroblasts of patients with SYS (n=7) compared with controls (n=11). We also transfected cell lines with vectors encoding wild-type (WT) or mutated MAGEL2 to assess stability and subcellular localisation of the truncated protein. RESULTS Functional studies show significantly decreased levels of secreted Aβ1-40 and intracellular glutamine in SYS fibroblasts compared with WT. We also identified 132 differentially expressed genes, including non-coding RNAs (ncRNAs) such as HOTAIR, and many of them related to developmental processes and mitotic mechanisms. The truncated form of MAGEL2 displayed a stability similar to the WT but it was significantly switched to the nucleus, compared with a mainly cytoplasmic distribution of the WT MAGEL2. Based on the updated knowledge, we offer guidelines for the clinical management of patients with SYS. CONCLUSION A truncated MAGEL2 protein is stable and localises mainly in the nucleus, where it might exert a pathogenic neomorphic effect. Aβ1-40 secretion levels and HOTAIR mRNA levels might be promising biomarkers for SYS. Our findings may improve SYS understanding and clinical management.
Collapse
Affiliation(s)
- Laura Castilla-Vallmanya
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
| | - Mónica Centeno-Pla
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Mercedes Serrano
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
- Neurology Department, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Héctor Franco-Valls
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Raúl Martínez-Cabrera
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Aina Prat-Planas
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
| | - Elena Rojano
- Department of Molecular Biology and Biochemistry; Institute of Biomedical Research in Málaga (IBIMA), University of Málaga, Málaga, Spain
| | - Juan A G Ranea
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
- Department of Molecular Biology and Biochemistry; Institute of Biomedical Research in Málaga (IBIMA), University of Málaga, Málaga, Spain
| | - Pedro Seoane
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
- Department of Molecular Biology and Biochemistry; Institute of Biomedical Research in Málaga (IBIMA), University of Málaga, Málaga, Spain
| | - Clara Oliva
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Abraham J Paredes-Fuentes
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Gemma Marfany
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
| | - Rafael Artuch
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
| | - Raquel Rabionet
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, IBUB, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
| | - Roser Urreizti
- Institut de Recerca Sant Joan de Déu, Espluques de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instiuto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
7
|
Epigenetic Differences Arise in Endothelial Cells Responding to Cobalt–Chromium. J Funct Biomater 2023; 14:jfb14030127. [PMID: 36976051 PMCID: PMC10052026 DOI: 10.3390/jfb14030127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Cobalt–chromium (Co-Cr)-based alloys are emerging with important characteristics for use in dentistry, but the knowledge of epigenetic mechanisms in endothelial cells has barely been achieved. In order to address this issue, we have prepared a previously Co-Cr-enriched medium to further treat endothelial cells (HUVEC) for up to 72 h. Our data show there is important involvement with epigenetic machinery. Based on the data, it is believed that methylation balance in response to Co-Cr is finely modulated by DNMTs (DNA methyltransferases) and TETs (Tet methylcytosine dioxygenases), especially DNMT3B and both TET1 and TET2. Additionally, histone compaction HDAC6 (histone deacetylase 6) seems to develop a significant effect in endothelial cells. The requirement of SIRT1 seems to have a crucial role in this scenario. SIRT1 is associated with a capacity to modulate the expression of HIF-1α in response to hypoxia microenvironments, thus presenting a protective effect. As mentioned previously, cobalt is able to prevent HIF1A degradation and maintain hypoxia-related signaling in eukaryotic cells. Together, our results show, for the first time, a descriptive study reporting the relevance of epigenetic machinery in endothelial cells responding to cobalt–chromium, and it opens new perspectives to better understand their repercussions as prerequisites for driving cell adhesion, cell cycle progression, and angiogenesis surrounding this Co-Cr-based implantable device.
Collapse
|
8
|
Integrated Analyses of DNA Methylation and Gene Expression of Rainbow Trout Muscle under Variable Ploidy and Muscle Atrophy Conditions. Genes (Basel) 2022; 13:genes13071151. [PMID: 35885934 PMCID: PMC9319582 DOI: 10.3390/genes13071151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/19/2022] [Accepted: 06/24/2022] [Indexed: 02/04/2023] Open
Abstract
Rainbow trout, Oncorhynchus mykiss, is an important cool, freshwater aquaculture species used as a model for biological research. However, its genome reference has not been annotated for epigenetic markers affecting various biological processes, including muscle growth/atrophy. Increased energetic demands during gonadogenesis/reproduction provoke muscle atrophy in rainbow trout. We described DNA methylation and its associated gene expression in atrophying muscle by comparing gravid, diploid females to sterile, triploid females. Methyl Mini-seq and RNA-Seq were simultaneously used to characterize genome-wide DNA methylation and its association with gene expression in rainbow trout muscle. Genome-wide enrichment in the number of CpGs, accompanied by depleted methylation levels, was noticed around the gene transcription start site (TSS). Hypermethylation of CpG sites within ±1 kb on both sides of TSS (promoter and gene body) was weakly/moderately associated with reduced gene expression. Conversely, hypermethylation of the CpG sites in downstream regions of the gene body +2 to +10 kb was weakly associated with increased gene expression. Unlike mammalian genomes, rainbow trout gene promotors are poor in CpG islands, at <1% compared to 60%. No signs of genome-wide, differentially methylated (DM) CpGs were observed due to the polyploidy effect; only 1206 CpGs (0.03%) were differentially methylated, and these were primarily associated with muscle atrophy. Twenty-eight genes exhibited differential gene expression consistent with methylation levels of 31 DM CpGs. These 31 DM CpGs represent potential epigenetic markers of muscle atrophy in rainbow trout. The DM CpG-harboring genes are involved in apoptosis, epigenetic regulation, autophagy, collagen metabolism, cell membrane functions, and Homeobox proteins. Our study also identified genes explaining higher water content and modulated glycolysis previously shown as characteristic biochemical signs of rainbow trout muscle atrophy associated with sexual maturation. This study characterized DNA methylation in the rainbow trout genome and its correlation with gene expression. This work also identified novel epigenetic markers associated with muscle atrophy in fish/lower vertebrates.
Collapse
|
9
|
Disatham J, Brennan L, Jiao X, Ma Z, Hejtmancik JF, Kantorow M. Changes in DNA methylation hallmark alterations in chromatin accessibility and gene expression for eye lens differentiation. Epigenetics Chromatin 2022; 15:8. [PMID: 35246225 PMCID: PMC8897925 DOI: 10.1186/s13072-022-00440-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
Background Methylation at cytosines (mCG) is a well-known regulator of gene expression, but its requirements for cellular differentiation have yet to be fully elucidated. A well-studied cellular differentiation model system is the eye lens, consisting of a single anterior layer of epithelial cells that migrate laterally and differentiate into a core of fiber cells. Here, we explore the genome-wide relationships between mCG methylation, chromatin accessibility and gene expression during differentiation of eye lens epithelial cells into fiber cells. Results Whole genome bisulfite sequencing identified 7621 genomic loci exhibiting significant differences in mCG levels between lens epithelial and fiber cells. Changes in mCG levels were inversely correlated with the differentiation state-specific expression of 1285 genes preferentially expressed in either lens fiber or lens epithelial cells (Pearson correlation r = − 0.37, p < 1 × 10–42). mCG levels were inversely correlated with chromatin accessibility determined by assay for transposase-accessible sequencing (ATAC-seq) (Pearson correlation r = − 0.86, p < 1 × 10–300). Many of the genes exhibiting altered regions of DNA methylation, chromatin accessibility and gene expression levels in fiber cells relative to epithelial cells are associated with lens fiber cell structure, homeostasis and transparency. These include lens crystallins (CRYBA4, CRYBB1, CRYGN, CRYBB2), lens beaded filament proteins (BFSP1, BFSP2), transcription factors (HSF4, SOX2, HIF1A), and Notch signaling pathway members (NOTCH1, NOTCH2, HEY1, HES5). Analysis of regions exhibiting cell-type specific alterations in DNA methylation revealed an overrepresentation of consensus sequences of multiple transcription factors known to play key roles in lens cell differentiation including HIF1A, SOX2, and the MAF family of transcription factors. Conclusions Collectively, these results link DNA methylation with control of chromatin accessibility and gene expression changes required for eye lens differentiation. The results also point to a role for DNA methylation in the regulation of transcription factors previously identified to be important for lens cell differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-022-00440-z.
Collapse
Affiliation(s)
- Joshua Disatham
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Lisa Brennan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Xiaodong Jiao
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhiwei Ma
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Fielding Hejtmancik
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marc Kantorow
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
| |
Collapse
|
10
|
Murach KA, Dungan CM, von Walden F, Wen Y. Epigenetic evidence for distinct contributions of resident and acquired myonuclei during long-term exercise adaptation using timed in vivo myonuclear labeling. Am J Physiol Cell Physiol 2022; 322:C86-C93. [PMID: 34817266 PMCID: PMC8765804 DOI: 10.1152/ajpcell.00358.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Muscle fibers are syncytial postmitotic cells that can acquire exogenous nuclei from resident muscle stem cells, called satellite cells. Myonuclei are added to muscle fibers by satellite cells during conditions such as load-induced hypertrophy. It is difficult to dissect the molecular contributions of resident versus satellite cell-derived myonuclei during adaptation due to the complexity of labeling distinct nuclear populations in multinuclear cells without label transference between nuclei. To sidestep this barrier, we used a genetic mouse model where myonuclear DNA can be specifically and stably labeled via nonconstitutive H2B-GFP at any point in the lifespan. Resident myonuclei (Mn) were GFP-tagged in vivo before 8 wk of progressive weighted wheel running (PoWeR) in adult mice (>4-mo-old). Resident + satellite cell-derived myonuclei (Mn+SC Mn) were labeled at the end of PoWeR in a separate cohort. Following myonuclear isolation, promoter DNA methylation profiles acquired with low-input reduced representation bisulfite sequencing (RRBS) were compared to deduce epigenetic contributions of satellite cell-derived myonuclei during adaptation. Resident myonuclear DNA has hypomethylated promoters in genes related to protein turnover, whereas the addition of satellite cell-derived myonuclei shifts myonuclear methylation profiles to favor transcription factor regulation and cell-cell signaling. By comparing myonucleus-specific methylation profiling to previously published single-nucleus transcriptional analysis in the absence (Mn) versus the presence of satellite cells (Mn+SC Mn) with PoWeR, we provide evidence that satellite cell-derived myonuclei may preferentially supply specific ribosomal proteins to growing myofibers and retain an epigenetic "memory" of prior stem cell identity. These data offer insights on distinct epigenetic myonuclear characteristics and contributions during adult muscle growth.
Collapse
Affiliation(s)
- Kevin A. Murach
- 1Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas,2Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas,3The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Cory M. Dungan
- 3The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky,4Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Ferdinand von Walden
- 5Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
| | - Yuan Wen
- 3The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky,6Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky,7Myoanalytics, LLC, Lexington, Kentucky
| |
Collapse
|
11
|
Li Y, Wang J, Elzo MA, Fan H, Du K, Xia S, Shao J, Lai T, Hu S, Jia X, Lai S. Molecular Profiling of DNA Methylation and Alternative Splicing of Genes in Skeletal Muscle of Obese Rabbits. Curr Issues Mol Biol 2021; 43:1558-1575. [PMID: 34698087 PMCID: PMC8929151 DOI: 10.3390/cimb43030110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022] Open
Abstract
DNA methylation and the alternative splicing of precursor messenger RNAs (pre-mRNAs) are two important genetic modification mechanisms. However, both are currently uncharacterized in the muscle metabolism of rabbits. Thus, we constructed the Tianfu black rabbit obesity model (obese rabbits fed with a 10% high-fat diet and control rabbits from 35 days to 70 days) and collected the skeletal muscle samples from the two groups for Genome methylation sequencing and RNA sequencing. DNA methylation data showed that the promoter regions of 599 genes and gene body region of 2522 genes had significantly differential methylation rates between the two groups, of which 288 genes had differential methylation rates in promoter and gene body regions. Analysis of alternative splicing showed 555 genes involved in exon skipping (ES) patterns, and 15 genes existed in differential methylation regions. Network analysis showed that 20 hub genes were associated with ubiquitinated protein degradation, muscle development pathways, and skeletal muscle energy metabolism. Our findings suggest that the two types of genetic modification have potential regulatory effects on skeletal muscle development and provide a basis for further mechanistic studies in the rabbit.
Collapse
Affiliation(s)
- Yanhong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Jie Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Mauricio A. Elzo
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA;
| | - Huimei Fan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Kun Du
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Siqi Xia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Jiahao Shao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Tianfu Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Shenqiang Hu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
| | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (J.W.); (H.F.); (K.D.); (S.X.); (J.S.); (T.L.); (S.H.); (X.J.)
- Correspondence:
| |
Collapse
|
12
|
The molecular pathway triggered by zirconia in endothelial cells involves epigenetic control. Tissue Cell 2021; 73:101627. [PMID: 34425516 DOI: 10.1016/j.tice.2021.101627] [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: 09/03/2020] [Revised: 07/14/2021] [Accepted: 08/14/2021] [Indexed: 11/24/2022]
Abstract
The requirement to achieve natural looking restorations is one of the most challenging aspects in dentistry. Although zirconia has provided new opportunities for achieving superior aesthetics and physicochemical outcomes, very little has been achieved for its cellular and molecular performance, especially considering angiogenesis and osteogenesis. As angiogenesis is a secondary event and concomitant to osteogenesis, an indirect effect of dental implant on endothelial cells could be the release of active molecules such as those already reported affecting osteoblasts. To better address this issue, we challenged human endothelial cells (HUVECs) with zirconia-conditioned medium up to 72 h to allow analysis specific gene expression and protein pattern of mediators of epigenetic machinery in full. Our data shows involvement of zirconia in triggering intracellular signaling through MAPK-ERK activation, leading the signal to activate histone deacetylase HDAC6 likely with concomitant well-modulated DNA methylation profile by DNMTs and TETs. These signaling pathways seem to culminate in cytoskeleton rearrangement of endothelial cells, an important prerequisite to cell migration expected in angiogenesis. Collectively, this study demonstrates for the first time epigenetic-related molecular mechanism involved in endothelial cells responding to zirconia, revealing a repertoire of signaling molecules capable of executing the reprogramming process of gene expression, which are necessary to drive cell proliferation, migration, and consequently angiogenesis. This set of data can further studies using gene editing approaches to better elucidate functional roles.
Collapse
|
13
|
Lin X, Yan M, Yu W, Ma Y, Zhang L, Wei W, Chen J. Hoxa11 and Hoxa13 facilitate slow-twitch muscle formation in C2C12 cells and indirectly affect the lipid deposition of 3T3-L1 cells. Anim Sci J 2021; 92:e13544. [PMID: 33738916 DOI: 10.1111/asj.13544] [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: 08/15/2020] [Revised: 01/18/2021] [Accepted: 02/15/2021] [Indexed: 11/29/2022]
Abstract
Muscle-fiber type in livestock skeletal muscles influences meat quality, but the underlying mechanisms remain unclear. We previously showed that Homeobox A11 (Hoxa11) and Homeobox A13 (Hoxa13) are differentially expressed in fast- and slow-twitch muscles, but their effects on the formation of muscle-fiber types and intramuscular fat deposition have not been investigated. Here, our results revealed that overexpression of Hoxa11 and Hoxa13 delayed cell-cycle progression in C2C12 myoblasts, reduced their proliferation, and promoted their differentiation into slow-twitch muscle fibers. Knockdown experiments produced the opposite results. The conditioned media of differentiated C2C12 cells with Hoxa11/Hoxa13 overexpression or knockdown were harvested. Staining results showed that adipogenesis of preadipocytes was significantly promoted by Hoxa13 knockdown C2C12 cell culture medium. Changes in lipid accumulation were due to a reduction in lipid decomposition and an increase in triglyceride synthesis; genes related to fatty-acid synthesis were decreased. In conclusion, our study showed that Hoxa11 and Hoxa13 promote slow-twitch muscle formation and indirectly regulate preadipocyte adipogenesis, which may facilitate meat-quality improvement in the future.
Collapse
Affiliation(s)
- Xiangsheng Lin
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Mengting Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wensai Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuchen Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
14
|
Yu S, Wang G, Liao J, Chen X. A functional mutation in the AMPD1 promoter region affects promoter activity and breast meat freshness in chicken. Anim Genet 2020; 52:121-125. [PMID: 33226134 DOI: 10.1111/age.13025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/26/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022]
Abstract
Freshness is an important index to determine the quality deterioration (protein degradation and changes in appearance) of chilled chicken meat and is a primary consideration of consumers. Adenosine monophosphate deaminase 1 (AMPD1) catalyzes the deamination of adenosine monophosphate to inosine monophosphate in skeletal muscle and is the rate-limiting step in the purine nucleotide cycle. Inosine monophosphate is regarded as an important indicator of meat freshness in chicken. This study investigated the association of polymorphisms in the chicken AMPD1 promoter region with meat freshness during freezing storage. An SNP (c. -905G>A) was found to be associated with the freshness (K-value) of chicken breast meat. Chickens with the AA genotype had significantly lower K-values than those with GG and AG genotypes (P < 0.01). Individuals with the AA genotype also had higher breast meat AMPD1 mRNA levels than did those with the GG and AG genotypes (P < 0.01, P < 0.05). A luciferase assay revealed that genotype AA had greater transcriptional activity than genotype GG. Transcription factor binding site analysis identified distinct putative transcription factor binding sites in the two alleles of mutation site c. -905. In summary, we identified an SNP (c. -905G>A) in the promoter region of the AMPD1 gene that may modulate the binding affinity of different transcription factors to control AMPD1 expression and affect the freshness K-value of chicken meat.
Collapse
Affiliation(s)
- S Yu
- Engineering Research Center of Sichuan Province Higher School of Local Chicken Breeds Industrialization in Southern Sichuan, College of Life Science, Leshan Normal University, Leshan, 614000, China
| | - G Wang
- Engineering Research Center of Sichuan Province Higher School of Local Chicken Breeds Industrialization in Southern Sichuan, College of Life Science, Leshan Normal University, Leshan, 614000, China
| | - J Liao
- Engineering Research Center of Sichuan Province Higher School of Local Chicken Breeds Industrialization in Southern Sichuan, College of Life Science, Leshan Normal University, Leshan, 614000, China
| | - X Chen
- Leshan Academy of Agricultural Sciences, Leshan, 614000, China
| |
Collapse
|
15
|
Zheng M, Xiao S, Guo T, Rao L, Li L, Zhang Z, Huang L. DNA methylomic homogeneity and heterogeneity in muscles and testes throughout pig adulthood. Aging (Albany NY) 2020; 12:25412-25431. [PMID: 33231562 PMCID: PMC7803572 DOI: 10.18632/aging.104143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/09/2020] [Indexed: 01/24/2023]
Abstract
DNA methylome pattern is significantly different among tissues, ages, breeds, and genders. We assessed 20 methylome and transcriptome data in longissimus dorsi (LD) or testicles from Bamaxiang (BMX) and Large White pigs (LW) by deep sequencing technology. We identified ~55.7M CpGs and 5.30M, 0.20M, 1.20M, and 0.16M differential CpGs (P<0.01) between tissues, ages, breeds, and genders, respectively. Interestingly, 7.54% of differentially methylated regions (DMRs) are co-localized with promoters, which potentially regulate gene expression. RNA-seq analysis revealed that 23.42% CpGs are significantly correlated with gene expression (mean |r|=0.58, P<0.01), most of which are enriched in tissue-specific functions. Specially, we also found that the methylation levels in promoters of 655 genes were strongly associated with their expression levels (mean |r|=0.66, P<0.01). In addition, differentially methylated CpGs (DMCpGs) between breeds in HOXC gene cluster imply important regulatory roles in myocytes hypertrophy and intermuscular fat (IMF) deposition. Dramatically, higher similarity of methylation pattern was observed within pedigree than across pedigrees, which indicates the existence of heritable methylation regions. In summary, a part of CpGs in promoter can change its methylation pattern and play a marked regulatory function in different physiological or natural environments.
Collapse
Affiliation(s)
- Min Zheng
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| | - Shijun Xiao
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| | - Tianfu Guo
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| | - Lin Rao
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| | - Longyun Li
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| | - Zhiyan Zhang
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| | - Lusheng Huang
- State Key Laboratory for Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
16
|
Van der Mude A. Structure encoding in DNA. J Theor Biol 2020; 492:110205. [PMID: 32070719 DOI: 10.1016/j.jtbi.2020.110205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 12/29/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
It is proposed that transposons and related long non-coding RNA define the fine structure of body parts. Although morphogens have long been known to direct the formation of many gross structures in early embryonic development, they do not have the necessary precision to define a structure down to the individual cellular level. Using the distinction between procedural and declarative knowledge in information processing as an analogy, it is hypothesized that DNA encodes fine structure in a manner that is different from the genetic code for proteins. The hypothesis states that repeated or near-repeated sequences that are in transposons and non-coding RNA define body part structures. As the cells in a body part go through the epigenetic process of differentiation, the action of methylation serves to inactivate all but the relevant structure definitions and some associated cell type genes. The transposons left active will then physically modify the DNA sequence in the heterochromatin to establish the local context in the three-dimensional body part structure. This brings the encoded definition of the cell type to the histone. The histone code for that cell type starts the regulatory cascade that turns on the genes associated with that particular type of cell, transforming it from a multipotent cell to a fully differentiated cell. This mechanism creates structures in the musculoskeletal system, the organs of the body, the major parts of the brain, and other systems.
Collapse
|
17
|
Ehrlich KC, Lacey M, Ehrlich M. Epigenetics of Skeletal Muscle-Associated Genes in the ASB, LRRC, TMEM, and OSBPL Gene Families. EPIGENOMES 2020; 4:1. [PMID: 34968235 PMCID: PMC8594701 DOI: 10.3390/epigenomes4010001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/21/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
Much remains to be discovered about the intersection of tissue-specific transcription control and the epigenetics of skeletal muscle (SkM), a very complex and dynamic organ. From four gene families, Leucine-Rich Repeat Containing (LRRC), Oxysterol Binding Protein Like (OSBPL), Ankyrin Repeat and Socs Box (ASB), and Transmembrane Protein (TMEM), we chose 21 genes that are preferentially expressed in human SkM relative to 52 other tissue types and analyzed relationships between their tissue-specific epigenetics and expression. We also compared their genetics, proteomics, and descriptions in the literature. For this study, we identified genes with little or no previous descriptions of SkM functionality (ASB4, ASB8, ASB10, ASB12, ASB16, LRRC14B, LRRC20, LRRC30, TMEM52, TMEM233, OSBPL6/ORP6, and OSBPL11/ORP11) and included genes whose SkM functions had been previously addressed (ASB2, ASB5, ASB11, ASB15, LRRC2, LRRC38, LRRC39, TMEM38A/TRIC-A, and TMEM38B/TRIC-B). Some of these genes have associations with SkM or heart disease, cancer, bone disease, or other diseases. Among the transcription-related SkM epigenetic features that we identified were: super-enhancers, promoter DNA hypomethylation, lengthening of constitutive low-methylated promoter regions, and SkM-related enhancers for one gene embedded in a neighboring gene (e.g., ASB8-PFKM, LRRC39-DBT, and LRRC14B-PLEKHG4B gene-pairs). In addition, highly or lowly co-expressed long non-coding RNA (lncRNA) genes probably regulate several of these genes. Our findings give insights into tissue-specific epigenetic patterns and functionality of related genes in a gene family and can elucidate normal and disease-related regulation of gene expression in SkM.
Collapse
Affiliation(s)
- Kenneth C. Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Michelle Lacey
- Department of Mathematics, Tulane University, New Orleans, LA 70118, USA;
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA;
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
18
|
da Silva RA, Fuhler GM, Janmaat VT, da C Fernandes CJ, da Silva Feltran G, Oliveira FA, Matos AA, Oliveira RC, Ferreira MR, Zambuzzi WF, Peppelenbosch MP. HOXA cluster gene expression during osteoblast differentiation involves epigenetic control. Bone 2019; 125:74-86. [PMID: 31054377 DOI: 10.1016/j.bone.2019.04.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/25/2019] [Accepted: 04/30/2019] [Indexed: 01/17/2023]
Abstract
The HOXA gene cluster is generally recognized as a pivotal mediator of positional identity in the skeletal system, expression of different orthologues conferring alternative locational phenotype of the vertebrate bone. Strikingly, however, the molecular mechanisms that regulate orthologue-specific expression of different HOXA cluster members in gestating osteoblasts remain largely obscure, but in analogy to the processes observed in acute lymphatic leukemia it is assumed that alternative methylation of HOXA promoter regions drives position specific expression patterns. In an effort to understand HOXA cluster gene expression in osteogenesis we characterize both expression and the epigenetic landscape of the HOXA gene cluster during in vitro osteoblast formation from mesenchymal precursors. We observe that osteoblast formation per se provokes strong upregulation of HOXA gene cluster expression, in particular of midcluster genes, and paradoxal downregulation of HOXA7 and HOXA10. These differences in expression appear related to promoter methylation. LnRNAs HOTAIR and HOTTIP, known to modulate HOXA expression, are also regulated by their promoter methylation processing, but do not correlate with HOXA cluster expression profile. We thus conclude that HOXA expression is profoundly regulated during osteoblast differentiation through canonical methylation-dependent mechanisms but not through the flanking lnRNAs.
Collapse
Affiliation(s)
- Rodrigo A da Silva
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, São Paulo State University - UNESP, Botucatu, São Paulo 18618-970, Brazil; Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Vincent T Janmaat
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Célio Júnior da C Fernandes
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, São Paulo State University - UNESP, Botucatu, São Paulo 18618-970, Brazil
| | - Geórgia da Silva Feltran
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, São Paulo State University - UNESP, Botucatu, São Paulo 18618-970, Brazil
| | - Flávia Amadeu Oliveira
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, 17012-901 Bauru, São Paulo, Brazil
| | - Adriana Arruda Matos
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, 17012-901 Bauru, São Paulo, Brazil
| | - Rodrigo Cardoso Oliveira
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, 17012-901 Bauru, São Paulo, Brazil
| | - Marcel Rodrigues Ferreira
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, São Paulo State University - UNESP, Botucatu, São Paulo 18618-970, Brazil
| | - Willian F Zambuzzi
- Laboratory of Bioassays and Cellular Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, São Paulo State University - UNESP, Botucatu, São Paulo 18618-970, Brazil.
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
19
|
Mahmoudian RA, Bahadori B, Rad A, Abbaszadegan MR, Forghanifard MM. MEIS1 knockdown may promote differentiation of esophageal squamous carcinoma cell line KYSE-30. Mol Genet Genomic Med 2019; 7:e00746. [PMID: 31090196 PMCID: PMC6625128 DOI: 10.1002/mgg3.746] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/01/2019] [Accepted: 04/27/2019] [Indexed: 12/11/2022] Open
Abstract
Background MEIS1 (Myeloid ecotropic viral integration site 1), as a homeobox (HOX) transcription factor, has a dual function in different types of cancer. Although numerous roles are proposed for MEIS1 in differentiation, stem cell function, gastrointestinal development and tumorigenesis, the involved molecular mechanisms are poor understood. Our aim in this study was to elucidate the functional correlation between MEIS1, as regulator of differentiation process, and the involved genes in cell differentiation in human esophageal squamous carcinoma (ESC) cell line KYSE‐30. Methods The KYSE‐30 cells were transduced using recombinant retroviral particles containing specific shRNA sequence against MEIS1 to knockdown MEIS1 gene expression. Following RNA extraction and cDNA synthesis, mRNA expression of MEIS1 and the selected genes including TWIST1, EGF, CDX2, and KRT4 was examined using relative comparative real‐time PCR. Results Retroviral transduction caused a significant underexpression of MEIS1 in GFP‐hMEIS1 compared to control GFP cells approximately 5.5‐fold. While knockdown of MEIS1 expression caused a significant decrease in EGF and TWIST1 mRNA expression, nearly ‐8‐ and ‐12‐fold respectively, it caused a significant increase in mRNA expression of differentiation markers including KRT4 and CDX2, approximately 34‐ and 1.14‐fold, correspondingly. Conclusion MEIS1 gene silencing in KYSE‐30 cells increased expression of epithelial markers and decreased expression of epithelial‐mesenchymal transition (EMT) marker TWIST1. It may highlight the role of MEIS1 in differentiation process of KYSE‐30 cells. These results may confirm that MEIS1 silencing promotes differentiation and decreases EMT capability of ESC cell line KYSE‐30.
Collapse
Affiliation(s)
| | - Bahareh Bahadori
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Abolfazl Rad
- Cellular and Molecular Research center, Sabzevar Univeristy of Medical Sciences, Sabzevar, Iran
| | | | | |
Collapse
|
20
|
de Las Heras-Saldana S, Chung KY, Lee SH, Gondro C. Gene expression of Hanwoo satellite cell differentiation in longissimus dorsi and semimembranosus. BMC Genomics 2019; 20:156. [PMID: 30808286 PMCID: PMC6390542 DOI: 10.1186/s12864-019-5530-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Korean Hanwoo cattle are known for their high meat quality, especially their high intramuscular fat compared to most other cattle breeds. Different muscles have very different meat quality traits and a study of the myogenic process in satellite cells can help us better understand the genes and pathways that regulate this process and how muscles differentiate. RESULTS Cell cultures of Longissimus dorsi muscle differentiated from myoblast into multinucleated myotubes faster than semimembranosus. Time-series RNA-seq identified a total of 13 differentially expressed genes between the two muscles during their development. These genes seem to be involved in determining muscle lineage development and appear to modulate the expression of myogenic regulatory factors (mainly MYOD and MYF5) during differentiation of satellite cells into multinucleate myotubes. Gene ontology enriched terms were consistent with the morphological changes observed in the histology. Most of the over-represented terms and genes expressed during myoblast differentiation were similar regardless of muscle type which indicates a highly conserved myogenic process albeit the rates of differentiation being different. There were more differences in the enriched GO terms during the end of proliferation compared to myoblast differentiation. CONCLUSIONS The use of satellite cells from newborn Hanwoo calves appears to be a good model to study embryonic myogenesis in muscle. Our findings provide evidence that the differential expression of HOXB2, HOXB4, HOXB9, HOXC8, FOXD1, IGFN1, ZIC2, ZIC4, HOXA11, HOXC11, PITX1, SIM2 and TBX4 genes could be involved in the differentiation of Longissimus dorsi and Semimembranosus muscles. These genes seem to modulate the muscle fate of the satellite cells during myogenesis through a differential expression profile that also controls the expression of some myogenic regulatory factors (MYOD and MYF5). The number of differentially expressed genes across time was unsurprisingly large. In relation to the baseline day 0, there were 631, 155, 175, 519 and 586 DE genes in LD, while in SM we found 204, 0, 615, 761 and 1154 DE genes at days 1, 2, 4, 7 and 14 respectively.
Collapse
Affiliation(s)
| | - Ki Yong Chung
- Hanwoo Research Institute, National Institute of Animal Science, RDA, Pyeongchang, South Korea
| | - Seung Hwan Lee
- Division of Animal and Dairy Science, Chungnam National University, Deajeon, South Korea.
| | - Cedric Gondro
- Department of Animal Science, Michigan State University, 474 S Shaw Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
21
|
Schubert MF, Noah AC, Bedi A, Gumucio JP, Mendias CL. Reduced Myogenic and Increased Adipogenic Differentiation Capacity of Rotator Cuff Muscle Stem Cells. J Bone Joint Surg Am 2019; 101:228-238. [PMID: 30730482 PMCID: PMC6791507 DOI: 10.2106/jbjs.18.00509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Fat accumulation commonly occurs in chronically torn rotator cuff muscles, and increased fat within the rotator cuff is correlated with poor clinical outcomes. The extent of lipid deposition is particularly pronounced in injured rotator cuff muscles compared with other commonly injured muscles such as the gastrocnemius. Satellite cells, which are a tissue-resident muscle stem-cell population, can differentiate into fat cells. We hypothesized that satellite cells from the rotator cuff have greater intrinsic adipogenic differentiation potential than do gastrocnemius satellite cells, and this difference is due to variations in epigenetic imprinting between the cells. METHODS Satellite cells from gastrocnemius and rotator cuff muscles of mice were cultured in adipogenic media, and the capacity to differentiate into mature muscle cells and adipogenic cells was assessed (n ≥ 9 plates per muscle group). We also performed DNA methylation analysis of gastrocnemius and rotator cuff satellite cells to determine whether epigenetic differences were present between the 2 groups (n = 5 mice per group). RESULTS Compared with the gastrocnemius, satellite cells from the rotator cuff had a 23% reduction in myogenic differentiation and an 87% decrease in the expression of the differentiated muscle cell marker MRF4 (myogenic regulatory factor 4). With respect to adipogenesis, rotator cuff satellite cells had a 4.3-fold increase in adipogenesis, a 12-fold increase in the adipogenic transcription factor PPARγ (peroxisome proliferator-activated receptor gamma), and a 65-fold increase in the adipogenic marker FABP4 (fatty-acid binding protein 4). Epigenetic analysis identified 355 differentially methylated regions of DNA between rotator cuff and gastrocnemius satellite cells, and pathway enrichment analysis suggested that these regions were involved with lipid metabolism and adipogenesis. CONCLUSIONS Satellite cells from rotator cuff muscles have reduced myogenic and increased adipogenic differentiation potential compared with gastrocnemius muscles. There appears to be a cellular and genetic basis behind the generally poor rates of rotator cuff muscle healing. CLINICAL RELEVANCE The reduced myogenic and increased adipogenic capacity of rotator cuff satellite cells is consistent with the increased fat content and poor muscle healing rates often observed for chronically torn rotator cuff muscles. For patients undergoing rotator cuff repair, transplantation of autologous satellite cells from other muscles less prone to fatty infiltration may improve clinical outcomes.
Collapse
Affiliation(s)
- Manuel F. Schubert
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew C. Noah
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Asheesh Bedi
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Jonathan P. Gumucio
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Christopher L. Mendias
- Departments of Orthopaedic Surgery (M.F.S., A.C.N., A.B, J.P.G, and C.L.M.) and Molecular and Integrative Physiology (A.C.N, J.P.G., and C.L.M.), University of Michigan Medical School, Ann Arbor, Michigan,Hospital for Special Surgery, New York, NY,Departments of Physiology and Biophysics and Orthopaedic Surgery, Weill Cornell Medical College, New York, NY
| |
Collapse
|
22
|
Terry EE, Zhang X, Hoffmann C, Hughes LD, Lewis SA, Li J, Wallace MJ, Riley LA, Douglas CM, Gutierrez-Monreal MA, Lahens NF, Gong MC, Andrade F, Esser KA, Hughes ME. Transcriptional profiling reveals extraordinary diversity among skeletal muscle tissues. eLife 2018; 7:34613. [PMID: 29809149 PMCID: PMC6008051 DOI: 10.7554/elife.34613] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/15/2018] [Indexed: 11/24/2022] Open
Abstract
Skeletal muscle comprises a family of diverse tissues with highly specialized functions. Many acquired diseases, including HIV and COPD, affect specific muscles while sparing others. Even monogenic muscular dystrophies selectively affect certain muscle groups. These observations suggest that factors intrinsic to muscle tissues influence their resistance to disease. Nevertheless, most studies have not addressed transcriptional diversity among skeletal muscles. Here we use RNAseq to profile mRNA expression in skeletal, smooth, and cardiac muscle tissues from mice and rats. Our data set, MuscleDB, reveals extensive transcriptional diversity, with greater than 50% of transcripts differentially expressed among skeletal muscle tissues. We detect mRNA expression of hundreds of putative myokines that may underlie the endocrine functions of skeletal muscle. We identify candidate genes that may drive tissue specialization, including Smarca4, Vegfa, and Myostatin. By demonstrating the intrinsic diversity of skeletal muscles, these data provide a resource for studying the mechanisms of tissue specialization. About 40% of our weight is formed of skeletal muscles, the hundreds of muscles in our bodies that can be voluntarily controlled by our nervous system. At the moment, the research community largely sees all these muscles as a single group whose tissues are virtually interchangeable. Yet, skeletal muscles have highly diverse origins, shapes and roles. For example, our diaphragm is a long muscle that contracts slowly and rhythmically so we can draw breaths, while tiny muscles in our eyes generate the short and precise movements of our eyeballs. Different skeletal muscles also respond in distinct ways to injuries, drugs and diseases. This suggests that these muscles may be diverse at the genetic level. While all the cells in our body have the same genetic information, exactly which genes are turned on and off (or ‘expressed’) changes between types of cells. On top of this ‘on or off’ regulation, the level of expression of a gene – how active it is – can also differ. However, the studies that examine the differences in gene expression between tissues usually overlook skeletal muscles. Here, Terry et al. use genetic techniques to measure how genes are expressed in over 20 types of muscle in mice and rats. The results show that the expression levels of over 50% of all the animals’ genes vary between muscles. In fact, any two types of muscles express on average 13% of their genes differently from each other. The analyses yield further unexpected findings. For example, the expression levels in a muscle in the foot that helps to flex the rodents’ toes are more similar to those found in eye muscles than to the ones observed in limb muscles. These conclusions indicate that skeletal muscles are a widely diverse family of tissues. The research community will be able to use the data collected by Terry et al. to explore further the origins and the consequences of the differences between skeletal muscles. This could help researchers to understand why specific groups of muscles are more susceptible to disease, or react differently to a drug. This knowledge could also be exploited to refine approaches in tissue engineering, which aims to replace damaged muscles in the body.
Collapse
Affiliation(s)
- Erin E Terry
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States
| | - Xiping Zhang
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, United States
| | - Christy Hoffmann
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States
| | - Laura D Hughes
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute, La Jolla, United States
| | - Scott A Lewis
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States
| | - Jiajia Li
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States
| | - Matthew J Wallace
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States
| | - Lance A Riley
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, United States
| | - Collin M Douglas
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, United States
| | - Miguel A Gutierrez-Monreal
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, United States
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Ming C Gong
- Department of Physiology, University of Kentucky School of Medicine, Lexington, United States
| | - Francisco Andrade
- Department of Physiology, University of Kentucky School of Medicine, Lexington, United States
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, United States
| | - Michael E Hughes
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
23
|
Baribault C, Ehrlich KC, Ponnaluri VKC, Pradhan S, Lacey M, Ehrlich M. Developmentally linked human DNA hypermethylation is associated with down-modulation, repression, and upregulation of transcription. Epigenetics 2018; 13:275-289. [PMID: 29498561 PMCID: PMC5997157 DOI: 10.1080/15592294.2018.1445900] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA methylation can affect tissue-specific gene transcription in ways that are difficult to discern from studies focused on genome-wide analyses of differentially methylated regions (DMRs). To elucidate the variety of associations between differentiation-related DNA hypermethylation and transcription, we used available epigenomic and transcriptomic profiles from 38 human cell/tissue types to focus on such relationships in 94 genes linked to hypermethylated DMRs in myoblasts (Mb). For 19 of the genes, promoter-region hypermethylation in Mb (and often a few heterologous cell types) was associated with gene repression but, importantly, DNA hypermethylation was absent in many other repressed samples. In another 24 genes, DNA hypermethylation overlapped cryptic enhancers or super-enhancers and correlated with down-modulated, but not silenced, gene expression. However, such methylation was absent, surprisingly, in both non-expressing samples and highly expressing samples. This suggests that some genes need DMR hypermethylation to help repress cryptic enhancer chromatin only when they are actively transcribed. For another 11 genes, we found an association between intergenic hypermethylated DMRs and positive expression of the gene in Mb. DNA hypermethylation/transcription correlations similar to those of Mb were evident sometimes in diverse tissues, such as aorta and brain. Our findings have implications for the possible involvement of methylated DNA in Duchenne's muscular dystrophy, congenital heart malformations, and cancer. This epigenomic analysis suggests that DNA methylation is not simply the inevitable consequence of changes in gene expression but, instead, is often an active agent for fine-tuning transcription in association with development.
Collapse
Affiliation(s)
- Carl Baribault
- a Tulane Cancer Center , Tulane University Health Sciences Center , New Orleans , LA 70112 , USA.,b Department of Mathematics , Tulane University , New Orleans , LA 70118 , USA
| | - Kenneth C Ehrlich
- c Center for Bioinformatics and Genomics , Tulane University Health Sciences Center , New Orleans , LA 70112 , USA
| | | | | | - Michelle Lacey
- b Department of Mathematics , Tulane University , New Orleans , LA 70118 , USA
| | - Melanie Ehrlich
- a Tulane Cancer Center , Tulane University Health Sciences Center , New Orleans , LA 70112 , USA.,c Center for Bioinformatics and Genomics , Tulane University Health Sciences Center , New Orleans , LA 70112 , USA.,e Hayward Genetics Center Tulane University Health Sciences Center , New Orleans , LA 70112 , USA
| |
Collapse
|
24
|
Rejlova K, Musilova A, Kramarzova KS, Zaliova M, Fiser K, Alberich-Jorda M, Trka J, Starkova J. Low HOX gene expression in PML-RARα-positive leukemia results from suppressed histone demethylation. Epigenetics 2018; 13:73-84. [PMID: 29224413 DOI: 10.1080/15592294.2017.1413517] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Homeobox (HOX) genes are frequently dysregulated in leukemia. Previous studies have shown that aberrant HOX gene expression accompanies leukemogenesis and affects disease progression and leukemia patient survival. Patients with acute myeloid leukemia (AML) bearing PML-RARα fusion gene have distinct HOX gene signature in comparison to other subtypes of AML patients, although the mechanism of transcription regulation is not completely understood. We previously found an association between the mRNA levels of HOX genes and those of the histone demethylases JMJD3 and UTX in PML-RARα- positive leukemia patients. Here, we demonstrate that the release of the PML-RARα-mediated block in PML-RARα-positive myeloid leukemia cells increased both JMJD3 and HOX gene expression, while inhibition of JMJD3 using the specific inhibitor GSK-J4 reversed the effect. This effect was driven specifically through PML-RARα fusion protein since expression changes did not occur in cells with mutated RARα and was independent of differentiation. We confirmed that gene expression levels were inversely correlated with alterations in H3K27me3 histone marks localized at HOX gene promoters. Furthermore, data from chromatin immunoprecipitation followed by sequencing broaden a list of clustered HOX genes regulated by JMJD3 in PML-RARα-positive leukemic cells. Interestingly, the combination of GSK-J4 and all-trans retinoic acid (ATRA) significantly increased PML-RARα-positive cell apoptosis compared with ATRA treatment alone. This effect was also observed in ATRA-resistant NB4 clones, which may provide a new therapeutic opportunity for patients with acute promyelocytic leukemia (APL) resistant to current treatment. The results of our study reveal the mechanism of HOX gene expression regulation and contribute to our understanding of APL pathogenesis.
Collapse
Affiliation(s)
- Katerina Rejlova
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic
| | - Alena Musilova
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic
| | - Karolina Skvarova Kramarzova
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic
| | - Marketa Zaliova
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic
| | - Karel Fiser
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic
| | | | - Jan Trka
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic.,c University Hospital Motol , Prague , Czech Republic
| | - Julia Starkova
- a CLIP - Childhood Leukaemia Investigation Prague.,b Department of Pediatric Hematology and Oncology , Second Faculty of Medicine, Charles University , Prague , Czech Republic
| |
Collapse
|
25
|
Increased methylation and decreased expression of homeobox genes TLX1, HOXA10 and DLX5 in human placenta are associated with trophoblast differentiation. Sci Rep 2017; 7:4523. [PMID: 28674422 PMCID: PMC5495813 DOI: 10.1038/s41598-017-04776-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 05/22/2017] [Indexed: 01/30/2023] Open
Abstract
Homeobox genes regulate embryonic and placental development, and are widely expressed in the human placenta, but their regulatory control by DNA methylation is unclear. DNA methylation analysis was performed on human placentae from first, second and third trimesters to determine methylation patterns of homeobox gene promoters across gestation. Most homeobox genes were hypo-methylated throughout gestation, suggesting that DNA methylation is not the primary mechanism involved in regulating HOX genes expression in the placenta. Nevertheless, several genes showed variable methylation patterns across gestation, with a general trend towards an increase in methylation over gestation. Three genes (TLX1, HOXA10 and DLX5) showed inverse gains of methylation with decreasing mRNA expression throughout pregnancy, supporting a role for DNA methylation in their regulation. Proteins encoded by these genes were primarily localised to the syncytiotrophoblast layer, and showed decreased expression later in gestation. siRNA mediated downregulation of DLX5, TLX1 and HOXA10 in primary term villous cytotrophoblast resulted in decreased proliferation and increased expression of differentiation markers, including ERVW-1. Our data suggest that loss of DLX5, TLX1 and HOXA10 expression in late gestation is required for proper placental differentiation and function.
Collapse
|
26
|
Hwang SY, Kang YJ, Sung B, Jang JY, Hwang NL, Oh HJ, Ahn YR, Kim HJ, Shin JH, Yoo MA, Kim CM, Chung HY, Kim ND. Folic acid is necessary for proliferation and differentiation of C2C12 myoblasts. J Cell Physiol 2017; 233:736-747. [PMID: 28471487 DOI: 10.1002/jcp.25989] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 05/03/2017] [Indexed: 02/04/2023]
Abstract
Folic acid, a water soluble B vitamin, plays an important role in cellular metabolic activities, such as functioning as a cofactor in one-carbon metabolism for DNA and RNA synthesis as well as nucleotide and amino acid biosynthesis in the body. A lack of dietary folic acid can lead to folic acid deficiency and result in several health problems, including macrocytic anemia, elevated plasma homocysteine, cardiovascular disease, birth defects, carcinogenesis, muscle weakness, and walking difficulty. However, the effect of folic acid deficiency on skeletal muscle development and its molecular mechanisms are unknown. We, therefore, investigated the effect of folic acid deficiency on myogenesis in skeletal muscle cells and found that folic acid deficiency induced proliferation inhibition and cell cycle breaking as well as cellular senescence in C2C12 myoblasts, implying that folic acid deficiency influences skeletal muscle development. Folic acid deficiency also inhibited differentiation of C2C12 myoblasts and induced deregulation of the cell cycle exit and many cell cycle regulatory genes. It inhibited expression of muscle-specific marker MyHC as well as myogenic regulatory factor (myogenin). Moreover, immunocytochemistry and Western blot analyses revealed that DNA damage was more increased in folic acid-deficient medium-treated differentiating C2C12 cells. Furthermore, we found that folic acid resupplementation reverses the effect on the cell cycle and senescence in folic acid-deficient C2C12 myoblasts but does not reverse the differentiation of C2C12 cells. Altogether, the study results suggest that folic acid is necessary for normal development of skeletal muscle cells.
Collapse
Affiliation(s)
- Seong Y Hwang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yong J Kang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Bokyung Sung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Jung Y Jang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Na L Hwang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Hye J Oh
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yu R Ahn
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Hong J Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Jin H Shin
- Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Mi-Ae Yoo
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Cheol M Kim
- Research Center for Anti-Aging Technology Development, Pusan National University, Busan, Republic of Korea.,Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hae Y Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Republic of Korea
| | - Nam D Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Republic of Korea.,Research Center for Anti-Aging Technology Development, Pusan National University, Busan, Republic of Korea.,Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
27
|
Frank-Bertoncelj M, Trenkmann M, Klein K, Karouzakis E, Rehrauer H, Bratus A, Kolling C, Armaka M, Filer A, Michel BA, Gay RE, Buckley CD, Kollias G, Gay S, Ospelt C. Epigenetically-driven anatomical diversity of synovial fibroblasts guides joint-specific fibroblast functions. Nat Commun 2017; 8:14852. [PMID: 28332497 PMCID: PMC5376654 DOI: 10.1038/ncomms14852] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 02/02/2017] [Indexed: 01/15/2023] Open
Abstract
A number of human diseases, such as arthritis and atherosclerosis, include characteristic pathology in specific anatomical locations. Here we show transcriptomic differences in synovial fibroblasts from different joint locations and that HOX gene signatures reflect the joint-specific origins of mouse and human synovial fibroblasts and synovial tissues. Alongside DNA methylation and histone modifications, bromodomain and extra-terminal reader proteins regulate joint-specific HOX gene expression. Anatomical transcriptional diversity translates into joint-specific synovial fibroblast phenotypes with distinct adhesive, proliferative, chemotactic and matrix-degrading characteristics and differential responsiveness to TNF, creating a unique microenvironment in each joint. These findings indicate that local stroma might control positional disease patterns not only in arthritis but in any disease with a prominent stromal component. Arthritis affects different joints variably despite systemic inflammatory cues. Here the authors show anatomical differences in the transcriptome, epigenome and function of synovial fibroblasts that might affect susceptibility to site-specific joint diseases.
Collapse
Affiliation(s)
- Mojca Frank-Bertoncelj
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland
| | - Michelle Trenkmann
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland
| | - Kerstin Klein
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland
| | - Emmanuel Karouzakis
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anna Bratus
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | | | - Maria Armaka
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming', 34, Fleming Street, 16672 Vari, Attica, Greece
| | - Andrew Filer
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - Beat A Michel
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland
| | - Renate E Gay
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland
| | - Christopher D Buckley
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - George Kollias
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming', 34, Fleming Street, 16672 Vari, Attica, Greece.,Department of Experimental Physiology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Goudi, Athens, Greece
| | - Steffen Gay
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland.,Center of Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Caroline Ospelt
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Wagistrasse 14, 8952 Schlieren, Zurich, Switzerland.,Center of Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
28
|
Ponnaluri VKC, Ehrlich KC, Zhang G, Lacey M, Johnston D, Pradhan S, Ehrlich M. Association of 5-hydroxymethylation and 5-methylation of DNA cytosine with tissue-specific gene expression. Epigenetics 2017; 12:123-138. [PMID: 27911668 PMCID: PMC5330441 DOI: 10.1080/15592294.2016.1265713] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022] Open
Abstract
Differentially methylated or hydroxymethylated regions (DMRs) in mammalian DNA are often associated with tissue-specific gene expression but the functional relationships are still being unraveled. To elucidate these relationships, we studied 16 human genes containing myogenic DMRs by analyzing profiles of their epigenetics and transcription and quantitatively assaying 5-hydroxymethylcytosine (5hmC) and 5-methylcytosine (5mC) at specific sites in these genes in skeletal muscle (SkM), myoblasts, heart, brain, and diverse other samples. Although most human promoters have little or no methylation regardless of expression, more than half of the genes that we chose to study-owing to their myogenic DMRs-overlapped tissue-specific alternative or cryptic promoters displaying corresponding tissue-specific differences in histone modifications. The 5mC levels in myoblast DMRs were significantly associated with 5hmC levels in SkM at the same site. Hypermethylated myogenic DMRs within CDH15, a muscle- and cerebellum-specific cell adhesion gene, and PITX3, a homeobox gene, were used for transfection in reporter gene constructs. These intragenic DMRs had bidirectional tissue-specific promoter activity that was silenced by in vivo-like methylation. The CDH15 DMR, which was previously associated with an imprinted maternal germline DMR in mice, had especially strong promoter activity in myogenic host cells. These findings are consistent with the controversial hypothesis that intragenic DNA methylation can facilitate transcription and is not just a passive consequence of it. Our results support varied roles for tissue-specific 5mC- or 5hmC-enrichment in suppressing inappropriate gene expression from cryptic or alternative promoters and in increasing the plasticity of gene expression required for development and rapid responses to tissue stress or damage.
Collapse
Affiliation(s)
| | - Kenneth C. Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA, USA
| | | | - Michelle Lacey
- Department of Mathematics, Tulane Health Sciences Center and Tulane University, New Orleans, LA, USA
| | - Douglas Johnston
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA, USA
| | | | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA, USA
- Hayward Genetics Center and Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
29
|
Ehrlich KC, Paterson HL, Lacey M, Ehrlich M. DNA Hypomethylation in Intragenic and Intergenic Enhancer Chromatin of Muscle-Specific Genes Usually Correlates with their Expression. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2016; 89:441-455. [PMID: 28018137 PMCID: PMC5168824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tissue-specific enhancers are critical for gene regulation. In this study, we help elucidate the contribution of muscle-associated differential DNA methylation to the enhancer activity of highly muscle-specific genes. By bioinformatic analysis of 44 muscle-associated genes, we show that preferential gene expression in skeletal muscle (SkM) correlates with SkM-specific intragenic and intergenic enhancer chromatin and overlapping foci of DNA hypomethylation. Some genes, e.g., CASQ1 and FBXO32, displayed broad regions of both SkM- and heart-specific enhancer chromatin but exhibited focal SkM-specific DNA hypomethylation. Half of the genes had SkM-specific super-enhancers. In contrast to simple enhancer/gene-expression correlations, a super-enhancer was associated with the myogenic MYOD1 gene in both SkM and myoblasts even though SkM has < 1 percent as much MYOD1 expression. Local chromatin differences in this super-enhancer probably contribute to the SkM/myoblast differential expression. Transfection assays confirmed the tissue-specificity of the 0.3-kb core enhancer within MYOD1's super-enhancer and demonstrated its repression by methylation of its three CG dinucleotides. Our study suggests that DNA hypomethylation increases enhancer tissue-specificity and that SkM super-enhancers sometimes are poised for physiologically important, rapid up-regulation.
Collapse
Affiliation(s)
- Kenneth C. Ehrlich
- Program in Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA
| | | | - Michelle Lacey
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA,Mathematics Department, Tulane University, New Orleans, LA
| | - Melanie Ehrlich
- Program in Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA,Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA,Hayward Genetics Center, Tulane University Health Sciences Center, New Orleans, LA,To whom all correspondence should be addressed: Melanie Ehrlich, PhD, Hayward Genetics Center, Tulane University Health Sciences Center, 1430 Tulane Ave., New Orleans, LA 70112; Tele: 504-988-2449; Fax: 504-988-1763;
| |
Collapse
|
30
|
Epigenetic Reprogramming of Muscle Progenitors: Inspiration for Clinical Therapies. Stem Cells Int 2015; 2016:6093601. [PMID: 26839565 PMCID: PMC4709771 DOI: 10.1155/2016/6093601] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/14/2015] [Accepted: 09/16/2015] [Indexed: 01/23/2023] Open
Abstract
In the context of regenerative medicine, based on the potential of stem cells to restore diseased tissues, epigenetics is becoming a pivotal area of interest. Therapeutic interventions that promote tissue and organ regeneration have as primary objective the selective control of gene expression in adult stem cells. This requires a deep understanding of the epigenetic mechanisms controlling transcriptional programs in tissue progenitors. This review attempts to elucidate the principle epigenetic regulations responsible of stem cells differentiation. In particular we focus on the current understanding of the epigenetic networks that regulate differentiation of muscle progenitors by the concerted action of chromatin-modifying enzymes and noncoding RNAs. The novel exciting role of exosome-bound microRNA in mediating epigenetic information transfer is also discussed. Finally we show an overview of the epigenetic strategies and therapies that aim to potentiate muscle regeneration and counteract the progression of Duchenne Muscular Dystrophy (DMD).
Collapse
|
31
|
Xin Y, Li Z, Chan MT, Wu WKK. Circulating epigenetic biomarkers in melanoma. Tumour Biol 2015; 37:1487-92. [DOI: 10.1007/s13277-015-4599-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/07/2015] [Indexed: 01/21/2023] Open
|
32
|
Ehrlich M, Ehrlich KC. DNA cytosine methylation and hydroxymethylation at the borders. Epigenomics 2015; 6:563-6. [PMID: 25531248 DOI: 10.2217/epi.14.48] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
33
|
Chandra S, Terragni J, Zhang G, Pradhan S, Haushka S, Johnston D, Baribault C, Lacey M, Ehrlich M. Tissue-specific epigenetics in gene neighborhoods: myogenic transcription factor genes. Hum Mol Genet 2015; 24:4660-73. [PMID: 26041816 PMCID: PMC4512632 DOI: 10.1093/hmg/ddv198] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/18/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022] Open
Abstract
Myogenic regulatory factor (MRF) genes, MYOD1, MYOG, MYF6 and MYF5, are critical for the skeletal muscle lineage. Here, we used various epigenome profiles from human myoblasts (Mb), myotubes (Mt), muscle and diverse non-muscle samples to elucidate the involvement of multigene neighborhoods in the regulation of MRF genes. We found more far-distal enhancer chromatin associated with MRF genes in Mb and Mt than previously reported from studies in mice. For the MYF5/MYF6 gene-pair, regions of Mb-associated enhancer chromatin were located throughout the adjacent 236-kb PTPRQ gene even though Mb expressed negligible amounts of PTPRQ mRNA. Some enhancer chromatin regions inside PTPRQ in Mb were also seen in PTPRQ mRNA-expressing non-myogenic cells. This suggests dual-purpose PTPRQ enhancers that upregulate expression of PTPRQ in non-myogenic cells and MYF5/MYF6 in myogenic cells. In contrast, the myogenic enhancer chromatin regions distal to MYOD1 were intergenic and up to 19 kb long. Two of them contain small, known MYOD1 enhancers, and one displayed an unusually high level of 5-hydroxymethylcytosine in a quantitative DNA hydroxymethylation assay. Unexpectedly, three regions of MYOD1-distal enhancer chromatin in Mb and Mt overlapped enhancer chromatin in umbilical vein endothelial cells, which might upregulate a distant gene (PIK3C2A). Lastly, genes surrounding MYOG were preferentially transcribed in Mt, like MYOG itself, and exhibited nearby myogenic enhancer chromatin. These neighboring chromatin regions may be enhancers acting in concert to regulate myogenic expression of multiple adjacent genes. Our findings reveal the very different and complex organization of gene neighborhoods containing closely related transcription factor genes.
Collapse
Affiliation(s)
- Sruti Chandra
- Program in Human Genetics, Tulane Cancer Center, and Center for Bioinformatics and Genomics, Tulane Health Sciences Center, New Orleans, LA 70122, USA
| | | | | | | | - Stephen Haushka
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA, and
| | - Douglas Johnston
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Carl Baribault
- Tulane Cancer Center and Department of Mathematics, Tulane Health Sciences Center and Tulane University, New Orleans, LA 70122, USA
| | - Michelle Lacey
- Tulane Cancer Center and Department of Mathematics, Tulane Health Sciences Center and Tulane University, New Orleans, LA 70122, USA
| | - Melanie Ehrlich
- Program in Human Genetics, Tulane Cancer Center, and Center for Bioinformatics and Genomics, Tulane Health Sciences Center, New Orleans, LA 70122, USA,
| |
Collapse
|
34
|
Covelo-Soto L, Saura M, Morán P. Does DNA methylation regulate metamorphosis? The case of the sea lamprey (Petromyzon marinus) as an example. Comp Biochem Physiol B Biochem Mol Biol 2015; 185:42-6. [DOI: 10.1016/j.cbpb.2015.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/20/2015] [Accepted: 03/31/2015] [Indexed: 02/03/2023]
|
35
|
Seifert A, Werheid DF, Knapp SM, Tobiasch E. Role of Hox genes in stem cell differentiation. World J Stem Cells 2015; 7:583-595. [PMID: 25914765 PMCID: PMC4404393 DOI: 10.4252/wjsc.v7.i3.583] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/20/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Hox genes are an evolutionary highly conserved gene family. They determine the anterior-posterior body axis in bilateral organisms and influence the developmental fate of cells. Embryonic stem cells are usually devoid of any Hox gene expression, but these transcription factors are activated in varying spatial and temporal patterns defining the development of various body regions. In the adult body, Hox genes are among others responsible for driving the differentiation of tissue stem cells towards their respective lineages in order to repair and maintain the correct function of tissues and organs. Due to their involvement in the embryonic and adult body, they have been suggested to be useable for improving stem cell differentiations in vitro and in vivo. In many studies Hox genes have been found as driving factors in stem cell differentiation towards adipogenesis, in lineages involved in bone and joint formation, mainly chondrogenesis and osteogenesis, in cardiovascular lineages including endothelial and smooth muscle cell differentiations, and in neurogenesis. As life expectancy is rising, the demand for tissue reconstruction continues to increase. Stem cells have become an increasingly popular choice for creating therapies in regenerative medicine due to their self-renewal and differentiation potential. Especially mesenchymal stem cells are used more and more frequently due to their easy handling and accessibility, combined with a low tumorgenicity and little ethical concerns. This review therefore intends to summarize to date known correlations between natural Hox gene expression patterns in body tissues and during the differentiation of various stem cells towards their respective lineages with a major focus on mesenchymal stem cell differentiations. This overview shall help to understand the complex interactions of Hox genes and differentiation processes all over the body as well as in vitro for further improvement of stem cell treatments in future regenerative medicine approaches.
Collapse
|
36
|
Carrió E, Suelves M. DNA methylation dynamics in muscle development and disease. Front Aging Neurosci 2015; 7:19. [PMID: 25798107 PMCID: PMC4350440 DOI: 10.3389/fnagi.2015.00019] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/15/2015] [Indexed: 12/12/2022] Open
Abstract
DNA methylation is an essential epigenetic modification for mammalian development and is crucial for the establishment and maintenance of cellular identity. Traditionally, DNA methylation has been considered as a permanent repressive epigenetic mark. However, the application of genome-wide approaches has allowed the analysis of DNA methylation in different genomic contexts revealing a more dynamic regulation than originally thought, since active DNA methylation and demethylation occur during cellular differentiation and tissue specification. Satellite cells are the primary stem cells in adult skeletal muscle and are responsible for postnatal muscle growth, hypertrophy, and muscle regeneration. This review outlines the published data regarding DNA methylation changes along the skeletal muscle program, in both physiological and pathological conditions, to better understand the epigenetic mechanisms that control myogenesis.
Collapse
Affiliation(s)
- Elvira Carrió
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC) and Health Sciences Research Institute Germans Trias I Pujol (IGTP) Badalona, Spain
| | - Mònica Suelves
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC) and Health Sciences Research Institute Germans Trias I Pujol (IGTP) Badalona, Spain
| |
Collapse
|
37
|
Regulation of skeletal muscle development and homeostasis by gene imprinting, histone acetylation and microRNA. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:309-16. [DOI: 10.1016/j.bbagrm.2015.01.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/17/2014] [Accepted: 01/10/2015] [Indexed: 12/13/2022]
|
38
|
Abstract
Cytosine methylation in DNA constitutes an important epigenetic layer of transcriptional and regulatory control in many eukaryotes. Profiling DNA methylation across the genome is critical to understanding the influence of epigenetics in normal biology and disease, such as cancer. Genome-wide analyses such as arrays and next-generation sequencing (NGS) technologies have been used to assess large fractions of the methylome at a single-base-pair resolution. However, the range of DNA methylation profiling techniques can make selecting the appropriate protocol a challenge. This chapter discusses the advantages and disadvantages of various methylome detection approaches to assess which is appropriate for the question at hand. Here, we focus on four prominent genome-wide approaches: whole-genome bisulfite sequencing (WGBS); methyl-binding domain capture sequencing (MBDCap-Seq); reduced-representation-bisulfite-sequencing (RRBS); and Infinium Methylation450 BeadChips (450 K, Illumina). We discuss some of the requirements, merits, and challenges that should be considered when choosing a methylome technology to ensure that it will be informative. In addition, we show how genome-wide methylation detection arrays and high-throughput sequencing have provided immense insight into ovarian cancer-specific methylation signatures that may serve as diagnostic biomarkers or predict patient response to epigenetic therapy.
Collapse
|
39
|
Skvarova Kramarzova K, Fiser K, Mejstrikova E, Rejlova K, Zaliova M, Fornerod M, Drabkin HA, van den Heuvel-Eibrink MM, Stary J, Trka J, Starkova J. Homeobox gene expression in acute myeloid leukemia is linked to typical underlying molecular aberrations. J Hematol Oncol 2014; 7:94. [PMID: 25539595 PMCID: PMC4310032 DOI: 10.1186/s13045-014-0094-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/04/2014] [Indexed: 02/02/2023] Open
Abstract
Background Although distinct patterns of homeobox (HOX) gene expression have been described in defined cytogenetic and molecular subsets of patients with acute myeloid leukemia (AML), it is unknown whether these patterns are the direct result of transcriptional alterations or rather represent the differentiation stage of the leukemic cell. Method To address this question, we used qPCR to analyze mRNA expression of HOXA and HOXB genes in bone marrow (BM) samples of 46 patients with AML and sorted subpopulations of healthy BM cells. These various stages of myeloid differentiation represent matched counterparts of morphological subgroups of AML. To further study the transcriptional alterations of HOX genes in hematopoiesis, we also analyzed gene expression of epigenetic modifiers in the subpopluations of healthy BM and leukemic cells. Results Unsupervised hierarchical clustering divided the AMLs into five clusters characterized by the presence of prevalent molecular genetic aberrations. Notably, the impact of genotype on HOX gene expression was significantly more pronounced than that of the differentiation stage of the blasts. This driving role of molecular aberrations was best exemplified by the repressive effect of the PML-RARa fusion gene on HOX gene expression, regardless of the presence of the FLT3/ITD mutation. Furthermore, HOX gene expression was positively correlated with mRNA levels of histone demethylases (JMJD3 and UTX) and negatively correlated with gene expression of DNA methyltranferases. No such relationships were observed in subpopulations of healthy BM cells. Conclusion Our results demonstrate that specific molecular genetic aberrations, rather than differentiation per se, underlie the observed differences in HOX gene expression in AML. Moreover, the observed correlations between epigenetic modifiers and HOX ex pression that are specific to malignant hematopoiesis, suggest their potential causal relationships. Electronic supplementary material The online version of this article (doi:10.1186/s13045-014-0094-0) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
den Hollander W, Ramos YFM, Bos SD, Bomer N, van der Breggen R, Lakenberg N, de Dijcker WJ, Duijnisveld BJ, Slagboom PE, Nelissen RGHH, Meulenbelt I. Knee and hip articular cartilage have distinct epigenomic landscapes: implications for future cartilage regeneration approaches. Ann Rheum Dis 2014; 73:2208-12. [PMID: 25261579 DOI: 10.1136/annrheumdis-2014-205980] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To elucidate the functional epigenomic landscape of articular cartilage in osteoarthritis (OA) affected knee and hip joints in relation to gene expression. METHODS Using Illumina Infinium HumanMethylation450 BeadChip arrays, genome-wide DNA methylation was measured in 31 preserved and lesioned cartilage sample pairs (14 knees and 17 hips) from patients who underwent a total joint replacement due to primary OA. Using previously published genome-wide expression data of 33 pairs of cartilage samples, of which 13 pairs were overlapping with the current methylation dataset, we assessed gene expression differences in differentially methylated regions (DMRs). RESULTS Principal component analysis of the methylation data revealed distinct clustering of knee and hip samples, irrespective of OA pathophysiology. A total of 6272 CpG dinucleotides were differentially methylated between the two joints, comprising a total of 357 DMRs containing 1817 CpGs and 245 unique genes. Enrichment analysis of genes proximal of the DMRs revealed significant enrichment for developmental pathways and homeobox (HOX) genes. Subsequent transcriptomic analysis of DMR genes exposed distinct knee and hip expression patterns. CONCLUSIONS Our findings reveal consistent DMRs between knee and hip articular cartilage that marked transcriptomic differences among HOX genes, which were not reflecting the temporal sequential HOX expression pattern during development. This implies distinct mechanisms for maintaining cartilage integrity in adulthood, thereby contributing to our understanding of cartilage homeostasis and future tissue regeneration approaches.
Collapse
Affiliation(s)
- W den Hollander
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands
| | - Y F M Ramos
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands
| | - S D Bos
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands Genomics Initiative, sponsored by the NCHA, Leiden, The Netherlands
| | - N Bomer
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands IDEAL, Leiden, The Netherlands
| | | | - N Lakenberg
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands
| | - W J de Dijcker
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands
| | | | - P E Slagboom
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands Genomics Initiative, sponsored by the NCHA, Leiden, The Netherlands IDEAL, Leiden, The Netherlands
| | | | - I Meulenbelt
- Department of Molecular Epidemiology, LUMC, Leiden, The Netherlands Genomics Initiative, sponsored by the NCHA, Leiden, The Netherlands
| |
Collapse
|
41
|
Comer BS, Ba M, Singer CA, Gerthoffer WT. Epigenetic targets for novel therapies of lung diseases. Pharmacol Ther 2014; 147:91-110. [PMID: 25448041 DOI: 10.1016/j.pharmthera.2014.11.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 12/13/2022]
Abstract
In spite of substantial advances in defining the immunobiology and function of structural cells in lung diseases there is still insufficient knowledge to develop fundamentally new classes of drugs to treat many lung diseases. For example, there is a compelling need for new therapeutic approaches to address severe persistent asthma that is insensitive to inhaled corticosteroids. Although the prevalence of steroid-resistant asthma is 5-10%, severe asthmatics require a disproportionate level of health care spending and constitute a majority of fatal asthma episodes. None of the established drug therapies including long-acting beta agonists or inhaled corticosteroids reverse established airway remodeling. Obstructive airways remodeling in patients with chronic obstructive pulmonary disease (COPD), restrictive remodeling in idiopathic pulmonary fibrosis (IPF) and occlusive vascular remodeling in pulmonary hypertension are similarly unresponsive to current drug therapy. Therefore, drugs are needed to achieve long-acting suppression and reversal of pathological airway and vascular remodeling. Novel drug classes are emerging from advances in epigenetics. Novel mechanisms are emerging by which cells adapt to environmental cues, which include changes in DNA methylation, histone modifications and regulation of transcription and translation by noncoding RNAs. In this review we will summarize current epigenetic approaches being applied to preclinical drug development addressing important therapeutic challenges in lung diseases. These challenges are being addressed by advances in lung delivery of oligonucleotides and small molecules that modify the histone code, DNA methylation patterns and miRNA function.
Collapse
Affiliation(s)
- Brian S Comer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, 36688, USA
| | - Mariam Ba
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Cherie A Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - William T Gerthoffer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, 36688, USA.
| |
Collapse
|
42
|
Marcinkiewicz KM, Gudas LJ. Altered histone mark deposition and DNA methylation at homeobox genes in human oral squamous cell carcinoma. J Cell Physiol 2014; 229:1405-16. [PMID: 24519855 DOI: 10.1002/jcp.24577] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/16/2014] [Indexed: 01/03/2023]
Abstract
We recently reported a role of polycomb repressive complex 2 (PRC2) and PRC2 trimethylation of histone 3 lysine 27 (H3K27me3) in the regulation of homeobox (HOX) (Marcinkiewicz and Gudas, 2013, Exp Cell Res) gene transcript levels in human oral keratinocytes (OKF6-TERT1R) and tongue squamous cell carcinoma (SCC) cells. Here, we assessed both the levels of various histone modifications at a subset of homeobox genes and genome wide DNA methylation patterns in OKF6-TERT1R and SCC-9 cells by using ERRBS (enhanced reduced representation bisulfite sequencing). We detected the H3K9me3 mark at HOXB7, HOXC10, HOXC13, and HOXD8 at levels higher in OKF6-TERT1R than in SCC-9 cells; at IRX1 and SIX2 the H3K9me3 levels were conversely higher in SCC-9 than in OKF6-TERT1R. The H3K79me3 mark was detectable only at IRX1 in OKF6-TERT1R and at IRX4 in SCC-9 cells. The levels of H3K4me3 and H3K36me3 marks correlate with the transcript levels of the assessed homeobox genes in both OKF6-TERT1R and SCC-9. We detected generally lower CpG methylation levels on DNA in SCC-9 cells at annotated genomic regions which were differentially methylated between OKF6-TERT1R and SCC-9 cells; however, some genomic regions, including the HOX gene clusters, showed DNA methylation at higher levels in SCC-9 than OKF6-TERT1R. Thus, both altered histone modification patterns and changes in DNA methylation are associated with dysregulation of homeobox gene expression in human oral cavity SCC cells, and this dysregulation potentially plays a role in the neoplastic phenotype of oral keratinocytes.
Collapse
Affiliation(s)
- Katarzyna M Marcinkiewicz
- Department of Pharmacology, Weill Cornell Medical College and Weill Graduate School of Biomedical Sciences of Cornell University, New York, New York
| | | |
Collapse
|
43
|
Chandra S, Baribault C, Lacey M, Ehrlich M. Myogenic differential methylation: diverse associations with chromatin structure. BIOLOGY 2014; 3:426-51. [PMID: 24949935 PMCID: PMC4085616 DOI: 10.3390/biology3020426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 11/16/2022]
Abstract
Employing a new algorithm for identifying differentially methylated regions (DMRs) from reduced representation bisulfite sequencing profiles, we identified 1972 hypermethylated and 3250 hypomethylated myogenic DMRs in a comparison of myoblasts (Mb) and myotubes (Mt) with 16 types of nonmuscle cell cultures. DMRs co-localized with a variety of chromatin structures, as deduced from ENCODE whole-genome profiles. Myogenic hypomethylation was highly associated with both weak and strong enhancer-type chromatin, while hypermethylation was infrequently associated with enhancer-type chromatin. Both myogenic hypermethylation and hypomethylation often overlapped weak transcription-type chromatin and Polycomb-repressed-type chromatin. For representative genes, we illustrate relationships between DNA methylation, the local chromatin state, DNaseI hypersensitivity, and gene expression. For example, MARVELD2 exhibited myogenic hypermethylation in transcription-type chromatin that overlapped a silenced promoter in Mb and Mt while TEAD4 had myogenic hypomethylation in intronic subregions displaying enhancer-type or transcription-type chromatin in these cells. For LSP1, alternative promoter usage and active promoter-type chromatin were linked to highly specific myogenic or lymphogenic hypomethylated DMRs. Lastly, despite its myogenesis-associated expression, TBX15 had multiple hypermethylated myogenic DMRs framing its promoter region. This could help explain why TBX15 was previously reported to be underexpressed and, unexpectedly, its promoter undermethylated in placentas exhibiting vascular intrauterine growth restriction.
Collapse
Affiliation(s)
- Sruti Chandra
- Center for Bioinformatics and Genomics, New Orleans, LA 70112, USA.
| | - Carl Baribault
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Michelle Lacey
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, New Orleans, LA 70112, USA.
| |
Collapse
|
44
|
Terragni J, Zhang G, Sun Z, Pradhan S, Song L, Crawford GE, Lacey M, Ehrlich M. Notch signaling genes: myogenic DNA hypomethylation and 5-hydroxymethylcytosine. Epigenetics 2014; 9:842-50. [PMID: 24670287 PMCID: PMC4065182 DOI: 10.4161/epi.28597] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/11/2014] [Accepted: 03/19/2014] [Indexed: 12/31/2022] Open
Abstract
Notch intercellular signaling is critical for diverse developmental pathways and for homeostasis in various types of stem cells and progenitor cells. Because Notch gene products need to be precisely regulated spatially and temporally, epigenetics is likely to help control expression of Notch signaling genes. Reduced representation bisulfite sequencing (RRBS) indicated significant hypomethylation in myoblasts, myotubes, and skeletal muscle vs. many nonmuscle samples at intragenic or intergenic regions of the following Notch receptor or ligand genes: NOTCH1, NOTCH2, JAG2, and DLL1. An enzymatic assay of sites in or near these genes revealed unusually high enrichment of 5-hydroxymethylcytosine (up to 81%) in skeletal muscle, heart, and cerebellum. Epigenetics studies and gene expression profiles suggest that hypomethylation and/or hydroxymethylation help control expression of these genes in heart, brain, myoblasts, myotubes, and within skeletal muscle myofibers. Such regulation could promote cell renewal, cell maintenance, homeostasis, and a poised state for repair of tissue damage.
Collapse
Affiliation(s)
| | | | - Zhiyi Sun
- New England Biolabs; Ipswich, MA USA
| | | | - Lingyun Song
- Institute for Genome Sciences & Policy; Duke University; Durham, NC USA
| | | | - Michelle Lacey
- Tulane Cancer Center and Department of Mathematics; Tulane Health Sciences Center and Tulane University; New Orleans, LA USA
| | - Melanie Ehrlich
- Program in Human Genetics; Tulane Cancer Center; Center for Bioinformatics and Genomics; Tulane Health Sciences Center; New Orleans, LA USA
| |
Collapse
|
45
|
Ehrlich M, Lacey M. DNA methylation and differentiation: silencing, upregulation and modulation of gene expression. Epigenomics 2013; 5:553-68. [PMID: 24059801 PMCID: PMC3864898 DOI: 10.2217/epi.13.43] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Differentiation-related DNA methylation is receiving increasing attention, partly owing to new, whole-genome analyses. These revealed that cell type-specific differential methylation in gene bodies is more frequent than in promoters. We review new insights into the functionality of DNA methylation during differentiation, with emphasis on the methylomes of myoblasts, myotubes and skeletal muscle versus non-muscle samples. Biostatistical analyses of data from reduced representation bisulfite sequencing are discussed. Lastly, a model is presented for how promoter and intragenic DNA hypermethylation affect gene expression, including increasing the efficiency of polycomb silencing at some promoters, downmodulating other promoters rather than silencing them, counteracting enhancers with heterologous specificity, altering chromatin conformation by inhibiting the binding of CTCF, modulating mRNA transcript levels by inhibiting overlapping promoters of noncoding RNA genes or by regulating the use of alternative mRNA promoters, modulating transcription termination, regulating alternative splicing and acting as barriers to the spread of activating chromatin.
Collapse
Affiliation(s)
- Melanie Ehrlich
- Hayward Human Genetics Program, Tulane Cancer Center, and Center for Bioinformatics & Genomics, Tulane Health Sciences Center, New Orleans, LA 70112, USA.
| | | |
Collapse
|