1
|
Endo T. Postnatal skeletal muscle myogenesis governed by signal transduction networks: MAPKs and PI3K-Akt control multiple steps. Biochem Biophys Res Commun 2023; 682:223-243. [PMID: 37826946 DOI: 10.1016/j.bbrc.2023.09.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Skeletal muscle myogenesis represents one of the most intensively and extensively examined systems of cell differentiation, tissue formation, and regeneration. Muscle regeneration provides an in vivo model system of postnatal myogenesis. It comprises multiple steps including muscle stem cell (or satellite cell) quiescence, activation, migration, myogenic determination, myoblast proliferation, myocyte differentiation, myofiber maturation, and hypertrophy. A variety of extracellular signaling and subsequent intracellular signal transduction pathways or networks govern the individual steps of postnatal myogenesis. Among them, MAPK pathways (the ERK, JNK, p38 MAPK, and ERK5 pathways) and PI3K-Akt signaling regulate multiple steps of myogenesis. Ca2+, cytokine, and Wnt signaling also participate in several myogenesis steps. These signaling pathways often control cell cycle regulatory proteins or the muscle-specific MyoD family and the MEF2 family of transcription factors. This article comprehensively reviews molecular mechanisms of the individual steps of postnatal skeletal muscle myogenesis by focusing on signal transduction pathways or networks. Nevertheless, no or only a partial signaling molecules or pathways have been identified in some responses during myogenesis. The elucidation of these unidentified signaling molecules and pathways leads to an extensive understanding of the molecular mechanisms of myogenesis.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
2
|
Hao Y, Xue T, Liu S, Geng S, Shi X, Qian P, He W, Zheng J, Li Y, Lou J, Shi T, Wang G, Wang X, Wang Y, Li Y, Song Y. Loss of CRY2 promotes regenerative myogenesis by enhancing PAX7 expression and satellite cell proliferation. MedComm (Beijing) 2023; 4:e202. [PMID: 36636367 PMCID: PMC9830134 DOI: 10.1002/mco2.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
The regenerative capacity of skeletal muscle is dependent on satellite cells. The circadian clock regulates the maintenance and function of satellite cells. Cryptochrome 2 (CRY2) is a critical component of the circadian clock, and its role in skeletal muscle regeneration remains controversial. Using the skeletal muscle lineage and satellite cell-specific CRY2 knockout mice (CRY2scko), we show that the deletion of CRY2 enhances muscle regeneration. Single myofiber analysis revealed that deletion of CRY2 stimulates the proliferation of myoblasts. The differentiation potential of myoblasts was enhanced by the loss of CRY2 evidenced by increased expression of myosin heavy chain (MyHC) and myotube formation in CRY2-/- cells versus CRY2+/+ cells. Immunostaining revealed that the number of mononucleated paired box protein 7 (PAX7+) cells associated with myotubes formed by CRY2-/- cells was increased compared with CRY2+/+ cells, suggesting that more reserve cells were produced in the absence of CRY2. Loss of CRY2 leads to the activation of the ERK1/2 signaling pathway and ETS1, which binds to the promoter of PAX7 to induce its transcription. CRY2 deficient myoblasts survived better in ischemic muscle. Therefore, CRY2 is essential in regulating skeletal muscle repair.
Collapse
Affiliation(s)
- Yingxue Hao
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Ting Xue
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Song‐Bai Liu
- Suzhou Vocational Health College, Suzhou Key Laboratory of Biotechnology for Laboratory MedicineSuzhouJiangsuP. R. China
| | - Sha Geng
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Xinghong Shi
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Panting Qian
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Wei He
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Jiqing Zheng
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Yanfang Li
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Jing Lou
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Tianze Shi
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Ge Wang
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Xiaoxiao Wang
- Suzhou Vocational Health College, Suzhou Key Laboratory of Biotechnology for Laboratory MedicineSuzhouJiangsuP. R. China
| | - Yanli Wang
- Institutefor Cardiovascular Science and Department of Cardiovascular SurgeryFirst Affiliated Hospital and Medical College of Soochow UniversitySuzhouJiangsuP. R. China
- Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuP. R. China
| | - Yangxin Li
- Institutefor Cardiovascular Science and Department of Cardiovascular SurgeryFirst Affiliated Hospital and Medical College of Soochow UniversitySuzhouJiangsuP. R. China
- Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuP. R. China
| | - Yao‐Hua Song
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| |
Collapse
|
3
|
Zheng J, Lou J, Li Y, Qian P, He W, Hao Y, Xue T, Li Y, Song YH. Satellite cell-specific deletion of Cipc alleviates myopathy in mdx mice. Cell Rep 2022; 39:110939. [PMID: 35705041 DOI: 10.1016/j.celrep.2022.110939] [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: 04/27/2021] [Revised: 04/18/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022] Open
Abstract
Skeletal muscle regeneration relies on satellite cells that can proliferate, differentiate, and form new myofibers upon injury. Emerging evidence suggests that misregulation of satellite cell fate and function influences the severity of Duchenne muscular dystrophy (DMD). The transcription factor Pax7 determines the myogenic identity and maintenance of the pool of satellite cells. The circadian clock regulates satellite cell proliferation and self-renewal. Here, we show that the CLOCK-interacting protein Circadian (CIPC) a negative-feedback regulator of the circadian clock, is up-regulated during myoblast differentiation. Specific deletion of Cipc in satellite cells alleviates myopathy, improves muscle function, and reduces fibrosis in mdx mice. Cipc deficiency leads to activation of the ERK1/2 and JNK1/2 signaling pathways, which activates the transcription factor SP1 to trigger the transcription of Pax7 and MyoD. Therefore, CIPC is a negative regulator of satellite cell function, and loss of Cipc in satellite cells promotes muscle regeneration.
Collapse
Affiliation(s)
- Jiqing Zheng
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Jing Lou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Yanfang Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Panting Qian
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Wei He
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Yingxue Hao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Ting Xue
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China
| | - Yangxin Li
- Department of Cardiovascular Surgery and Institute of Cardiovascular Science, First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, P.R. China.
| | - Yao-Hua Song
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou 215123, P.R. China; National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P.R. China.
| |
Collapse
|
4
|
Yin YH, Zhang XH, Wang XA, Li RH, Zhang YW, Shan XX, You XX, Huang XD, Wu AL, Wang M, Pan XF, Bian C, Jiang WS, Shi Q, Yang JX. Construction of a chromosome-level genome assembly for genome-wide identification of growth-related quantitative trait loci in Sinocyclocheilus grahami (Cypriniformes, Cyprinidae). Zool Res 2021; 42:262-266. [PMID: 33764016 PMCID: PMC8175956 DOI: 10.24272/j.issn.2095-8137.2020.321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The Dianchi golden-line barbel, Sinocyclocheilus grahami (Regan, 1904), is one of the “Four Famous Fishes” of Yunnan Province, China. Given its economic value, this species has been artificially bred successfully since 2007, with a nationally selected breed (“S. grahami, Bayou No. 1”) certified in 2018. For the future utilization of this species, its growth rate, disease resistance, and wild adaptability need to be improved, which could be achieved with the help of molecular marker-assisted selection (MAS). In the current study, we constructed the first chromosome-level genome of S. grahami, assembled 48 pseudo-chromosomes, and obtained a genome assembly of 1.49 Gb. We also performed QTL-seq analysis of S. grahami using the highest and lowest bulks (i.e., largest and smallest size) in both a sibling and random population. We screened two quantitative trait loci (QTLs) (Chr3, 14.9–39.1 Mb and Chr17, 4.1–27.4 Mb) as the major growth-related locations. Several candidate genes (e.g., map2k5, stat1, phf21a, sox6, and smad6) were also identified, with functions related to growth, such as cell differentiation, neuronal development, skeletal muscle development, chondrogenesis, and immunity. These results built a solid foundation for in-depth MAS studies on the growth traits of S. grahami.
Collapse
Affiliation(s)
- Yan-Hui Yin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin-Hui Zhang
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Xiao-Ai Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Rui-Han Li
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Yuan-Wei Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Xin-Xin Shan
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Xin-Xin You
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Xin-Di Huang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - An-Li Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Mo Wang
- Key Laboratory for Conserving Wildlife with Small Populations in Yunnan, Faculty of Biodiversity Conservation, Southwest Forestry University, Kunming, Yunnan 650224, China
| | - Xiao-Fu Pan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Chao Bian
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Wan-Sheng Jiang
- Hunan Engineering Laboratory for Chinese Giant Salamander's Resource Protection and Comprehensive Utilization, and Key Laboratory of Hunan Forest and Chemical Industry Engineering, Jishou University, Zhangjiajie, Hunan 427000, China. E-mail:
| | - Qiong Shi
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Jun-Xing Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China. E-mail:
| |
Collapse
|
5
|
Deolal P, Male G, Mishra K. The challenge of staying in shape: nuclear size matters. Curr Genet 2021; 67:605-612. [PMID: 33779777 DOI: 10.1007/s00294-021-01176-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
Cellular organelles have unique morphology and the organelle size to cell size ratio is regulated. Nucleus is one of the most prominent, usually round in shape, organelle of a eukaryotic cell that occupies 8-10% of cellular volume. The shape and size of nucleus is known to undergo remodeling during processes such as cell growth, division and certain stresses. Regulation of protein and lipid distribution at the nuclear envelope is crucial for preserving the nuclear morphology and size. As size and morphology are interlinked, altering one influences the other. In this perspective, we discuss the relationship between size and shape regulation of the nucleus.
Collapse
Affiliation(s)
- Pallavi Deolal
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Gurranna Male
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Krishnaveni Mishra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
6
|
Kong SH, Yoon JW, Kim JH, Park J, Choi J, Lee JH, Hong AR, Cho NH, Shin CS. Identification of Novel Genetic Variants Related to Trabecular Bone Score in Community-Dwelling Older Adults. Endocrinol Metab (Seoul) 2020; 35:801-810. [PMID: 33232597 PMCID: PMC7803610 DOI: 10.3803/enm.2020.735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/22/2020] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND As the genetic variants of trabecular bone microarchitecture are not well-understood, we performed a genome-wide association study to identify genetic determinants of bone microarchitecture analyzed by trabecular bone score (TBS). METHODS TBS-associated genes were discovered in the Ansung cohort (discovery cohort), a community-based rural cohort in Korea, and then validated in the Gene-Environment Interaction and Phenotype (GENIE) cohort (validation cohort), consisting of subjects who underwent health check-up programs. In the discovery cohort, 2,451 participants were investigated for 1.42 million genotyped and imputed markers. RESULTS In the validation cohort, identified as significant variants were evaluated in 2,733 participants. An intronic variant in iroquois homeobox 3 (IRX3), rs1815994, was significantly associated with TBS in men (P=3.74E-05 in the discovery cohort, P=0.027 in the validation cohort). Another intronic variant in mitogen-activated protein kinase kinase 5 (MAP2K5), rs11630730, was significantly associated with TBS in women (P=3.05E-09 in the discovery cohort, P=0.041 in the validation cohort). Men with the rs1815994 variant and women with the rs11630730 variant had lower TBS and lumbar spine bone mineral density. The detrimental effects of the rs1815994 variant in men and rs11630730 variant in women were also identified in association analysis (β=-0.0281, β=-0.0465, respectively). CONCLUSION In this study, the rs1815994 near IRX3 in men and rs11630730 near MAP2K5 in women were associated with deterioration of the bone microarchitecture. It is the first study to determine the association of genetic variants with TBS. Further studies are needed to confirm our findings and identify additional variants contributing to the trabecular bone microarchitecture.
Collapse
Affiliation(s)
- Sung Hye Kong
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Won Yoon
- Department of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Jung Hee Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - JooYong Park
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jiyeob Choi
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - A Ram Hong
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Nam H. Cho
- Department of Preventive Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
Phillips CA, Reading BJ, Livingston M, Livingston K, Ashwell CM. Evaluation via Supervised Machine Learning of the Broiler Pectoralis Major and Liver Transcriptome in Association With the Muscle Myopathy Wooden Breast. Front Physiol 2020; 11:101. [PMID: 32158398 PMCID: PMC7052112 DOI: 10.3389/fphys.2020.00101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/27/2020] [Indexed: 01/07/2023] Open
Abstract
The muscle myopathy wooden breast (WB) has recently appeared in broiler production and has a negative impact on meat quality. WB is described as hard/firm consistency found within the pectoralis major (PM). In the present study, we use machine learning from our PM and liver transcriptome dataset to capture the complex relationships that are not typically revealed by traditional statistical methods. Gene expression data was evaluated between the PM and liver of birds with WB and those that were normal. Two separate machine learning algorithms were performed to analyze the data set including the sequential minimal optimization (SMO) of support vector machines (SVMs) and Multilayer Perceptron (MLP) Artificial Neural Network (ANN). Machine learning algorithms were compared to identify genes within a gene expression data set of approximately 16,000 genes for both liver and PM, which can be correctly classified from birds with or without WB. The performance of both machine learning algorithms SMO and MLP was determined using percent correct classification during the cross-validations. By evaluating the WB transcriptome datasets by 5× cross-validation using ANNs, the expression of nine genes ranked based on Shannon Entropy (Information Gain) from PM were able to correctly classify if the individual bird was normal or exhibited WB 100% of the time. These top nine genes were all protein coding and potential biomarkers. When PM gene expression data were evaluated between normal birds and those with WB using SVMs they were correctly classified 95% of the time using 450 of the top genes sorted ranked based on Shannon Entropy (Information Gain) as a preprocessing step. When evaluating the 450 attributes that were 95% correctly classified using SVMs through Ingenuity Pathway Analysis (IPA) there was an overlap in top genes identified through MLP. This analysis allowed the identification of critical transcriptional responses for the first time in both liver and muscle during the onset of WB. The information provided has revealed many molecules and pathways making up a complex molecular mechanism involved with the progression of wooden breast and suggests that the etiology of the myopathy is not limited to activity in the muscle alone, but is an altered systemic pathology.
Collapse
Affiliation(s)
- Chelsea A. Phillips
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, United States
| | - Benjamin J. Reading
- Department of Applied Ecology, North Carolina State University, Raleigh, NC, United States
| | - Matthew Livingston
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, United States
| | - Kimberly Livingston
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, United States
| | - Chris M. Ashwell
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
8
|
Akirin1 promotes myoblast differentiation by modulating multiple myoblast differentiation factors. Biosci Rep 2019; 39:BSR20182152. [PMID: 30777932 PMCID: PMC6395299 DOI: 10.1042/bsr20182152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/25/2019] [Accepted: 02/07/2019] [Indexed: 11/17/2022] Open
Abstract
Akirin1 is found to be involved in myoblast differentiation. However, the mechanism by which the Akirin1 gene regulates myoblast differentiation still remains unclear. In the present study, we found that ectopic expression of Akirin1 promoted myoblast differentiation by increasing the expression of myogenic regulatory factor (MRF) 4 (MRF4) and myocyte enhancer factor 2B (MEF2B) mRNA. Additionally, we showed that ectopic Akirin1 induced cell cycle arrest by up-regulating p21 mRNA. To further uncover the mechanism by which Akirin1 promotes myoblast differentiation, we showed that the enhanced Akirin1 increased the mRNA expression of P38α. Importantly, the enhanced MRF4 expression by Akirin1 can be abrogated by treatment of SB203580, a p38 inhibitor. Similarly, we found that enhanced MEF2B expression by Akirin1 can be abrogated by treatment with LY294002, a PI3K inhibitor. Together, our results indicate that Akirin1 promotes myoblast differentiation by acting on the p38 and PI3K pathways and subsequently inducing the expression of myoblast differentiation factors.
Collapse
|
9
|
Insulin-Like Growth Factor Binding Protein-6 Promotes the Differentiation of Placental Mesenchymal Stem Cells into Skeletal Muscle Independent of Insulin-Like Growth Factor Receptor-1 and Insulin Receptor. Stem Cells Int 2019; 2019:9245938. [PMID: 30911300 PMCID: PMC6397983 DOI: 10.1155/2019/9245938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/16/2018] [Accepted: 01/06/2019] [Indexed: 12/15/2022] Open
Abstract
As mesenchymal stem cells (MSCs) are being investigated for regenerative therapies to be used in the clinic, delineating the roles of the IGF system in MSC growth and differentiation, in vitro, is vital in developing these cellular therapies to treat degenerative diseases. Muscle differentiation is a multistep process, starting with commitment to the muscle lineage and ending with the formation of multinucleated fibers. Insulin-like growth factor binding protein-6 (IGFBP-6), relative to other IGFBPs, has high affinity for IGF-2. However, the role of IGFBP-6 in muscle development has not been clearly defined. Our previous studies showed that in vitro extracellular IGFBP-6 increased myogenesis in early stages and could enhance the muscle differentiation process in the absence of IGF-2. In this study, we identified the signal transduction mechanisms of IGFBP-6 on muscle differentiation by placental mesenchymal stem cells (PMSCs). We showed that muscle differentiation required activation of both AKT and MAPK pathways. Interestingly, we demonstrated that IGFBP-6 could compensate for IGF-2 loss and help enhance the muscle differentiation process by triggering predominantly the MAPK pathway independent of activating either IGF-1R or the insulin receptor (IR). These findings indicate the complex interactions between IGFBP-6 and IGFs in PMSC differentiation into the skeletal muscle and that the IGF signaling axis, specifically involving IGFBP-6, is important in muscle differentiation. Moreover, although the major role of IGFBP-6 is IGF-2 inhibition, it is not necessarily the case that IGFBP-6 is the main modulator of IGF-2.
Collapse
|
10
|
Zhang J, Xu X, Liu Y, Zhang L, Odle J, Lin X, Zhu H, Wang X, Liu Y. EPA and DHA Inhibit Myogenesis and Downregulate the Expression of Muscle-related Genes in C2C12 Myoblasts. Genes (Basel) 2019; 10:genes10010064. [PMID: 30669396 PMCID: PMC6356802 DOI: 10.3390/genes10010064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/11/2019] [Indexed: 12/31/2022] Open
Abstract
This study was conducted to elucidate the biological effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on cell proliferation, differentiation and gene expression in C2C12 myoblasts. C2C12 were treated with various concentrations of EPA or DHA under proliferation and differentiation conditions. Cell viability was analyzed using cell counting kit-8 assays (CCK-8). The Edu assays were performed to analyze cell proliferation. To analyze cell differentiation, the expressions of myogenic marker genes were determined at the transcriptional and translational levels by qRT-PCR, immunoblotting and immunofluorescence. Global gene expression patterns were characterized using RNA-sequencing. Phosphorylation levels of ERK and Akt were examined by immunoblotting. Cell viability and proliferation was significantly inhibited after incubation with EPA (50 and 100 μM) or DHA (100 μM). Both EPA and DHA suppressed C2C12 myoblasts differentiation. RNA-sequencing analysis revealed that some muscle-related genes were significantly downregulated following EPA or DHA (50 μM) treatment, including insulin-like growth factor 2 (IGF-2), troponin T3 (Tnnt3), myoglobin (Mb), myosin light chain phosphorylatable fast skeletal muscle (Mylpf) and myosin heavy polypeptide 3 (Myh3). IGF-2 was crucial for the growth and differentiation of skeletal muscle and could activate the PI3K/Akt and the MAPK/ERK cascade. We found that EPA and DHA (50 μM) decreased the phosphorylation levels of ERK1/2 and Akt in C2C12 myoblasts. Thus, this study suggested that EPA and DHA exerted an inhibitory effect on myoblast proliferation and differentiation and downregulated muscle-related genes expression.
Collapse
Affiliation(s)
- Jing Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Xin Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Yan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Lin Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Jack Odle
- Laboratory of Development Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA.
| | - Xi Lin
- Laboratory of Development Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA.
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Xiuying Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
11
|
Different Effects of Insulin-Like Growth Factor-1 and Insulin-Like Growth Factor-2 on Myogenic Differentiation of Human Mesenchymal Stem Cells. Stem Cells Int 2017; 2017:8286248. [PMID: 29387091 PMCID: PMC5745708 DOI: 10.1155/2017/8286248] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/28/2017] [Accepted: 10/10/2017] [Indexed: 11/18/2022] Open
Abstract
Insulin-like growth factors (IGFs) are critical components of the stem cell niche, as they regulate proliferation and differentiation of stem cells into different lineages, including skeletal muscle. We have previously reported that insulin-like growth factor binding protein-6 (IGFBP-6), which has high affinity for IGF-2, alters the differentiation process of placental mesenchymal stem cells (PMSCs) into skeletal muscle. In this study, we determined the roles of IGF-1 and IGF-2 and their interactions with IGFBP-6. We showed that IGF-1 increased IGFBP-6 levels within 24 hours but decreased after 3 days, while IGF-2 maintained higher levels of IGFBP-6 throughout myogenesis. IGF-1 increased IGFBP-6 in the early phase as a requirement for muscle commitment. In contrast, IGF-2 enhanced muscle differentiation as shown by the expression of muscle differentiation markers MyoD, MyoG, and MHC. IGF-1 and IGF-2 had different effects on muscle differentiation with IGF-1 promoting early commitment to muscle and IGF-2 promoting complete muscle differentiation. We also showed that PMSCs acquired increasing capacity to synthesize IGF-2 during muscle differentiation, and the capacity increased as the differentiation progressed suggesting an autocrine and/or paracrine effect. Additionally, we demonstrated that IGFBP-6 could enhance the muscle differentiation process in the absence of IGF-2.
Collapse
|
12
|
Effects of insulin like growth factors on early embryonic chick limb myogenesis. PLoS One 2017; 12:e0185775. [PMID: 28972999 PMCID: PMC5626492 DOI: 10.1371/journal.pone.0185775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/19/2017] [Indexed: 11/19/2022] Open
Abstract
Limb muscles derive from pax3 expressing precursor cells that migrate from the hypaxial somite into the developing limb bud. Once there they begin to differentiate and express muscle determination genes such as MyoD. This process is regulated by a combination of inductive or inhibitory signals including Fgf18, retinoic acid, HGF, Notch and IGFs. IGFs are well known to affect late stages of muscle development and to promote both proliferation and differentiation. We examined their roles in early stage limb bud myogenesis using chicken embryos as an experimental model. Grafting beads soaked in purified recombinant IGF-I, IGF-II or small molecule inhibitors of specific signaling pathways into developing chick embryo limbs showed that both IGF-I and IGF-II induce expression of the early stage myogenic markers pax3 and MyoD as well as myogenin. Their effects on pax3 and MyoD expression were blocked by inhibitors of both the IGF type I receptor (picropodophyllotoxin, PPP) and MEK (U0126). The PI3K inhibitor LY294002 blocked IGF-II, but not IGF-I, induction of pax3 mRNA as well as the IGF-I, but not IGF-II, induction of MyoD mRNA. In addition SU5402, an FGFR/ VEGFR inhibitor, blocked the induction of MyoD by both IGFs but had no effect on pax3 induction, suggesting a role for FGF or VEGF signaling in their induction of MyoD. This was confirmed by in situ hybridization showing that FGF18, a known regulator of MyoD in limb myoblasts, was induced by IGF-I. In addition to their well-known effects on later stages of myogenesis via their induction of myogenin expression, both IGF-I and IGF-II induced pax3 and MyoD expression in developing chick embryos, indicating that they also regulate early stages of myogenesis. The data suggests that the IGFs may have slightly different effects on IGF1R signal transduction via PI3K and that their stimulatory effects on MyoD expression may be indirect, possibly via induction of FGF18 expression.
Collapse
|
13
|
Gilbert AS, Seoane PI, Sephton-Clark P, Bojarczuk A, Hotham R, Giurisato E, Sarhan AR, Hillen A, Velde GV, Gray NS, Alessi DR, Cunningham DL, Tournier C, Johnston SA, May RC. Vomocytosis of live pathogens from macrophages is regulated by the atypical MAP kinase ERK5. SCIENCE ADVANCES 2017; 3:e1700898. [PMID: 28835924 PMCID: PMC5559206 DOI: 10.1126/sciadv.1700898] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/24/2017] [Indexed: 06/07/2023]
Abstract
Vomocytosis, or nonlytic extrusion, is a poorly understood process through which macrophages release live pathogens that they have failed to kill back into the extracellular environment. Vomocytosis is conserved across vertebrates and occurs with a diverse range of pathogens, but to date, the host signaling events that underpin expulsion remain entirely unknown. We use a targeted inhibitor screen to identify the MAP kinase ERK5 as a critical suppressor of vomocytosis. Pharmacological inhibition or genetic manipulation of ERK5 activity significantly raises vomocytosis rates in human macrophages, whereas stimulation of the ERK5 signaling pathway inhibits vomocytosis. Lastly, using a zebrafish model of cryptococcal disease, we show that reducing ERK5 activity in vivo stimulates vomocytosis and results in reduced dissemination of infection. ERK5 therefore represents the first host signaling regulator of vomocytosis to be identified and a potential target for the future development of vomocytosis-modulating therapies.
Collapse
Affiliation(s)
- Andrew S. Gilbert
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Paula I. Seoane
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Poppy Sephton-Clark
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Aleksandra Bojarczuk
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Richard Hotham
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Emanuele Giurisato
- Division of Molecular and Clinical Cancer, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Adil R. Sarhan
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland
| | - Amy Hillen
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven–University of Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven–University of Leuven, Leuven, Belgium
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, SGM 628, Boston, MA 02115, USA
| | - Dario R. Alessi
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland
| | - Debbie L. Cunningham
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Cathy Tournier
- Division of Molecular and Clinical Cancer, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Simon A. Johnston
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Robin C. May
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
14
|
Extracellular Signal-Regulated Kinase 5 is Required for Low-Concentration H 2O 2-Induced Angiogenesis of Human Umbilical Vein Endothelial Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6895730. [PMID: 28540300 PMCID: PMC5429924 DOI: 10.1155/2017/6895730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/22/2017] [Accepted: 03/09/2017] [Indexed: 01/28/2023]
Abstract
Background. The aim of this study was to assess the effects of low concentrations of H2O2 on angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro and explore the underlying mechanisms. Methods. HUVECs were cultured and stimulated with different concentrations of H2O2. Flow cytometric analysis was used to select an optimal concentration of H2O2 for the following experiments. Cell proliferation, migration, and tubule formation were evaluated by Cell Counting Kit-8 (CCK-8) assays, scratch wound assays, and Matrigel tubule formation assays, respectively. For gain and loss of function studies, constitutively active MEK5 (CA-MEK5) and ERK5 shRNA lentiviruses were used to activate or knock down extracellular signal-regulated kinase 5 (ERK5). Results. We found that low concentrations of H2O2 promoted HUVECs proliferation, migration, and tubule formation. ERK5 in HUVECs was significantly activated by H2O2. Enhanced ERK5 activity significantly amplified the proangiogenic effects of H2O2; in contrast, ERK5 knock-down abrogated the effects of H2O2. Conclusions. Our results confirmed that low concentrations of H2O2 promoted HUVECs angiogenesis in vitro, and ERK5 is an essential mediator of this process. Therefore, ERK5 may be a potential therapeutic target for promoting angiogenesis and improving graft survival.
Collapse
|
15
|
Chen TH, Chen CY, Wen HC, Chang CC, Wang HD, Chuu CP, Chang CH. YAP promotes myogenic differentiation via the MEK5-ERK5 pathway. FASEB J 2017; 31:2963-2972. [PMID: 28356344 DOI: 10.1096/fj.201601090r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/13/2017] [Indexed: 01/26/2023]
Abstract
Yes-associated protein (YAP) is a transcriptional coactivator in the Hippo pathway that regulates cell proliferation, differentiation, and apoptosis. The MEK5/ERK5 MAPK cascade is essential for the early step of myogenesis. In this study, we generated C2C12 stable cell lines that expressed YAP (C2C12-YAP cells) and found that ERK5 and MEK5 were activated in C2C12-YAP cells compared with control C2C12 (C2C12-vector) cells. C2C12-YAP stable cells also differentiated into myotubes better than C2C12-vector cells, and expressed elevated levels of myogenin, a transcription factor that regulates myogenesis, as well as elevated levels of myosin heavy chain, a skeletal muscle marker. Western blot analysis revealed that Src and c-Abl (Abelson murine leukemia viral oncogene homolog 1) activation were enhanced in C2C12-YAP cells. Conversely, treatment of inhibitors of c-Abl, Src, or MEK5 inhibited activation of MEK5 and ERK5 and myogenesis of C2C12 myoblasts. Specific interactions between YAP and proteins in the ERK5 pathway, such as MEK kinase 3 (MEKK3) and ERK5, were illustrated by coimmunoprecipitation experiments. MEKK3 contains the PPGY motif (aa 178-181), which may interact with YAP. Site-directed mutagenesis experiments revealed that expression of MEKK3 Y181F mutant inhibited MEK5/ERK5 activation and myogenic differentiation. These results suggest that YAP promotes muscle differentiation by activating the Abl/Src/MEKK3/MEK5/ERK5 kinase cascade.-Chen, T.-H., Chen, C.-Y., Wen, H.-C., Chang, C.-C., Wang, H.-D., Chuu, C.-P., Chang, C.-H. YAP promotes myogenic differentiation via the MEK5-ERK5 pathway.
Collapse
Affiliation(s)
- Ting-Huan Chen
- Department of Life Science, National Tsing Hua University, Hsin-Chu, Taiwan, China.,Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Chen-Yu Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Hui-Chin Wen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Chia-Chu Chang
- Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan, China
| | - Horng-Dar Wang
- Department of Life Science, National Tsing Hua University, Hsin-Chu, Taiwan, China
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China;
| | - Chung-Ho Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China; .,Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan, China
| |
Collapse
|
16
|
Bortell N, Basova L, Semenova S, Fox HS, Ravasi T, Marcondes MCG. Astrocyte-specific overexpressed gene signatures in response to methamphetamine exposure in vitro. J Neuroinflammation 2017; 14:49. [PMID: 28279172 PMCID: PMC5345234 DOI: 10.1186/s12974-017-0825-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/27/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Astrocyte activation is one of the earliest findings in the brain of methamphetamine (Meth) abusers. Our goal in this study was to identify the characteristics of the astrocytic acute response to the drug, which may be critical in pathogenic outcomes secondary to the use. METHODS We developed an integrated analysis of gene expression data to study the acute gene changes caused by the direct exposure to Meth treatment of astrocytes in vitro, and to better understand how astrocytes respond, what are the early molecular markers associated with this response. We examined the literature in search of similar changes in gene signatures that are found in central nervous system disorders. RESULTS We identified overexpressed gene networks represented by genes of an inflammatory and immune nature and that are implicated in neuroactive ligand-receptor interactions. The overexpressed networks are linked to molecules that were highly upregulated in astrocytes by all doses of methamphetamine tested and that could play a role in the central nervous system. The strongest overexpressed signatures were the upregulation of MAP2K5, GPR65, and CXCL5, and the gene networks individually associated with these molecules. Pathway analysis revealed that these networks are involved both in neuroprotection and in neuropathology. We have validated several targets associated to these genes. CONCLUSIONS Gene signatures for the astrocytic response to Meth were identified among the upregulated gene pool, using an in vitro system. The identified markers may participate in dysfunctions of the central nervous system but could also provide acute protection to the drug exposure. Further in vivo studies are necessary to establish the role of these gene networks in drug abuse pathogenesis.
Collapse
Affiliation(s)
- Nikki Bortell
- Cellular and Molecular Neurosciences Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.,Anschutz Medical Campus, University of Colorado, Denver, CO, USA
| | - Liana Basova
- Cellular and Molecular Neurosciences Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Svetlana Semenova
- Department of Psychiatry, University of California San Diego, San Diego, CA, 92093, USA
| | - Howard S Fox
- Department of Experimental Pharmacology, University of Nebraska Medical School, Omaha, NE, 68198, USA
| | - Timothy Ravasi
- KAUST Environmental Epigenetic Program (KEEP), Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.,Department of Medicine, Division of Genetic, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Maria Cecilia G Marcondes
- Cellular and Molecular Neurosciences Department, The Scripps Research Institute, La Jolla, CA, 92037, USA. .,Present address: San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100 - San Diego, San Diego, CA, 92121, USA.
| |
Collapse
|
17
|
Gu N, Ge K, Hao C, Ji Y, Li H, Guo Y. Neuregulin1β Effects on Brain Tissue via ERK5-Dependent MAPK Pathway in a Rat Model of Cerebral Ischemia-Reperfusion Injury. J Mol Neurosci 2017; 61:607-616. [PMID: 28265860 DOI: 10.1007/s12031-017-0902-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/16/2017] [Indexed: 11/29/2022]
Abstract
Neuregulin1β (NRG1β), a member of the excitomotor of tyrosine kinase receptor (erbB) family, was recently shown to play a neuroprotective role in cerebral ischemia-reperfusion injury. The present study analyzed the effects and its possible signaling pathway of NRG1β on brain tissues after cerebral ischemia-reperfusion injury. A focal cerebral ischemic model was established by inserting a monofilament thread to achieve middle cerebral artery occlusion, followed by an NRG1β injection via the internal carotid artery. NRG1β injection resulted in significantly improved neurobehavioral activity according to the modified neurological severity score test. Tetrazolium chloridestaining revealed a smaller cerebral infarction volume; hematoxylin-eosin staining and transmission electron microscopy showed significantly alleviated neurodegeneration in the middle cerebral artery occlusion rats. Moreover, expression of phosphorylated MEK5, phosphorylated ERK5, and phosphorylated MEK2C increased after NRG1β treatment, and the neuroprotective effect of NRG1β was attenuated by an injection of the MEK5 inhibitor, BIX02189. Results from the present study demonstrate that NRG1β provides neuroprotection following cerebral ischemia-reperfusion injury via the ERK5-dependent MAPK pathway.
Collapse
Affiliation(s)
- Ning Gu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Institute of Integrated Medicine, Qingdao University Medical College, Qingdao, Shandong, China
| | - Keli Ge
- Institute of Integrated Medicine, Qingdao University Medical College, Qingdao, Shandong, China
| | - Cui Hao
- Institute of Cerebrovascular Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yaqing Ji
- Institute of Integrated Medicine, Qingdao University Medical College, Qingdao, Shandong, China
| | - Hongyun Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Yunliang Guo
- Institute of Cerebrovascular Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
18
|
Edea Z, Kim KS. A whole genomic scan to detect selection signatures between Berkshire and Korean native pig breeds. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2014; 56:23. [PMID: 26290712 PMCID: PMC4540274 DOI: 10.1186/2055-0391-56-23] [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: 06/29/2014] [Accepted: 10/26/2014] [Indexed: 11/11/2022]
Abstract
Background Scanning of the genome for selection signatures between breeds may play important role in understanding the underlie causes for observable phenotypic variations. The discovery of high density single nucleotide polymorphisms (SNPs) provide a useful starting point to perform genome–wide scan in pig populations in order to identify loci/candidate genes underlie phenotypic variation in pig breeds and facilitate genetic improvement programs. However, prior to this study genomic region under selection in commercially selected Berkshire and Korean native pig breeds has never been detected using high density SNP markers. To this end, we have genotyped 45 animals using Porcine SNP60 chip to detect selection signatures in the genome of the two breeds by using the FST approach. Results In the comparison of Berkshire and KNP breeds using the FDIST approach, a total of 1108 outlier loci (3.48%) were significantly different from zero at 99% confidence level with 870 of the outlier SNPs displaying high level of genetic differentiation (FST ≥0.490). The identified candidate genes were involved in a wide array of biological processes and molecular functions. Results revealed that 19 candidate genes were enriched in phosphate metabolism (GO: 0006796; ADCK1, ACYP1, CAMK2D, CDK13, CDK13, ERN1, GALK2, INPP1; MAK, MAP2K5, MAP3K1, MAPK14, P14KB, PIK3C3, PRKC1, PTPRK, RNASEL, THBS1, BRAF, VRK1). We have identified a set of candidate genes under selection and have known to be involved in growth, size and pork quality (CART, AGL, CF7L2, MAP2K5, DLK1, GLI3, CA3 and MC3R), ear morphology and size (HMGA2 and SOX5) stress response (ATF2, MSRB3, TMTC3 and SCAF8) and immune response ( HCST and RYR1). Conclusions Some of the genes may be used to facilitate genetic improvement programs. Our results also provide insights for better understanding of the process and influence of breed development on the pattern of genetic variations. Electronic supplementary material The online version of this article (doi:10.1186/2055-0391-56-23) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zewdu Edea
- Department of Animal Science, Chungbuk National University, Cheongju, 361-763 Korea
| | - Kwan-Suk Kim
- Department of Animal Science, Chungbuk National University, Cheongju, 361-763 Korea
| |
Collapse
|
19
|
Jiang L, Huang M, Wang L, Fan X, Wang P, Wang D, Fu X, Wang J. Overexpression of MEKK2 is associated with colorectal carcinogenesis. Oncol Lett 2013; 6:1333-1337. [PMID: 24179519 PMCID: PMC3813537 DOI: 10.3892/ol.2013.1553] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/05/2013] [Indexed: 01/04/2023] Open
Abstract
Mitogen-activated protein kinase kinase kinase 2 (MEKK2) is an important upstream mediator of the extracellular signal-regulated kinase 5 signaling cascade that is essential for a number of cellular functions, including mitogenesis, differentiation and oncogenic transformation. Using western blotting to examine MEKK2 expression in 16 cases of primary colorectal cancer (CRC) lesions with paired normal mucosa, it was identified that MEKK2 is highly expressed in CRC lesions compared with that of the normal mucosa. Immunohistochemistry of 24 normal mucosa, 24 adenoma and 96 adenocarcinoma colorectal specimens indicated that the expression of MEKK2 was significantly increased in the adenoma and carcinoma specimens compared with that of the normal mucosa cases (P<0.0001 for both). However, no significant differences were detected in MEKK2 expression between the carcinoma and adenoma specimens (P=0.85). Similarly, no correlations were identified between MEKK2 expression and clinicopathological features, including gender, age, body mass index, histological differentiation, depth of invasion, lymph node metastasis, UICC stage and K-ras mutations (P>0.05). The present study demonstrated that MEKK2 functions as a promotive factor in CRC.
Collapse
Affiliation(s)
- Li Jiang
- Department of Gastrointestinal Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Differential regulation of IGF-I and IGF-II gene expression in skeletal muscle cells. Mol Cell Biochem 2012; 373:107-13. [PMID: 23054195 DOI: 10.1007/s11010-012-1479-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 09/26/2012] [Indexed: 12/22/2022]
Abstract
Insulin-like growth factor (IGF)-I and IGF-II play major roles in the regulation of skeletal muscle growth and differentiation, and both are locally expressed in muscle cells. Recent studies have demonstrated that IGF-II up-regulates its own gene expression during myogenesis and this auto-regulatory loop is critical for muscle differentiation. How local IGF-I is regulated in this process is unclear. Here, we report that while IGF-II up-regulated its own gene expression, it suppressed IGF-I gene expression during myogenesis. These opposite effects of IGF-II on IGF-I and IGF-II genes expression were time dependent and dose dependent. It has been shown that IGFs activate the PI3K-Akt-mTOR, p38 MAPK, and Erk1/2 MAPK pathways. In myoblasts, we examined their role(s) in mediating the opposite effects of IGF-II. Our results showed that both the PI3K-Akt-mTOR and p38 MAPK pathways played critical roles in increasing IGF-II mRNA expression. In contrast, mTOR was required for down-regulating the IGF-I gene expression by IGF-II. In addition, Akt, Erk1/2 MAPK, and p38 MAPK pathways were also involved in the regulation of basal levels of IGF-I and IGF-II genes during myogenesis. These findings reveal a previously unrecognized negative feedback mechanism and extend our knowledge of IGF-I and IGF-II gene expression and regulation during myogenesis.
Collapse
|
21
|
Rask-Andersen M, Jacobsson JA, Moschonis G, Ek AE, Chrousos GP, Marcus C, Manios Y, Fredriksson R, Schiöth HB. The MAP2K5-linked SNP rs2241423 is associated with BMI and obesity in two cohorts of Swedish and Greek children. BMC MEDICAL GENETICS 2012; 13:36. [PMID: 22594783 PMCID: PMC3459804 DOI: 10.1186/1471-2350-13-36] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/04/2012] [Indexed: 11/10/2022]
Abstract
Background Recent genome-wide association studies have identified a single nucleotide polymorphism within the last intron of MAP2K5 associated with a higher body mass index (BMI) in adults. MAP2K5 is a component of the MAPK-family intracellular signaling pathways, responding to extracellular growth factors such as brain derived neurotrophic factor (BDNF) and nerve growth factor (NGF). In this study, we examined the association of this variant in two cohorts of children from Sweden and Greece. Methods We examine the association of rs2241423 to BMI in a cohort of 474 Swedish children admitted for treatment of childhood obesity and 519 children matched for gender, ethnicity and socioeconomic background from the Stockholm area, as well as a cross-sectional cohort of 2308 Greek school children (Healthy Growth Study). Children were genotyped using a predesigned TaqMan polymorphism assay. Logistic regression was used to test for an association of rs2241423 to obesity in the cohort of Swedish children. Linear regression was used to test for an association of rs2241423 to BMI z-score and phenotypic measurements of body adiposity in the cohort of Greek children. Models were adjusted for age and gender. In the cohort of Greek children the model was also adjusted for stage of pubertal development. Results The minor allele of rs2241423, allele A, was associated with a protective effect against obesity in the cohort of Swedish children (p = 0.029, OR = 0.79 (95% CI: 0.64–0.98)), and with a lower BMI z-score in the cohort of Greek children (p = 0.028, β = −0.092). No association to phenotypic measurements of body fat distribution could be observed in our study. Conclusions rs2241423 was associated with BMI and obesity in two independent European cohorts suggesting a role for MAP2K5 in early weight regulation.
Collapse
Affiliation(s)
- Mathias Rask-Andersen
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Uppsala SE 75124, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Insulin-like growth factor-II (IGF-II) affects many aspects of cellular function through its ability to activate several different receptors and, consequently, numerous intracellular signalling molecules. Thus, IGF-II is a key regulator of normal foetal development and growth. However, abnormalities in IGF-II function are associated with cardiovascular disease and cancer. Here, we review the cellular mechanisms by which IGF-II's physiological and pathophysiological actions are exerted by discussing the involvement of the type 1 and type 2 IGF receptors (IGF1R and IGF2R), the insulin receptor and the downstream MAP kinase, PI-3 kinase and G-protein-coupled signalling pathways in mediating IGF-II stimulated cellular proliferation, survival, differentiation and migration.
Collapse
Affiliation(s)
- Lynda K Harris
- Maternal and Fetal Health Research Centre, University of Manchester, UK
| | | |
Collapse
|
23
|
The myogenic kinome: protein kinases critical to mammalian skeletal myogenesis. Skelet Muscle 2011; 1:29. [PMID: 21902831 PMCID: PMC3180440 DOI: 10.1186/2044-5040-1-29] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 09/08/2011] [Indexed: 12/13/2022] Open
Abstract
Myogenesis is a complex and tightly regulated process, the end result of which is the formation of a multinucleated myofibre with contractile capability. Typically, this process is described as being regulated by a coordinated transcriptional hierarchy. However, like any cellular process, myogenesis is also controlled by members of the protein kinase family, which transmit and execute signals initiated by promyogenic stimuli. In this review, we describe the various kinases involved in mammalian skeletal myogenesis: which step of myogenesis a particular kinase regulates, how it is activated (if known) and what its downstream effects are. We present a scheme of protein kinase activity, similar to that which exists for the myogenic transcription factors, to better clarify the complex signalling that underlies muscle development.
Collapse
|
24
|
Su C, Underwood W, Rybalchenko N, Singh M. ERK1/2 and ERK5 have distinct roles in the regulation of brain-derived neurotrophic factor expression. J Neurosci Res 2011; 89:1542-50. [DOI: 10.1002/jnr.22683] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 04/05/2011] [Accepted: 04/06/2011] [Indexed: 11/05/2022]
|
25
|
Paxton CW, Cosgrove RA, Drozd AC, Wiggins EL, Woodhouse S, Watson RA, Spence HJ, Ozanne BW, Pell JM. BTB-Kelch protein Krp1 regulates proliferation and differentiation of myoblasts. Am J Physiol Cell Physiol 2011; 300:C1345-55. [PMID: 21368295 DOI: 10.1152/ajpcell.00321.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The BTB-Kelch protein Krp1 is highly and specifically expressed in skeletal muscle, where it is proposed to have a role in myofibril formation. We observed significant upregulation of Krp1 in C2 cells early in myoblast differentiation, well before myofibrillogenesis. Krp1 has a role in cytoskeletal organization and cell motility; since myoblast migration and elongation/alignment are important events in early myogenesis, we hypothesized that Krp1 is involved with earlier regulation of differentiation. Krp1 protein levels were detectable by 24 h after induction of differentiation in C2 cells and were significantly upregulated by 48 h, i.e., following the onset myogenin expression and preceding myosin heavy chain (MHC) upregulation. Upregulation of Krp1 required a myogenic stimulus as signaling derived from increased myoblast cell density was insufficient to activate Krp1 expression. Examination of putative Krp1 proximal promoter regions revealed consensus E box elements associated with myogenic basic helix-loop-helix binding. The activity of a luciferase promoter-reporter construct encompassing this 2,000-bp region increased in differentiating C2 myoblasts and in C2 cells transfected with myogenin and/or MyoD. Knockdown of Krp1 via short hairpin RNA resulted in increased C2 cell number and proliferation rate as assessed by bromodeoxyuridine incorporation, whereas overexpression of Krp1-myc had the opposite effect; apoptosis was unchanged. No effects of changed Krp1 protein levels on cell migration were observed, either by scratch wound assay or live cell imaging. Paradoxically, both knockdown and overexpression of Krp1 inhibited myoblast differentiation assessed by expression of myogenin, MEF2C, MHC, and cell fusion.
Collapse
Affiliation(s)
- Camille W Paxton
- Laboratory of Molecular Signalling, The Babraham Institute, Cambridge, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ciemerych MA, Archacka K, Grabowska I, Przewoźniak M. Cell cycle regulation during proliferation and differentiation of mammalian muscle precursor cells. Results Probl Cell Differ 2011; 53:473-527. [PMID: 21630157 DOI: 10.1007/978-3-642-19065-0_20] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Proliferation and differentiation of muscle precursor cells are intensively studied not only in the developing mouse embryo but also using models of skeletal muscle regeneration or analyzing in vitro cultured cells. These analyses allowed to show the universality of the cell cycle regulation and also uncovered tissue-specific interplay between major cell cycle regulators and factors crucial for the myogenic differentiation. Examination of the events accompanying proliferation and differentiation leading to the formation of functional skeletal muscle fibers allows understanding the molecular basis not only of myogenesis but also of skeletal muscle regeneration. This chapter presents the basis of the cell cycle regulation in proliferating and differentiating muscle precursor cells during development and after muscle injury. It focuses at major cell cycle regulators, myogenic factors, and extracellular environment impacting on the skeletal muscle.
Collapse
Affiliation(s)
- Maria A Ciemerych
- Department of Cytology, Institute of Zoology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland.
| | | | | | | |
Collapse
|
27
|
Brzoska E, Ciemerych MA, Przewozniak M, Zimowska M. Regulation of Muscle Stem Cells Activation. STEM CELL REGULATORS 2011; 87:239-76. [DOI: 10.1016/b978-0-12-386015-6.00031-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
28
|
Roberts OL, Holmes K, Müller J, Cross DAE, Cross MJ. ERK5 is required for VEGF-mediated survival and tubular morphogenesis of primary human microvascular endothelial cells. J Cell Sci 2010; 123:3189-200. [DOI: 10.1242/jcs.072801] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is activated in response to environmental stress and growth factors. Gene ablation of Erk5 in mice is embryonically lethal as a result of disruption of cardiovascular development and vascular integrity. We investigated vascular endothelial growth factor (VEGF)-mediated ERK5 activation in primary human dermal microvascular endothelial cells (HDMECs) undergoing proliferation on a gelatin matrix, and tubular morphogenesis within a collagen gel matrix. VEGF induced sustained ERK5 activation on both matrices. However, manipulation of ERK5 activity by siRNA-mediated gene silencing disrupted tubular morphogenesis without impacting proliferation. Overexpression of constitutively active MEK5 and ERK5 stimulated tubular morphogenesis in the absence of VEGF. Analysis of intracellular signalling revealed that ERK5 regulated AKT phosphorylation. On a collagen gel, ERK5 regulated VEGF-mediated phosphorylation of the pro-apoptotic protein BAD and increased expression of the anti-apoptotic protein BCL2, resulting in decreased caspase-3 activity and apoptosis suppression. Our findings suggest that ERK5 is required for AKT phosphorylation and cell survival and is crucial for endothelial cell differentiation in response to VEGF.
Collapse
Affiliation(s)
- Owain Llŷr Roberts
- NWCRF Institute, School of Biological Sciences, College of Natural Sciences, Bangor University, Bangor, LL57 2UW, UK
| | - Katherine Holmes
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Jürgen Müller
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Darren A. E. Cross
- AstraZeneca, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Michael J. Cross
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| |
Collapse
|
29
|
Yao Z, Yoon S, Kalie E, Raviv Z, Seger R. Calcium regulation of EGF-induced ERK5 activation: role of Lad1-MEKK2 interaction. PLoS One 2010; 5:e12627. [PMID: 20830310 PMCID: PMC2935384 DOI: 10.1371/journal.pone.0012627] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 08/13/2010] [Indexed: 12/17/2022] Open
Abstract
The ERK5 cascade is a MAPK pathway that transmits both mitogenic and stress signals, yet its mechanism of activation is not fully understood. Using intracellular calcium modifiers, we found that ERK5 activation by EGF is inhibited both by the depletion and elevation of intracellular calcium levels. This calcium effect was found to occur upstream of MEKK2, which is the MAP3K of the ERK5 cascade. Co-immunoprecipitation revealed that EGF increases MEKK2 binding to the adaptor protein Lad1, and this interaction was reduced by the intracellular calcium modifiers, indicating that a proper calcium concentration is required for the interactions and transmission of EGF signals to ERK5. In vitro binding assays revealed that the proper calcium concentration is required for a direct binding of MEKK2 to Lad1. The binding of these proteins is not affected by c-Src-mediated phosphorylation on Lad1, but slightly affects the Tyr phosphorylation of MEKK2, suggesting that the interaction with Lad1 is necessary for full Tyr phosphorylation of MEKK2. In addition, we found that changes in calcium levels affect the EGF-induced nuclear translocation of MEKK2 and thereby its effect on the nuclear ERK5 activity. Taken together, these findings suggest that calcium is required for EGF-induced ERK5 activation, and this effect is probably mediated by securing proper interaction of MEKK2 with the upstream adaptor protein Lad1.
Collapse
Affiliation(s)
- Zhong Yao
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Seunghee Yoon
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Kalie
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Raviv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
30
|
Lovett FA, Cosgrove RA, Gonzalez I, Pell JM. Essential role for p38alpha MAPK but not p38gamma MAPK in Igf2 expression and myoblast differentiation. Endocrinology 2010; 151:4368-80. [PMID: 20610565 DOI: 10.1210/en.2010-0209] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The muscle satellite cell is established as the major stem cell contributing to fiber growth and repair. p38 MAPK signaling is essential for myoblast differentiation and in particular for up-regulation of promyogenic Igf2 expression. p38 exists as four isoforms (alpha, beta, gamma, and delta), of which p38gamma is uniquely abundant in muscle. The aim of this study was to characterize p38 isoform expression and importance (using shRNA knockdown; demonstrated via both reduced protein and kinase activities) during myoblast differentiation. p38alpha and -gamma mRNA levels were most abundant in differentiating C2 cells with low/negligible contributions from p38beta and -delta, respectively. Increased phosphorylation of p38alpha and -gamma occurred during differentiation but via different mechanisms: p38alpha protein levels remained constant, whereas total p38gamma levels increased. Following shRNA knockdown of p38alpha, myoblast differentiation was dramatically inhibited [reduced myosin heavy chain (MHC), myogenin, pAkt protein levels]; significantly, Igf2 mRNA levels and promoter-reporter activities decreased. In contrast, knockdown of p38gamma induced a transient increase in both myogenin and MHC protein levels with no effect on Igf2 mRNA levels or promoter-reporter activity. Knockdown of p38alpha/beta markedly increased but that of p38gamma decreased caspase 3 activity, suggesting opposite actions on apoptosis. p38gamma was initially proposed to have a promyogenic function; however, p38gamma overexpression could not rescue reduced myoblast differentiation following p38alpha/beta inhibition. Therefore, p38alpha is essential for myoblast differentiation, and part of its action is to convert signals that indicate cell density into promyogenic gene expression in the form of the key peptide, IGF-II; p38gamma has a minor, yet opposing antimyogenic, function.
Collapse
Affiliation(s)
- Fiona A Lovett
- The Babraham Institute, Cambridge CB22 3AT, United Kingdom
| | | | | | | |
Collapse
|