1
|
Hashemolhosseini S, Gessler L. Crosstalk among canonical Wnt and Hippo pathway members in skeletal muscle and at the neuromuscular junction. Neural Regen Res 2025; 20:2464-2479. [PMID: 39248171 DOI: 10.4103/nrr.nrr-d-24-00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Skeletal muscles are essential for locomotion, posture, and metabolic regulation. To understand physiological processes, exercise adaptation, and muscle-related disorders, it is critical to understand the molecular pathways that underlie skeletal muscle function. The process of muscle contraction, orchestrated by a complex interplay of molecular events, is at the core of skeletal muscle function. Muscle contraction is initiated by an action potential and neuromuscular transmission requiring a neuromuscular junction. Within muscle fibers, calcium ions play a critical role in mediating the interaction between actin and myosin filaments that generate force. Regulation of calcium release from the sarcoplasmic reticulum plays a key role in excitation-contraction coupling. The development and growth of skeletal muscle are regulated by a network of molecular pathways collectively known as myogenesis. Myogenic regulators coordinate the differentiation of myoblasts into mature muscle fibers. Signaling pathways regulate muscle protein synthesis and hypertrophy in response to mechanical stimuli and nutrient availability. Several muscle-related diseases, including congenital myasthenic disorders, sarcopenia, muscular dystrophies, and metabolic myopathies, are underpinned by dysregulated molecular pathways in skeletal muscle. Therapeutic interventions aimed at preserving muscle mass and function, enhancing regeneration, and improving metabolic health hold promise by targeting specific molecular pathways. Other molecular signaling pathways in skeletal muscle include the canonical Wnt signaling pathway, a critical regulator of myogenesis, muscle regeneration, and metabolic function, and the Hippo signaling pathway. In recent years, more details have been uncovered about the role of these two pathways during myogenesis and in developing and adult skeletal muscle fibers, and at the neuromuscular junction. In fact, research in the last few years now suggests that these two signaling pathways are interconnected and that they jointly control physiological and pathophysiological processes in muscle fibers. In this review, we will summarize and discuss the data on these two pathways, focusing on their concerted action next to their contribution to skeletal muscle biology. However, an in-depth discussion of the non-canonical Wnt pathway, the fibro/adipogenic precursors, or the mechanosensory aspects of these pathways is not the focus of this review.
Collapse
Affiliation(s)
- Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
2
|
Desmarquet-Trin Dinh C, Manceau M. Structure, function and formation of the amniote skin pattern. Dev Biol 2024; 517:203-216. [PMID: 39326486 DOI: 10.1016/j.ydbio.2024.09.011] [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: 01/17/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
From feather and hair dotted arrays to pigmented stripes and spots, the spatial distribution of skin appendages and colouration often forms visible ornaments crucial for fitness in the coat of birds and mammals. These geometrical motifs are extremely diverse in nature. Yet, phenotypic surveys evidenced common themes in variation: the orientation, appendage-specificity or pigmentation of a given region may be conserved across groups or species. Here, we review naturalist observations of natural variation in the anatomy and ecological function of the skin pattern in amniotes. We then describe several decades of genetics, mathematical modelling and experimental embryology work aiming at understanding the molecular and morphogenetic mechanisms responsible for pattern formation. We discuss how these studies provided evidence that the morphological trends and differences representative of the phenotypic landscape of skin patterns in wild amniote species is rooted in the mechanisms controlling the production of distinct compartments in the embryonic skin.
Collapse
Affiliation(s)
| | - Marie Manceau
- Centre for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, France.
| |
Collapse
|
3
|
Tombarkiewicz B, Trzeciak K, Lis MW, Makulska J, Pawlak K, Bojarski B. The effect of methionine and folic acid administered in ovo on the blood biochemical parameters of chickens (Gallus gallus domesticus). Poult Sci 2024; 103:103731. [PMID: 38669817 PMCID: PMC11066548 DOI: 10.1016/j.psj.2024.103731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/26/2024] [Accepted: 03/31/2024] [Indexed: 04/28/2024] Open
Abstract
Methionine is one of the most frequently supplemented amino acids in raising of poultry. However, an overdose of methionine can cause hyperhomocysteinemia. Folic acid, taking part in the process of homocysteine remethylation, is a factor affecting the reduction of the concentration of this amino acid. The study was carried out in 2 stages. The experiment of step I was to investigate the effect of methionine and/or folic acid administration in ovo in the early stage of embryogenesis (E4), and the experiment of the second stage - in the late stage of embryogenesis (E17) on the following biochemical parameters of chicken blood: glucose concentration in whole blood and concentration of homocysteine and uric acid in plasma of domestic chickens (Gallus gallus domesticus). Our results confirm that methionine supplementation may increase the concentration of uric acid and homocysteine. Moreover, we demonstrated that folic acid administered during embryogenesis decreased homocysteine concentration, also in groups simultaneously supplemented with methionine, especially in the initial stage of postnatal life of the bird.
Collapse
Affiliation(s)
- Barbara Tombarkiewicz
- Department of Zoology and Animal Welfare, Faculty of Animal Science, University of Agriculture in Krakow, Krakow 30-059, Poland
| | - Karolina Trzeciak
- Department of Zoology and Animal Welfare, Faculty of Animal Science, University of Agriculture in Krakow, Krakow 30-059, Poland
| | - Marcin W Lis
- Department of Zoology and Animal Welfare, Faculty of Animal Science, University of Agriculture in Krakow, Krakow 30-059, Poland
| | - Joanna Makulska
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Science, University of Agriculture in Krakow, Krakow 30-059, Poland
| | - Krzysztof Pawlak
- Department of Zoology and Animal Welfare, Faculty of Animal Science, University of Agriculture in Krakow, Krakow 30-059, Poland
| | - Bartosz Bojarski
- Department of Animal Physiology, Institute of Biology, Pomeranian University in Słupsk, Słupsk 76-200, Poland.
| |
Collapse
|
4
|
Rauen KA, Tidyman WE. RASopathies - what they reveal about RAS/MAPK signaling in skeletal muscle development. Dis Model Mech 2024; 17:dmm050609. [PMID: 38847227 PMCID: PMC11179721 DOI: 10.1242/dmm.050609] [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] [Indexed: 06/12/2024] Open
Abstract
RASopathies are rare developmental genetic syndromes caused by germline pathogenic variants in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Although the incidence of each RASopathy syndrome is rare, collectively, they represent one of the largest groups of multiple congenital anomaly syndromes and have severe developmental consequences. Here, we review our understanding of how RAS/MAPK dysregulation in RASopathies impacts skeletal muscle development and the importance of RAS/MAPK pathway regulation for embryonic myogenesis. We also discuss the complex interactions of this pathway with other intracellular signaling pathways in the regulation of skeletal muscle development and growth, and the opportunities that RASopathy animal models provide for exploring the use of pathway inhibitors, typically used for cancer treatment, to correct the unique skeletal myopathy caused by the dysregulation of this pathway.
Collapse
Affiliation(s)
- Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, CA, 95817, USA
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| | - William E Tidyman
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
5
|
Sahoo S, Kumar KP, Narayan RK. Genitofemoral Nerve Variation: An Attempt to Explain the Embryological Basis via a Case Report. Cureus 2024; 16:e61763. [PMID: 38975486 PMCID: PMC11226732 DOI: 10.7759/cureus.61763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
The genitofemoral nerve (GFN) presents with a variable course in nearly half of the population. This variation can be seen in its availability, course, and branching. Here, a notable case during a cadaveric dissection revealed an unusually high bifurcation of the GFN on the left side, contrasting with the typical bifurcation observed on the right. This divergence was highlighted using colored markers to aid educational visualization, facilitating a comprehensive learning experience about the nerve's variability and its functional implications, such as the cremasteric reflex. Embryologically, these variations stem from the migratory paths of myotomes during development, influenced by extrinsic signals and growth factors. Despite the high incidence of anatomical variability, the muscular structure remains consistent, suggesting that the nerve's formation is more susceptible to developmental shifts than the muscles it innervates. Clinically, understanding GFN variations is crucial due to the nerve's involvement in conditions like genitofemoral neuropathy, which can arise from surgical procedures. Accurate knowledge of these variations aids in precise diagnostic and therapeutic interventions, reducing complications, and enhancing patient outcomes in lower abdominal and groin surgeries. However, further research is needed to elucidate the exact embryological and genetic underpinnings of these variations.
Collapse
Affiliation(s)
- Sanjukta Sahoo
- Anatomy, All India Institute of Medical Sciences, Bhubaneswar, IND
| | | | - Ravi K Narayan
- Anatomy, All India Institute of Medical Sciences, Bhubaneswar, IND
| |
Collapse
|
6
|
Bhat GR, Sethi I, Sadida HQ, Rah B, Mir R, Algehainy N, Albalawi IA, Masoodi T, Subbaraj GK, Jamal F, Singh M, Kumar R, Macha MA, Uddin S, Akil ASAS, Haris M, Bhat AA. Cancer cell plasticity: from cellular, molecular, and genetic mechanisms to tumor heterogeneity and drug resistance. Cancer Metastasis Rev 2024; 43:197-228. [PMID: 38329598 PMCID: PMC11016008 DOI: 10.1007/s10555-024-10172-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
Cancer is a complex disease displaying a variety of cell states and phenotypes. This diversity, known as cancer cell plasticity, confers cancer cells the ability to change in response to their environment, leading to increased tumor diversity and drug resistance. This review explores the intricate landscape of cancer cell plasticity, offering a deep dive into the cellular, molecular, and genetic mechanisms that underlie this phenomenon. Cancer cell plasticity is intertwined with processes such as epithelial-mesenchymal transition and the acquisition of stem cell-like features. These processes are pivotal in the development and progression of tumors, contributing to the multifaceted nature of cancer and the challenges associated with its treatment. Despite significant advancements in targeted therapies, cancer cell adaptability and subsequent therapy-induced resistance remain persistent obstacles in achieving consistent, successful cancer treatment outcomes. Our review delves into the array of mechanisms cancer cells exploit to maintain plasticity, including epigenetic modifications, alterations in signaling pathways, and environmental interactions. We discuss strategies to counteract cancer cell plasticity, such as targeting specific cellular pathways and employing combination therapies. These strategies promise to enhance the efficacy of cancer treatments and mitigate therapy resistance. In conclusion, this review offers a holistic, detailed exploration of cancer cell plasticity, aiming to bolster the understanding and approach toward tackling the challenges posed by tumor heterogeneity and drug resistance. As articulated in this review, the delineation of cellular, molecular, and genetic mechanisms underlying tumor heterogeneity and drug resistance seeks to contribute substantially to the progress in cancer therapeutics and the advancement of precision medicine, ultimately enhancing the prospects for effective cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Gh Rasool Bhat
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Jammu and Kashmir, India
| | - Itty Sethi
- Institute of Human Genetics, University of Jammu, Jammu, Jammu and Kashmir, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Bilal Rah
- Iron Biology Group, Research Institute of Medical and Health Science, University of Sharjah, Sharjah, UAE
| | - Rashid Mir
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk, Saudi Arabia
| | | | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | | | - Farrukh Jamal
- Dr. Rammanohar, Lohia Avadh University, Ayodhya, India
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Institute of Medical Sciences (AIIMS), Dr. BRAIRCH, All India, New Delhi, India
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory Animal Research Centre, Qatar University, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Mohammad Haris
- Laboratory Animal Research Centre, Qatar University, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
7
|
Thiery JP, Sheng G, Shu X, Runyan R. How studies in developmental epithelial-mesenchymal transition and mesenchymal-epithelial transition inspired new research paradigms in biomedicine. Development 2024; 151:dev200128. [PMID: 38300897 DOI: 10.1242/dev.200128] [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] [Indexed: 02/03/2024]
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse mechanism, mesenchymal-epithelial transition (MET), are evolutionarily conserved mechanisms initially identified in studies of early metazoan development. EMT may even have been established in choanoflagellates, the closest unicellular relative of Metazoa. These crucial morphological transitions operate during body plan formation and subsequently in organogenesis. These findings have prompted an increasing number of investigators in biomedicine to assess the importance of such mechanisms that drive epithelial cell plasticity in multiple diseases associated with congenital disabilities and fibrosis, and, most importantly, in the progression of carcinoma. EMT and MET also play crucial roles in regenerative medicine, notably by contributing epigenetic changes in somatic cells to initiate reprogramming into stem cells and their subsequent differentiation into distinct lineages.
Collapse
Affiliation(s)
| | - Guojun Sheng
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Xiaodong Shu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
8
|
Yahya I, Abduelmula A, Hockman D, Brand-Saberi B, Morosan-Puopolo G. The development of thoracic and abdominal muscle depends on SDF1 and CXCR4. Dev Biol 2024; 506:52-63. [PMID: 38070699 DOI: 10.1016/j.ydbio.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 11/29/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024]
Abstract
In vertebrates, the lateral body wall muscle formation is thought to be initiated by direct outgrowth of the dermomyotomes resulting in the elongation of the hypaxial myotomes. This contrasts with the formation of the muscles of the girdle, limbs and intrinsic tongue muscles, which originate from long-range migrating progenitors. Previous work shows that the migration of these progenitors requires CXCR4 which is specifically expressed in the migrating cells, but not in the dermomyotome. Here, we show that cells in the ventrolateral-lip (VLL) of the dermomyotome at the flank level express CXCR4 in a pattern consistent with that of Pax3 and MyoR. In ovo gain-of-function experiments using electroporation of SDF-1 constructs into the VLL resulted in increased expression of c-Met, Pax3 and MyoD. In contrast, a loss-of-function approach by implantation of CXCR4-inhibitor beads into the VLL of the flank region caused a reduction in the expression of these markers. These data show that CXCR4 is expressed in the VLL, and by experimentally manipulating the CXCR4/SDF-1 signaling, we demonstrate the importance of this axis in body wall muscle development.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany; Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan.
| | - Aisha Abduelmula
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany.
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, South Africa.
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany.
| | | |
Collapse
|
9
|
Szoszkiewicz A, Bukowska-Olech E, Jamsheer A. Molecular landscape of congenital vertebral malformations: recent discoveries and future directions. Orphanet J Rare Dis 2024; 19:32. [PMID: 38291488 PMCID: PMC10829358 DOI: 10.1186/s13023-024-03040-0] [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: 07/18/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
Vertebral malformations (VMs) pose a significant global health problem, causing chronic pain and disability. Vertebral defects occur as isolated conditions or within the spectrum of various congenital disorders, such as Klippel-Feil syndrome, congenital scoliosis, spondylocostal dysostosis, sacral agenesis, and neural tube defects. Although both genetic abnormalities and environmental factors can contribute to abnormal vertebral development, our knowledge on molecular mechanisms of numerous VMs is still limited. Furthermore, there is a lack of resource that consolidates the current knowledge in this field. In this pioneering review, we provide a comprehensive analysis of the latest research on the molecular basis of VMs and the association of the VMs-related causative genes with bone developmental signaling pathways. Our study identifies 118 genes linked to VMs, with 98 genes involved in biological pathways crucial for the formation of the vertebral column. Overall, the review summarizes the current knowledge on VM genetics, and provides new insights into potential involvement of biological pathways in VM pathogenesis. We also present an overview of available data regarding the role of epigenetic and environmental factors in VMs. We identify areas where knowledge is lacking, such as precise molecular mechanisms in which specific genes contribute to the development of VMs. Finally, we propose future research avenues that could address knowledge gaps.
Collapse
Affiliation(s)
- Anna Szoszkiewicz
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8, 60-806, Poznan, Poland.
| | - Ewelina Bukowska-Olech
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8, 60-806, Poznan, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8, 60-806, Poznan, Poland.
- Centers for Medical Genetics GENESIS, Dąbrowskiego 77A, 60-529, Poznan, Poland.
| |
Collapse
|
10
|
Draga M, Scaal M. Building a vertebra: Development of the amniote sclerotome. J Morphol 2024; 285:e21665. [PMID: 38100740 DOI: 10.1002/jmor.21665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/13/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
In embryonic development, the vertebral column arises from the sclerotomal compartment of the somites. The sclerotome is a mesenchymal cell mass which can be subdivided into several subpopulations specified by different regulatory mechanisms and giving rise to different parts of the vertebrae like vertebral body, vertebral arch, ribs, and vertebral joints. This review gives a short overview on the molecular and cellular basis of the formation of sclerotomal subdomains and the morphogenesis of their vertebral derivatives.
Collapse
Affiliation(s)
- Margarethe Draga
- Faculty of Medicine and University Hospital Cologne, Center of Anatomy, University of Cologne, Cologne, Germany
| | - Martin Scaal
- Faculty of Medicine and University Hospital Cologne, Center of Anatomy, University of Cologne, Cologne, Germany
| |
Collapse
|
11
|
Zeng Q, Du ZQ. Advances in the discovery of genetic elements underlying longissimus dorsi muscle growth and development in the pig. Anim Genet 2023; 54:709-720. [PMID: 37796678 DOI: 10.1111/age.13365] [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: 07/25/2022] [Revised: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 10/07/2023]
Abstract
As a major source of protein in human diets, pig meat plays a crucial role in ensuring global food security. Key determinants of meat production refer to the chemical and physical compositions or characteristics of muscle fibers, such as the number, hypertrophy potential, fiber-type conversion and intramuscular fat deposition. However, the growth and formation of muscle fibers comprises a complex process under spatio-temporal regulation, that is, the intermingled and concomitant proliferation, differentiation, migration and fusion of myoblasts. Recently, with the fast and continuous development of next-generation sequencing technology, the integration of quantitative trait loci mapping with genome-wide association studies (GWAS) has greatly helped animal geneticists to discover and explore thousands of functional or causal genetic elements underlying muscle growth and development. However, owing to the underlying complex molecular mechanisms, challenges to in-depth understanding and utilization remain, and the cost of large-scale sequencing, which requires integrated analyses of high-throughput omics data, is high. In this review, we mainly elaborate on research advances in integrative analyses (e.g. GWAS, omics) for identifying functional genes or genomic elements for longissimus dorsi muscle growth and development for different pig breeds, describing several successful transcriptome analyses and functional genomics cases, in an attempt to provide some perspective on the future functional annotation of genetic elements for muscle growth and development in pigs.
Collapse
Affiliation(s)
- Qingjie Zeng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Zhi-Qiang Du
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
12
|
Ibarra-Soria X, Thierion E, Mok GF, Münsterberg AE, Odom DT, Marioni JC. A transcriptional and regulatory map of mouse somite maturation. Dev Cell 2023; 58:1983-1995.e7. [PMID: 37499658 PMCID: PMC10563765 DOI: 10.1016/j.devcel.2023.07.003] [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: 01/25/2023] [Revised: 05/12/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023]
Abstract
The mammalian body plan is shaped by rhythmic segmentation of mesoderm into somites, which are transient embryonic structures that form down each side of the neural tube. We have analyzed the genome-wide transcriptional and chromatin dynamics occurring within nascent somites, from early inception of somitogenesis to the latest stages of body plan establishment. We created matched gene expression and open chromatin maps for the three leading pairs of somites at six time points during mouse embryonic development. We show that the rate of somite differentiation accelerates as development progresses. We identified a conserved maturation program followed by all somites, but somites from more developed embryos concomitantly switch on differentiation programs from derivative cell lineages soon after segmentation. Integrated analysis of the somitic transcriptional and chromatin activities identified opposing regulatory modules controlling the onset of differentiation. Our results provide a powerful, high-resolution view of the molecular genetics underlying somitic development in mammals.
Collapse
Affiliation(s)
- Ximena Ibarra-Soria
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| | - Elodie Thierion
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Gi Fay Mok
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Andrea E Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Duncan T Odom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; DKFZ, Division of Regulatory Genomics and Cancer Evolution B270, Im Neunheimer Feld 280, Heidelberg, 69120, Germany.
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK.
| |
Collapse
|
13
|
Sahai-Hernandez P, Pouget C, Eyal S, Svoboda O, Chacon J, Grimm L, Gjøen T, Traver D. Dermomyotome-derived endothelial cells migrate to the dorsal aorta to support hematopoietic stem cell emergence. eLife 2023; 12:e58300. [PMID: 37695317 PMCID: PMC10495111 DOI: 10.7554/elife.58300] [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: 04/27/2020] [Accepted: 09/03/2023] [Indexed: 09/12/2023] Open
Abstract
Development of the dorsal aorta is a key step in the establishment of the adult blood-forming system, since hematopoietic stem and progenitor cells (HSPCs) arise from ventral aortic endothelium in all vertebrate animals studied. Work in zebrafish has demonstrated that arterial and venous endothelial precursors arise from distinct subsets of lateral plate mesoderm. Here, we profile the transcriptome of the earliest detectable endothelial cells (ECs) during zebrafish embryogenesis to demonstrate that tissue-specific EC programs initiate much earlier than previously appreciated, by the end of gastrulation. Classic studies in the chick embryo showed that paraxial mesoderm generates a subset of somite-derived endothelial cells (SDECs) that incorporate into the dorsal aorta to replace HSPCs as they exit the aorta and enter circulation. We describe a conserved program in the zebrafish, where a rare population of endothelial precursors delaminates from the dermomyotome to incorporate exclusively into the developing dorsal aorta. Although SDECs lack hematopoietic potential, they act as a local niche to support the emergence of HSPCs from neighboring hemogenic endothelium. Thus, at least three subsets of ECs contribute to the developing dorsal aorta: vascular ECs, hemogenic ECs, and SDECs. Taken together, our findings indicate that the distinct spatial origins of endothelial precursors dictate different cellular potentials within the developing dorsal aorta.
Collapse
Affiliation(s)
- Pankaj Sahai-Hernandez
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Claire Pouget
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Shai Eyal
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Ondrej Svoboda
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
- Department of Cell Differentiation, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic v.v.i, Prague, Czech Republic
| | - Jose Chacon
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Lin Grimm
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Tor Gjøen
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - David Traver
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| |
Collapse
|
14
|
Deguchi K, Zambaiti E, De Coppi P. Regenerative medicine: current research and perspective in pediatric surgery. Pediatr Surg Int 2023; 39:167. [PMID: 37014468 PMCID: PMC10073065 DOI: 10.1007/s00383-023-05438-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/05/2023]
Abstract
The field of regenerative medicine, encompassing several disciplines including stem cell biology and tissue engineering, continues to advance with the accumulating research on cell manipulation technologies, gene therapy and new materials. Recent progress in preclinical and clinical studies may transcend the boundaries of regenerative medicine from laboratory research towards clinical reality. However, for the ultimate goal to construct bioengineered transplantable organs, a number of issues still need to be addressed. In particular, engineering of elaborate tissues and organs requires a fine combination of different relevant aspects; not only the repopulation of multiple cell phenotypes in an appropriate distribution but also the adjustment of the host environmental factors such as vascularisation, innervation and immunomodulation. The aim of this review article is to provide an overview of the recent discoveries and development in stem cells and tissue engineering, which are inseparably interconnected. The current status of research on tissue stem cells and bioengineering, and the possibilities for application in specific organs relevant to paediatric surgery have been specifically focused and outlined.
Collapse
Affiliation(s)
- Koichi Deguchi
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Elisa Zambaiti
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK
- UOC Chirurgia Pediatrica, Ospedale Infantile Regina Margherita, Turin, Italy
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK.
- NIHR BRC SNAPS Great Ormond Street Hospitals, London, UK.
- Stem Cells and Regenerative Medicine Section, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
15
|
Piatkowska AM, Adhikari K, Moverley AA, Turmaine M, Glazier JA, Plachta N, Evans SE, Stern CD. Sequential changes in cellular properties accompanying amniote somite formation. J Anat 2022; 242:417-435. [PMID: 36423208 PMCID: PMC9919497 DOI: 10.1111/joa.13791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/30/2022] [Accepted: 10/28/2022] [Indexed: 11/26/2022] Open
Abstract
Somites are transient structures derived from the pre-somitic mesoderm (PSM), involving mesenchyme-to-epithelial transition (MET) where the cells change their shape and polarize. Using Scanning electron microscopy (SEM), immunocytochemistry and confocal microscopy, we study the progression of these events along the tail-to-head axis of the embryo, which mirrors the progression of somitogenesis (younger cells located more caudally). SEM revealed that PSM epithelialization is a gradual process, which begins much earlier than previously thought, starting with the dorsalmost cells, then the medial ones, and then, simultaneously, the ventral and lateral cells, before a somite fully separates from the PSM. The core (internal) cells of the PSM and somites never epithelialize, which suggests that the core cells could be 'trapped' within the somitocoele after cells at the surfaces of the PSM undergo MET. Three-dimensional imaging of the distribution of the cell polarity markers PKCζ, PAR3, ZO1, the Golgi marker GM130 and the apical marker N-cadherin reveal that the pattern of polarization is distinctive for each marker and for each surface of the PSM, but the order of these events is not the same as the progression of cell elongation. These observations challenge some assumptions underlying existing models of somite formation.
Collapse
Affiliation(s)
- Agnieszka M. Piatkowska
- Department of Cell & Developmental BiologyUniversity College London, Gower Street (Anatomy Building)LondonUK
| | - Kaustubh Adhikari
- Department of Cell & Developmental BiologyUniversity College London, Gower Street (Anatomy Building)LondonUK,Present address:
The Open UniversityMilton KeynesUK
| | - Adam A. Moverley
- Department of Cell & Developmental BiologyUniversity College London, Gower Street (Anatomy Building)LondonUK
| | - Mark Turmaine
- Department of Cell & Developmental BiologyUniversity College London, Gower Street (Anatomy Building)LondonUK
| | - James A. Glazier
- Department of Intelligent Systems EngineeringBiocomplexity InstituteBloomingtonIndianaUSA
| | - Nicolas Plachta
- Department of Cell and Developmental Biology, 9‐123 Smilow Center for Translational Research, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Susan E. Evans
- Department of Cell & Developmental BiologyUniversity College London, Gower Street (Anatomy Building)LondonUK
| | - Claudio D. Stern
- Department of Cell & Developmental BiologyUniversity College London, Gower Street (Anatomy Building)LondonUK
| |
Collapse
|
16
|
Sinha S, Elbaz‐Alon Y, Avinoam O. Ca 2+ as a coordinator of skeletal muscle differentiation, fusion and contraction. FEBS J 2022; 289:6531-6542. [PMID: 35689496 PMCID: PMC9795905 DOI: 10.1111/febs.16552] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/05/2022] [Accepted: 06/09/2022] [Indexed: 12/30/2022]
Abstract
Muscle regeneration is essential for vertebrate muscle homeostasis and recovery after injury. During regeneration, muscle stem cells differentiate into myocytes, which then fuse with pre-existing muscle fibres. Hence, differentiation, fusion and contraction must be tightly regulated during regeneration to avoid the disastrous consequences of premature fusion of myocytes to actively contracting fibres. Cytosolic calcium (Ca2+ ), which is coupled to both induction of myogenic differentiation and contraction, has more recently been implicated in the regulation of myocyte-to-myotube fusion. In this viewpoint, we propose that Ca2+ -mediated coordination of differentiation, fusion and contraction is a feature selected in the amniotes to facilitate muscle regeneration.
Collapse
Affiliation(s)
- Sansrity Sinha
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Yael Elbaz‐Alon
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Ori Avinoam
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
17
|
Yahya I, Hockman D, Brand-Saberi B, Morosan-Puopolo G. New Insights into the Diversity of Branchiomeric Muscle Development: Genetic Programs and Differentiation. BIOLOGY 2022; 11:biology11081245. [PMID: 36009872 PMCID: PMC9404950 DOI: 10.3390/biology11081245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/30/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022]
Abstract
Simple Summary We review the transcription factors and signaling molecules driving differentiation of a subset of head muscles known as the branchiomeric muscles due to their origin in the pharyngeal arches. We provide novel data on the distinct myogenic programs within these muscles and explore how the cranial neural crest cell regulates branchiomeric muscle patterning and differentiation. Abstract Branchiomeric skeletal muscles are a subset of head muscles originating from skeletal muscle progenitor cells in the mesodermal core of pharyngeal arches. These muscles are involved in facial expression, mastication, and function of the larynx and pharynx. Branchiomeric muscles have been the focus of many studies over the years due to their distinct developmental programs and common origin with the heart muscle. A prerequisite for investigating these muscles’ properties and therapeutic potential is understanding their genetic program and differentiation. In contrast to our understanding of how branchiomeric muscles are formed, less is known about their differentiation. This review focuses on the differentiation of branchiomeric muscles in mouse embryos. Furthermore, the relationship between branchiomeric muscle progenitor and neural crest cells in the pharyngeal arches of chicken embryos is also discussed. Additionally, we summarize recent studies into the genetic networks that distinguish between first arch-derived muscles and other pharyngeal arch muscles.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum 11115, Sudan
- Division of Cell Biology, Department of Human Biology, Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Correspondence: (I.Y.); (G.M.-P.)
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Gabriela Morosan-Puopolo
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany
- Correspondence: (I.Y.); (G.M.-P.)
| |
Collapse
|
18
|
Shi J, Li W, Liu A, Ren L, Zhang P, Jiang T, Han Y, Liu L. MiRNA sequencing of Embryonic Myogenesis in Chengkou Mountain Chicken. BMC Genomics 2022; 23:571. [PMID: 35948880 PMCID: PMC9364561 DOI: 10.1186/s12864-022-08795-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/27/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Skeletal muscle tissue is among the largest organ systems in mammals, essential for survival and movement. Embryonic muscle development determines the quantity and quality of muscles after the birth of an individual. MicroRNAs (miRNAs) are a significant class of non-coding RNAs that bind to the 3'UTR region of mRNA to regulate gene function. Total RNA was extracted from the leg muscles of chicken embryos in different developmental stages of Chengkou Mountain Chicken and used to generate 171,407,341 clean small RNA reads. Target prediction, GO, and KEGG enrichment analyses determined the significantly enriched genes and pathways. Differential analysis determined the significantly different miRNAs between chicken embryo leg muscles at different developmental stages. Meanwhile, the weighted correlation network analysis (WGCNA) identified key modules in different developmental stages, and the hub miRNAs were screened following the KME value. RESULTS The clean reads contained 2047 miRNAs, including 721 existing miRNAs, 1059 known miRNAs, and 267 novel miRNAs. Many genes and pathways related to muscle development were identified, including ERBB4, MEF2C, FZD4, the Wnt, Notch, and MAPK signaling pathways. The WGCNA established the greenyellow module and gga-miR-130b-5p for E12, magenta module and gga-miR-1643-5p for E16, purple module and gga-miR-12218-5p for E19, cyan module and gga-miR-132b-5p for E21. CONCLUSION These results lay a foundation for further research on the molecular regulatory mechanism of embryonic muscle development in Chengkou mountain chicken and provide a reference for other poultry and livestock muscle development studies.
Collapse
Affiliation(s)
- Jun'an Shi
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Wendong Li
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Anfang Liu
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Lingtong Ren
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Pusen Zhang
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Ting Jiang
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Yuqing Han
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China
| | - Lingbin Liu
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Beibei, Chongqing, 400700, China.
| |
Collapse
|
19
|
Unique Features of River Lamprey (Lampetra fluviatilis) Myogenesis. Int J Mol Sci 2022; 23:ijms23158595. [PMID: 35955736 PMCID: PMC9368804 DOI: 10.3390/ijms23158595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
The river lamprey (L. fluviatilis) is a representative of the ancestral jawless vertebrate group. We performed a histological analysis of trunk muscle fiber differentiation during embryonal, larval, and adult musculature development in this previously unstudied species. Investigation using light, transmission electron (TEM), and confocal microscopy revealed that embryonal and larval musculature differs from adult muscle mass. Here, we present the morphological analysis of L. fluviatilis myogenesis, from unsegmented mesoderm through somite formation, and their differentiation into multinucleated muscle lamellae. Our analysis also revealed the presence of myogenic factors LfPax3/7 and Myf5 in the dermomyotome. In the next stages of development, two types of muscle lamellae can be distinguished: central surrounded by parietal. This pattern is maintained until adulthood, when parietal muscle fibers surround the central muscles on both sides. The two types show different morphological characteristics. Although lampreys are phylogenetically distant from jawed vertebrates, somite morphology, especially dermomyotome function, shows similarity. Here we demonstrate that somitogenesis is a conservative process among all vertebrates. We conclude that river lamprey myogenesis shares features with both ancestral and higher vertebrates.
Collapse
|
20
|
Clayton SW, Angermeier A, Halbrooks JE, McCardell R, Serra R. TGFβ signaling is required for sclerotome resegmentation during development of the spinal column in Gallus gallus. Dev Biol 2022; 488:120-130. [PMID: 35644252 PMCID: PMC9552462 DOI: 10.1016/j.ydbio.2022.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 12/14/2022]
Abstract
We previously showed the importance of TGFβ signaling in development of the mouse axial skeleton. Here, we provide the first direct evidence that TGFβ signaling is required for resegmentation of the sclerotome using chick embryos. Lipophilic fluorescent tracers, DiO and DiD, were microinjected into adjacent somites of embryos treated with or without TGFβRI inhibitors, SB431542, SB525334 or SD208, at developmental day E2.5 (HH16). Lineage tracing of labeled cells was observed over the course of 4 days until the completion of resegmentation at E6.5 (HH32). Vertebrae were malformed and intervertebral discs were small and misshapen in inhibitor injected embryos. Hypaxial myofibers were also increased in thickness after treatment with the inhibitor. Inhibition of TGFβ signaling resulted in alterations in resegmentation that ranged between full, partial, and slanted shifts in distribution of DiO or DiD labeled cells within vertebrae. Patterning of rostro-caudal markers within sclerotome was disrupted at E3.5 after treatment with TGFβRI inhibitor with rostral domains expressing both rostral and caudal markers. We propose that TGFβ signaling regulates rostro-caudal polarity and subsequent resegmentation in sclerotome during spinal column development.
Collapse
Affiliation(s)
- Sade W Clayton
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA; Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Allyson Angermeier
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA
| | - Jacob E Halbrooks
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA
| | - Ronisha McCardell
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA; Dillard University, Greensburg, LA, USA
| | - Rosa Serra
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA.
| |
Collapse
|
21
|
Schaeffer J, Weber IP, Thompson AJ, Keynes RJ, Franze K. Axons in the Chick Embryo Follow Soft Pathways Through Developing Somite Segments. Front Cell Dev Biol 2022; 10:917589. [PMID: 35874821 PMCID: PMC9304555 DOI: 10.3389/fcell.2022.917589] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
During patterning of the peripheral nervous system, motor axons grow sequentially out of the neural tube in a segmented fashion to ensure functional integration of the motor roots between the surrounding cartilage and bones of the developing vertebrae. This segmented outgrowth is regulated by the intrinsic properties of each segment (somite) adjacent to the neural tube, and in particular by chemical repulsive guidance cues expressed in the posterior half. Yet, knockout models for such repulsive cues still display initial segmentation of outgrowing motor axons, suggesting the existence of additional, yet unknown regulatory mechanisms of axon growth segmentation. As neuronal growth is not only regulated by chemical but also by mechanical signals, we here characterized the mechanical environment of outgrowing motor axons. Using atomic force microscopy-based indentation measurements on chick embryo somite strips, we identified stiffness gradients in each segment, which precedes motor axon growth. Axon growth was restricted to the anterior, softer tissue, which showed lower cell body densities than the repulsive stiffer posterior parts at later stages. As tissue stiffness is known to regulate axon growth during development, our results suggest that motor axons also respond to periodic stiffness gradients imposed by the intrinsic mechanical properties of somites.
Collapse
Affiliation(s)
- Julia Schaeffer
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Inserm, U1216, Grenoble Institut Neurosciences, Univ. Grenoble Alpes, Grenoble, France
- *Correspondence: Julia Schaeffer, ; Kristian Franze,
| | - Isabell P. Weber
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Amelia J. Thompson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Roger J. Keynes
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Institute of Medical Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- *Correspondence: Julia Schaeffer, ; Kristian Franze,
| |
Collapse
|
22
|
Wang J, Wen Y, Xu J, Yue B, Zhong J, Zheng L, Lei C, Chen H, Huang Y. Circ RIMKLB promotes myoblast proliferation and inhibits differentiation by sponging miR-29c to release KCNJ12. Epigenetics 2022; 17:1686-1700. [PMID: 35348434 PMCID: PMC9621043 DOI: 10.1080/15592294.2022.2058211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Muscle development is a complex process that was regulated by many factors, among which non-coding RNAs (ncRNAs) play a vital role in regulating multiple life activities of muscle cells. Circular RNA (circRNA), a type of non-coding RNA with closed-loop structure, has been reported to affect multiple life processes. However, the roles of circRNAs on muscle development have not been fully elucidated. The present study aimed to determine whether and how circRIMKLB affects muscle development. Our study revealed that circRIMKLB promoted myoblast proliferation and inhibited differentiation. Besides, miR-29c was proved as a downstream target of circRIMKLB using dual-luciferase reporter assay and RNA-binding protein immunoprecipitation (RIP) assay. Also, potassium inwardly rectifying channel subfamily J member 12 (KCNJ12) was identified as a novel target of miR-29c via dual-luciferase reporter assay, quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), and western blot. CircRIMKLB and KCNJ12 were both proved to regulate cell cycle on muscle regeneration after injury in vivo. In conclusion, we demonstrated that circRIMKLB sponged miR-29c, releasing KCNJ12 to regulate myoblast proliferation and differentiation and regulating cell cycle during muscle regeneration after injury in vivo.
Collapse
Affiliation(s)
- Jian Wang
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Yifan Wen
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Jiawei Xu
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Binglin Yue
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Jialin Zhong
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Li Zheng
- College of Animal Science & Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China
| | - Chuzhao Lei
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Hong Chen
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| | - Yongzhen Huang
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&f University, Yangling, Shaanxi, China
| |
Collapse
|
23
|
Della Gaspera B, Weill L, Chanoine C. Evolution of Somite Compartmentalization: A View From Xenopus. Front Cell Dev Biol 2022; 9:790847. [PMID: 35111756 PMCID: PMC8802780 DOI: 10.3389/fcell.2021.790847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Somites are transitory metameric structures at the basis of the axial organization of vertebrate musculoskeletal system. During evolution, somites appear in the chordate phylum and compartmentalize mainly into the dermomyotome, the myotome, and the sclerotome in vertebrates. In this review, we summarized the existing literature about somite compartmentalization in Xenopus and compared it with other anamniote and amniote vertebrates. We also present and discuss a model that describes the evolutionary history of somite compartmentalization from ancestral chordates to amniote vertebrates. We propose that the ancestral organization of chordate somite, subdivided into a lateral compartment of multipotent somitic cells (MSCs) and a medial primitive myotome, evolves through two major transitions. From ancestral chordates to vertebrates, the cell potency of MSCs may have evolved and gave rise to all new vertebrate compartments, i.e., the dermomyome, its hypaxial region, and the sclerotome. From anamniote to amniote vertebrates, the lateral MSC territory may expand to the whole somite at the expense of primitive myotome and may probably facilitate sclerotome formation. We propose that successive modifications of the cell potency of some type of embryonic progenitors could be one of major processes of the vertebrate evolution.
Collapse
|
24
|
Manneken JD, Dauer MVP, Currie PD. Dynamics of muscle growth and regeneration: Lessons from the teleost. Exp Cell Res 2021; 411:112991. [PMID: 34958765 DOI: 10.1016/j.yexcr.2021.112991] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/16/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022]
Abstract
The processes of myogenesis during both development and regeneration share a number of similarities across both amniotes and teleosts. In amniotes, the process of muscle formation is considered largely biphasic, with developmental myogenesis occurring through hyperplastic fibre deposition and postnatal muscle growth driven through hypertrophy of existing fibres. In contrast, teleosts continue generating new muscle fibres during adult myogenesis through a process of eternal hyperplasia using a dedicated stem cell system termed the external cell layer. During developmental and regenerative myogenesis alike, muscle progenitors interact with their niche to receive cues guiding their transition into myoblasts and ultimately mature myofibres. During development, muscle precursors receive input from neighbouring embryological tissues; however, during repair, this role is fulfilled by other injury resident cell types, such as those of the innate immune response. Recent work has focused on the role of macrophages as a pro-regenerative cell type which provides input to muscle satellite cells during regenerative myogenesis. As zebrafish harbour a satellite cell system analogous to that of mammals, the processes of regeneration can be interrogated in vivo with the imaging intensive approaches afforded in the zebrafish system. This review discusses the strengths of zebrafish with a focus on both the similarities and differences to amniote myogenesis during both development and repair.
Collapse
Affiliation(s)
- Jessica D Manneken
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria, 3800, Australia
| | - Mervyn V P Dauer
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria, 3800, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria, 3800, Australia; EMBL Australia, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
25
|
Taylor L, Wankell M, Saxena P, McFarlane C, Hebbard L. Cell adhesion an important determinant of myogenesis and satellite cell activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119170. [PMID: 34763027 DOI: 10.1016/j.bbamcr.2021.119170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
Skeletal muscles represent a complex and highly organised tissue responsible for all voluntary body movements. Developed through an intricate and tightly controlled process known as myogenesis, muscles form early in development and are maintained throughout life. Due to the constant stresses that muscles are subjected to, skeletal muscles maintain a complex course of regeneration to both replace and repair damaged myofibers and to form new functional myofibers. This process, made possible by a pool of resident muscle stem cells, termed satellite cells, and controlled by an array of transcription factors, is additionally reliant on a diverse range of cell adhesion molecules and the numerous signaling cascades that they initiate. This article will review the literature surrounding adhesion molecules and their roles in skeletal muscle myogenesis and repair.
Collapse
Affiliation(s)
- Lauren Taylor
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia
| | - Miriam Wankell
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia
| | - Pankaj Saxena
- Department of Cardiothoracic Surgery, The Townsville University Hospital, Townsville, Queensland, Australia; College of Medicine, Dentistry, James Cook University, Townsville, Queensland, Australia
| | - Craig McFarlane
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia.
| | - Lionel Hebbard
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia; Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
26
|
Kusakabe R, Tanaka M, Kuratani S. Developmental Evolution of Hypaxial Muscles: Insights From Cyclostomes and Chondrichthyans. Front Cell Dev Biol 2021; 9:760366. [PMID: 34650989 PMCID: PMC8505881 DOI: 10.3389/fcell.2021.760366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/10/2021] [Indexed: 11/30/2022] Open
Abstract
Jawed vertebrates possess two distinct groups of muscles in the trunk (epaxial and hypaxial muscles) primarily defined by the pattern of motor innervation from the spinal cord. Of these, the hypaxial group includes muscles with highly differentiated morphology and function, such as the muscles associated with paired limbs, shoulder girdles and tongue/infrahyoid (hypobranchial) muscles. Here we summarize the latest findings on the evolutionary mechanisms underlying the morphological variety of hypaxial musculature, with special reference to the molecular insights obtained from several living species that diverged early in vertebrate evolution. Lampreys, extant jawless vertebrates, lack many of derived traits characteristic of the gnathostomes, such as jaws, paired fins and epaxial/hypaxial distinction of the trunk skeletal musculatures. However, these animals possess the primitive form of the hypobranchial muscle. Of the gnathostomes, the elasmobranchs exhibit developmental mode of hypaxial muscles that is not identical to that of other gnathostomes in that the muscle primordia relocate as coherent cell aggregates. Comparison of expression of developmental genes, including Lbx genes, has delineated the temporal order of differentiation of various skeletal muscles, such as the hypobranchial, posterior pharyngeal and cucullaris (trapezius) muscles. We have proposed that the sequential addition of distal muscles, associated with expression of duplicated Lbx genes, promoted the elaboration of skeletal musculature. These analyses have revealed the framework of an evolutionary pathway that gave rise to the morphological complexity and diversity of vertebrate body patterns.
Collapse
Affiliation(s)
- Rie Kusakabe
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Masako Tanaka
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan.,Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan
| |
Collapse
|
27
|
Lu P, Lu Y. Born to Run? Diverse Modes of Epithelial Migration. Front Cell Dev Biol 2021; 9:704939. [PMID: 34540829 PMCID: PMC8448196 DOI: 10.3389/fcell.2021.704939] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Bundled with various kinds of adhesion molecules and anchored to the basement membrane, the epithelium has historically been considered as an immotile tissue and, to migrate, it first needs to undergo epithelial-mesenchymal transition (EMT). Since its initial description more than half a century ago, the EMT process has fascinated generations of developmental biologists and, more recently, cancer biologists as it is believed to be essential for not only embryonic development, organ formation, but cancer metastasis. However, recent progress shows that epithelium is much more motile than previously realized. Here, we examine the emerging themes in epithelial collective migration and how this has impacted our understanding of EMT.
Collapse
Affiliation(s)
- Pengfei Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yunzhe Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
28
|
Xu S, He X, Shi J, Li Z, Song J, Wang J, Wang G, Brand-Saberi B, Cheng X, Yang X. Interaction between retinoic acid and FGF/ERK signals are involved in Dexamethasone-induced abnormal myogenesis during embryonic development. Toxicology 2021; 461:152917. [PMID: 34464682 DOI: 10.1016/j.tox.2021.152917] [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: 04/17/2021] [Revised: 08/01/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Despite the common application in pregnancy at clinical practice, it remains ambiguous whether dexamethasone (Dex) exposure can affect embryonic myogenesis. In this study, firstly we showed that 10-6 M Dex (Cheng et al., 2016; 2017) treatment resulted in abnormal myogenesis in chicken embryos. Secondly, we demonstrated that 10-6 M Dex-induced abnormality of myogenesis resulted from aberrant cell proliferation, as well as from alteration of the differentiation process from the early stage of somitogenesis up to the late stage of myogenesis. The above-mentioned results caused by Dex exposure might be due to the aberrant gene expressions of somite formation (Raldh2, Fgf8, Wnt3a, β-catenin, Slug, Paraxis, N-cadherin) and differentiation (Pax3, MyoD, Wnt3a, Msx1, Shh). Thirdly, RNA sequencing implied the statistically significant differential gene expressions in regulating the myofibril and systemic development, as well as a dramatical alteration of retinoic acid (RA) signaling during somite development in the chicken embryos exposed to Dex. The subsequent validation experiments verified that Dex treatment indeed led to a metabolic change of RA signaling, which was up-regulated and principally mediated by FGF-ERK signaling revealed by means of the combination of chicken embryos and in vitro C2C12 cells. These findings highlight that 10-6 M Dex exposure enhances the risk of abnormal myogenesis through interfering with RA signaling during development.
Collapse
Affiliation(s)
- Shujie Xu
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Xiangyue He
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China; Department of Pathology, Medical School, Jinan University, Guangzhou, 510632, China
| | - Junzhu Shi
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Ziguang Li
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Jinhuan Song
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Jingyun Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Guang Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstrasse 150, 44801, Bochum, Germany
| | - Xin Cheng
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Xuesong Yang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
29
|
Function and regulation of muscle stem cells in skeletal muscle development and regeneration: a narrative review. JOURNAL OF BIO-X RESEARCH 2021. [DOI: 10.1097/jbr.0000000000000105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
30
|
Kim H, Kwon Y, Zhu C, Wu F, Kwon S, Yeo W, Choo HJ. Real-Time Functional Assay of Volumetric Muscle Loss Injured Mouse Masseter Muscles via Nanomembrane Electronics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101037. [PMID: 34218527 PMCID: PMC8425913 DOI: 10.1002/advs.202101037] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Indexed: 05/11/2023]
Abstract
Skeletal muscle has a remarkable regeneration capacity to recover its structure and function after injury, except for the traumatic loss of critical muscle volume, called volumetric muscle loss (VML). Although many extremity VML models have been conducted, craniofacial VML has not been well-studied due to unavailable in vivo assay tools. Here, this paper reports a wireless, noninvasive nanomembrane system that integrates skin-wearable printed sensors and electronics for real-time, continuous monitoring of VML on craniofacial muscles. The craniofacial VML model, using biopsy punch-induced masseter muscle injury, shows impaired muscle regeneration. To measure the electrophysiology of small and round masseter muscles of active mice during mastication, a wearable nanomembrane system with stretchable graphene sensors that can be laminated to the skin over target muscles is utilized. The noninvasive system provides highly sensitive electromyogram detection on masseter muscles with or without VML injury. Furthermore, it is demonstrated that the wireless sensor can monitor the recovery after transplantation surgery for craniofacial VML. Overall, the presented study shows the enormous potential of the masseter muscle VML injury model and wearable assay tool for the mechanism study and the therapeutic development of craniofacial VML.
Collapse
Affiliation(s)
- Hojoong Kim
- George W. Woodruff School of Mechanical EngineeringCollege of EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Center for Human‐Centric Interfaces and EngineeringInstitute for Electronics and NanotechnologyGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Young‐Tae Kwon
- Department for Metal PowderKorea Institute of Materials ScienceChangwon51508South Korea
| | - Carol Zhu
- Department of Cell BiologySchool of MedicineEmory UniversityAtlantaGA30322USA
| | - Fang Wu
- Department of Cell BiologySchool of MedicineEmory UniversityAtlantaGA30322USA
| | - Shinjae Kwon
- George W. Woodruff School of Mechanical EngineeringCollege of EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Center for Human‐Centric Interfaces and EngineeringInstitute for Electronics and NanotechnologyGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Woon‐Hong Yeo
- George W. Woodruff School of Mechanical EngineeringCollege of EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Center for Human‐Centric Interfaces and EngineeringInstitute for Electronics and NanotechnologyGeorgia Institute of TechnologyAtlantaGA30332USA
- Wallace H. Coulter Department of Biomedical EngineeringParker H. Petit Institute for Bioengineering and BiosciencesInstitute for MaterialsNeural Engineering CenterInstitute for Robotics and Intelligent MachinesGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Hyojung J. Choo
- Department of Cell BiologySchool of MedicineEmory UniversityAtlantaGA30322USA
| |
Collapse
|
31
|
Piatkowska AM, Evans SE, Stern CD. Cellular aspects of somite formation in vertebrates. Cells Dev 2021; 168:203732. [PMID: 34391979 DOI: 10.1016/j.cdev.2021.203732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
Vertebrate segmentation, the process that generates a regular arrangement of somites and thereby establishes the pattern of the adult body and of the musculoskeletal and peripheral nervous systems, was noticed many centuries ago. In the last few decades, there has been renewed interest in the process and especially in the molecular mechanisms that might account for its regularity and other spatial-temporal properties. Several models have been proposed but surprisingly, most of these do not provide clear links between the molecular mechanisms and the cell behaviours that generate the segmental pattern. Here we present a short survey of our current knowledge about the cellular aspects of vertebrate segmentation and the similarities and differences between different vertebrate groups in how they achieve their metameric pattern. Taking these variations into account should help to assess each of the models more appropriately.
Collapse
Affiliation(s)
- Agnieszka M Piatkowska
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK
| | - Susan E Evans
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK.
| |
Collapse
|
32
|
Costa ML, Jurberg AD, Mermelstein C. The Role of Embryonic Chick Muscle Cell Culture in the Study of Skeletal Myogenesis. Front Physiol 2021; 12:668600. [PMID: 34093232 PMCID: PMC8173222 DOI: 10.3389/fphys.2021.668600] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/21/2021] [Indexed: 11/13/2022] Open
Abstract
The mechanisms involved in the development of skeletal muscle fibers have been studied in the last 70 years and yet many aspects of this process are still not completely understood. A myriad of in vivo and in vitro invertebrate and vertebrate animal models has been used for dissecting the molecular and cellular events involved in muscle formation. Among the most used animal models for the study of myogenesis are the rodents rat and mouse, the fruit fly Drosophila, and the birds chicken and quail. Here, we describe the robustness and advantages of the chick primary muscle culture model for the study of skeletal myogenesis. In the myoblast culture obtained from embryonic chick pectoralis muscle it is possible to analyze all the steps involved in skeletal myogenesis, such as myoblast proliferation, withdrawal from cell cycle, cell elongation and migration, myoblast alignment and fusion, the assembly of striated myofibrils, and the formation of multinucleated myotubes. The fact that in vitro chick myotubes can harbor hundreds of nuclei, whereas myotubes from cell lines have only a dozen nuclei demonstrates the high level of differentiation of the autonomous chick myogenic program. This striking differentiation is independent of serum withdrawal, which points to the power of the model. We also review the major pro-myogenic and anti-myogenic molecules and signaling pathways involved in chick myogenesis, in addition to providing a detailed protocol for the preparation of embryonic chick myogenic cultures. Moreover, we performed a bibliometric analysis of the articles that used this model to evaluate which were the main explored topics of interest and their contributors. We expect that by describing the major findings, and their advantages, of the studies using the embryonic chick myogenic model we will foster new studies on the molecular and cellular process involved in muscle proliferation and differentiation that are more similar to the actual in vivo condition than the muscle cell lines.
Collapse
Affiliation(s)
- Manoel L Costa
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arnon D Jurberg
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Faculdade de Medicina-Presidente Vargas, Universidade Estácio de Sá, Rio de Janeiro, Brazil
| | - Claudia Mermelstein
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Yong LW, Lu TM, Tung CH, Chiou RJ, Li KL, Yu JK. Somite Compartments in Amphioxus and Its Implications on the Evolution of the Vertebrate Skeletal Tissues. Front Cell Dev Biol 2021; 9:607057. [PMID: 34041233 PMCID: PMC8141804 DOI: 10.3389/fcell.2021.607057] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Mineralized skeletal tissues of vertebrates are an evolutionary novelty within the chordate lineage. While the progenitor cells that contribute to vertebrate skeletal tissues are known to have two embryonic origins, the mesoderm and neural crest, the evolutionary origin of their developmental process remains unclear. Using cephalochordate amphioxus as our model, we found that cells at the lateral wall of the amphioxus somite express SPARC (a crucial gene for tissue mineralization) and various collagen genes. During development, some of these cells expand medially to surround the axial structures, including the neural tube, notochord and gut, while others expand laterally and ventrally to underlie the epidermis. Eventually these cell populations are found closely associated with the collagenous matrix around the neural tube, notochord, and dorsal aorta, and also with the dense collagen sheets underneath the epidermis. Using known genetic markers for distinct vertebrate somite compartments, we showed that the lateral wall of amphioxus somite likely corresponds to the vertebrate dermomyotome and lateral plate mesoderm. Furthermore, we demonstrated a conserved role for BMP signaling pathway in somite patterning of both amphioxus and vertebrates. These results suggest that compartmentalized somites and their contribution to primitive skeletal tissues are ancient traits that date back to the chordate common ancestor. The finding of SPARC-expressing skeletal scaffold in amphioxus further supports previous hypothesis regarding SPARC gene family expansion in the elaboration of the vertebrate mineralized skeleton.
Collapse
Affiliation(s)
- Luok Wen Yong
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Tsai-Ming Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Che-Huang Tung
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Department of Aquatic Biology, Chia-Yi University, Chia-Yi, Taiwan
| | - Ruei-Jen Chiou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kun-Lung Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jr-Kai Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Yilan, Taiwan
| |
Collapse
|
34
|
Wang M, Song W, Jin C, Huang K, Yu Q, Qi J, Zhang Q, He Y. Pax3 and Pax7 Exhibit Distinct and Overlapping Functions in Marking Muscle Satellite Cells and Muscle Repair in a Marine Teleost, Sebastes schlegelii. Int J Mol Sci 2021; 22:ijms22073769. [PMID: 33916485 PMCID: PMC8038590 DOI: 10.3390/ijms22073769] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 11/16/2022] Open
Abstract
Pax3 and Pax7 are members of the Pax gene family which are essential for embryo and organ development. Both genes have been proved to be markers of muscle satellite cells and play key roles in the process of muscle growth and repair. Here, we identified two Pax3 genes (SsPax3a and SsPax3b) and two Pax7 genes (SsPax7a and SsPax7b) in a marine teleost, black rockfish (Sebastes schlegelii). Our results showed SsPax3 and SsPax7 marked distinct populations of muscle satellite cells, which originated from the multi-cell stage and somite stage, respectively. In addition, we constructed a muscle injury model to explore the function of these four genes during muscle repair. Hematoxylin–eosin (H–E) of injured muscle sections showed new-formed myofibers occurred at 16 days post-injury (dpi). ISH (in situ hybridization) analysis demonstrated that the expression level of SsPax3a and two SsPax7 genes increased gradually during 0–16 dpi and peaked at 16 dpi. Interestingly, SsPax3b showed no significant differences during the injury repair process, indicating that the satellite cells labeled by SsPax3b were not involved in muscle repair. These results imply that the muscle stem cell populations in teleosts are more complicated than in mammals. This lays the foundation for future studies on the molecular mechanism of indeterminant growth and muscle repair of large fish species.
Collapse
Affiliation(s)
- Mengya Wang
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
| | - Weihao Song
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
| | - Chaofan Jin
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
| | - Kejia Huang
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
| | - Qianwen Yu
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
| | - Jie Qi
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
| | - Quanqi Zhang
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
| | - Yan He
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (M.W.); (W.S.); (C.J.); (K.H.); (Q.Y.); (J.Q.); (Q.Z.)
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
- Correspondence:
| |
Collapse
|
35
|
A mechanical model of early somite segmentation. iScience 2021; 24:102317. [PMID: 33889816 PMCID: PMC8050378 DOI: 10.1016/j.isci.2021.102317] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 11/21/2022] Open
Abstract
Somitogenesis is often described using the clock-and-wavefront (CW) model, which does not explain how molecular signaling rearranges the pre-somitic mesoderm (PSM) cells into somites. Our scanning electron microscopy analysis of chicken embryos reveals a caudally-progressing epithelialization front in the dorsal PSM that precedes somite formation. Signs of apical constriction and tissue segmentation appear in this layer 3-4 somite lengths caudal to the last-formed somite. We propose a mechanical instability model in which a steady increase of apical contractility leads to periodic failure of adhesion junctions within the dorsal PSM and positions the future inter-somite boundaries. This model produces spatially periodic segments whose size depends on the speed of the activation front of contraction (F), and the buildup rate of contractility (Λ). The Λ/F ratio determines whether this mechanism produces spatially and temporally regular or irregular segments, and whether segment size increases with the front speed. Dorsal pre-somitic mesoderm of chicken embryos epithelializes before somite formation Dorsal epithelium shows signs of apical constriction and early segmentation A mechanical instability model can reproduce sequential segmentation A single ratio describes spatial and temporal patterns of segmentation
Collapse
|
36
|
Wu N, Li Y, He X, Lin J, Long D, Cheng X, Brand-Saberi B, Wang G, Yang X. Retinoic Acid Signaling Plays a Crucial Role in Excessive Caffeine Intake-Disturbed Apoptosis and Differentiation of Myogenic Progenitors. Front Cell Dev Biol 2021; 9:586767. [PMID: 33791291 PMCID: PMC8006404 DOI: 10.3389/fcell.2021.586767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/04/2021] [Indexed: 11/13/2022] Open
Abstract
Whether or not the process of somitogenesis and myogenesis is affected by excessive caffeine intake still remains ambiguous. In this study, we first showed that caffeine treatment results in chest wall deformities and simultaneously reduced mRNA expressions of genes involved in myogenesis in the developing chicken embryos. We then used embryo cultures to assess in further detail how caffeine exposure affects the earliest steps of myogenesis, and we demonstrated that the caffeine treatment suppressed somitogenesis of chicken embryos by interfering with the expressions of crucial genes modulating apoptosis, proliferation, and differentiation of myogenic progenitors in differentiating somites. These phenotypes were abrogated by a retinoic acid (RA) antagonist in embryo cultures, even at low caffeine doses in C2C12 cells, implying that excess RA levels are responsible for these phenotypes in cells and possibly in vivo. These findings highlight that excessive caffeine exposure is negatively involved in regulating the development of myogenic progenitors through interfering with RA signaling. The RA somitogenesis/myogenesis pathway might be directly impacted by caffeine signaling rather than reflecting an indirect effect of the toxicity of excess caffeine dosage.
Collapse
Affiliation(s)
- Nian Wu
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Yingshi Li
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Xiangyue He
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Department of Pathology, Medical School, Jinan University, Guangzhou, China
| | - Jiayi Lin
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Denglu Long
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Xin Cheng
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Guang Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Xuesong Yang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
37
|
Viaut C, Weldon S, Münsterberg A. Fine-tuning of the PAX-SIX-EYA-DACH network by multiple microRNAs controls embryo myogenesis. Dev Biol 2021; 469:68-79. [PMID: 33080252 DOI: 10.1016/j.ydbio.2020.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/06/2020] [Accepted: 10/14/2020] [Indexed: 01/27/2023]
Abstract
MicroRNAs (miRNAs), short non-coding RNAs, which act post-transcriptionally to regulate gene expression, are of widespread significance during development and disease, including muscle disease. Advances in sequencing technology and bioinformatics led to the identification of a large number of miRNAs in vertebrates and other species, however, for many of these miRNAs specific roles have not yet been determined. LNA in situ hybridisation has revealed expression patterns of somite-enriched miRNAs, here we focus on characterising the functions of miR-128. We show that antagomiR-mediated knockdown (KD) of miR-128 in developing chick somites has a negative impact on skeletal myogenesis. Computational analysis identified the transcription factor EYA4 as a candidate target consistent with the observation that miR-128 and EYA4 display similar expression profiles. Luciferase assays confirmed that miR-128 interacts with the EYA4 3'UTR. In vivo experiments also suggest that EYA4 is regulated by miR-128. EYA4 is a member of the PAX-SIX-EYA-DACH (PSED) network of transcription factors. Therefore, we identified additional candidate miRNA binding sites in the 3'UTR of SIX1/4, EYA1/2/3 and DACH1. Using the miRanda algorithm, we found sites for miR-128, as well as for other myogenic miRNAs, miR-1a, miR-206 and miR-133a, some of these were experimentally confirmed as functional miRNA target sites. Our results reveal that miR-128 is involved in regulating skeletal myogenesis by directly targeting EYA4 with indirect effects on other PSED members, including SIX4 and PAX3. Hence, the inhibitory effect on myogenesis observed after miR-128 knockdown was rescued by concomitant knockdown of PAX3. Moreover, we show that the PSED network of transcription factors is co-regulated by multiple muscle-enriched microRNAs.
Collapse
Affiliation(s)
- Camille Viaut
- School of Biological Sciences, Cell and Developmental Biology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Shannon Weldon
- School of Biological Sciences, Cell and Developmental Biology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrea Münsterberg
- School of Biological Sciences, Cell and Developmental Biology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
38
|
Kusakabe R, Higuchi S, Tanaka M, Kadota M, Nishimura O, Kuratani S. Novel developmental bases for the evolution of hypobranchial muscles in vertebrates. BMC Biol 2020; 18:120. [PMID: 32907560 PMCID: PMC7488077 DOI: 10.1186/s12915-020-00851-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Vertebrates are characterized by possession of hypobranchial muscles (HBMs). Cyclostomes, or modern jawless vertebrates, possess a rudimentary and superficial HBM lateral to the pharynx, whereas the HBM in jawed vertebrates is internalized and anteroposteriorly specified. Precursor cells of the HBM, marked by expression of Lbx1, originate from somites and undergo extensive migration before becoming innervated by the hypoglossal nerve. How the complex form of HBM arose in evolution is relevant to the establishment of the vertebrate body plan, but despite having long been assumed to be similar to that of limb muscles, modification of developmental mechanisms of HBM remains enigmatic. RESULTS Here we characterize the expression of Lbx genes in lamprey and hagfish (cyclostomes) and catshark (gnathostome; jawed vertebrates). We show that the expression patterns of the single cyclostome Lbx homologue, Lbx-A, do not resemble the somitic expression of mammalian Lbx1. Disruption of Lbx-A revealed that LjLbx-A is required for the formation of both HBM and body wall muscles, likely due to the insufficient extension of precursor cells rather than to hindered muscle differentiation. Both homologues of Lbx in the catshark were expressed in the somitic muscle primordia, unlike in amniotes. During catshark embryogenesis, Lbx2 is expressed in the caudal HBM as well as in the abdominal rectus muscle, similar to lamprey Lbx-A, whereas Lbx1 marks the rostral HBM and pectoral fin muscle. CONCLUSIONS We conclude that the vertebrate HBM primarily emerged as a specialized somatic muscle to cover the pharynx, and the anterior internalized HBM of the gnathostomes is likely a novelty added rostral to the cyclostome-like HBM, for which duplication and functionalization of Lbx genes would have been a prerequisite.
Collapse
Affiliation(s)
- Rie Kusakabe
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Shinnosuke Higuchi
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Department of Biology, Graduate School of Science, Kobe University, Kobe, 657-8501, Japan
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Masako Tanaka
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Mitsutaka Kadota
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, 650-0047, Japan
| | - Osamu Nishimura
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, 650-0047, Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, 650-0047, Japan
| |
Collapse
|
39
|
Verbeek RJ, Mulder PB, Sollie KM, van der Hoeven JH, den Dunnen WFA, Maurits NM, Sival DA. Development of muscle ultrasound density in healthy fetuses and infants. PLoS One 2020; 15:e0235836. [PMID: 32649730 PMCID: PMC7351181 DOI: 10.1371/journal.pone.0235836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/23/2020] [Indexed: 11/19/2022] Open
Abstract
Muscle ultrasound density (MUD) is a non-invasive parameter to indicate neuromuscular integrity in both children and adults. In healthy fetuses and infants, physiologic MUD values during development are still lacking. We therefore aimed to determine the physiologic, age-related MUD trend of biceps, quadriceps, tibialis anterior, hamstrings, gluteal and calf muscles, from pre- to the first year of postnatal life. To avoid a bias by pregnancy-related signal disturbances, we expressed fetal MUD as a ratio against bone ultrasound density. We used the full-term prenatal MUD ratio and the newborn postnatal MUD value as reference points, so that MUD development could be quantified from early pre- into postnatal life. Results: During the prenatal period, the total muscle group revealed a developmental MUD trend concerning a fetal increase in MUD-ratio from the 2nd trimester up to the end of the 3rd trimester [median increase: 27% (range 16-45), p < .001]. After birth, MUD-values increased up to the sixth month [median increase: 11% (range -7-27), p = 0.025] and stabilized thereafter. Additionally, there were also individual MUD characteristics per muscle group and developmental stage, such as relatively low MUD values of fetal hamstrings and high values of the paediatric gluteus muscles. These MUD trends are likely to concur with analogous developmentally, maturation-related alterations in the muscle water to peptide content ratios.
Collapse
Affiliation(s)
- Renate J. Verbeek
- Department of (Pediatric) Neurology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- * E-mail:
| | - Petra B. Mulder
- Department of Obstetrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Krystyna M. Sollie
- Department of Obstetrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johannes H. van der Hoeven
- Department of (Pediatric) Neurology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wilfred F. A. den Dunnen
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Natalia M. Maurits
- Department of (Pediatric) Neurology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Deborah A. Sival
- Department of Pediatrics, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, The Netherlands
| |
Collapse
|
40
|
Disabled-2: a positive regulator of the early differentiation of myoblasts. Cell Tissue Res 2020; 381:493-508. [PMID: 32607799 PMCID: PMC7431403 DOI: 10.1007/s00441-020-03237-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/05/2020] [Indexed: 11/25/2022]
Abstract
Dab2 is an adaptor protein and a tumor suppressor. Our previous study has found that Dab2 was expressed in early differentiating skeletal muscles in mouse embryos. In this study, we determined the role of Dab2 in the skeletal muscle differentiation using C2C12 myoblasts in vitro and Xenopus laevis embryos in vivo. The expression of Dab2 was increased in C2C12 myoblasts during the formation of myotubes in vitro. Knockdown of Dab2 expression in C2C12 myoblasts resulted in a reduction of myotube formation, whereas the myotube formation was enhanced upon overexpression of Dab2. Re-expression of Dab2 in C2C12 myoblasts with downregulated expression of Dab2 restored their capacity to form myotubes. Microarray profiling and subsequent network analyses on the 155 differentially expressed genes after Dab2 knockdown showed that Mef2c was an important myogenic transcription factor regulated by Dab2 through the p38 MAPK pathway. It was also involved in other pathways that are associated with muscular development and functions. In Xenopus embryos developed in vivo, XDab2 was expressed in the myotome of somites where various myogenic markers were also expressed. Knockdown of XDab2 expression with antisense morpholinos downregulated the expression of myogenic markers in somites. In conclusion, this study is the first to provide solid evidence to show that Dab2 is a positive regulator of the early myoblast differentiation.
Collapse
|
41
|
Scaal M. Development of the amniote ventrolateral body wall. Dev Dyn 2020; 250:39-59. [PMID: 32406962 DOI: 10.1002/dvdy.193] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
In vertebrates, the trunk consists of the musculoskeletal structures of the back and the ventrolateral body wall, which together enclose the internal organs of the circulatory, digestive, respiratory and urogenital systems. This review gives an overview on the development of the thoracic and abdominal wall during amniote embryogenesis. Specifically, I briefly summarize relevant historical concepts and the present knowledge on the early embryonic development of ribs, sternum, intercostal muscles and abdominal muscles with respect to anatomical bauplan, origin and specification of precursor cells, initial steps of pattern formation, and cellular and molecular regulation of morphogenesis.
Collapse
Affiliation(s)
- Martin Scaal
- Faculty of Medicine, Institute of Anatomy II, University of Cologne, Cologne, Germany
| |
Collapse
|
42
|
Hwang M, Lee EJ, Chung MJ, Park S, Jeong KS. Five transcriptional factors reprogram fibroblast into myogenic lineage cells via paraxial mesoderm stage. Cell Cycle 2020; 19:1804-1816. [PMID: 32579865 DOI: 10.1080/15384101.2020.1780384] [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] [Indexed: 01/01/2023] Open
Abstract
It is hard to supply satellite cells as a cell source for therapy of muscle degenerative disease since the sampling of muscle tissue is very invasive to a patient with muscular disease. Direct conversion allows us to get specific cell types by transduction of defined transcriptional factors. To induce myogenic direct conversion, we transduced five transcriptional factors including Pax3, Sox2, Klf4, c-Myc, and Esrrb into mouse embryonic fibroblasts. We found that the transduction of the five transcriptional factors induced myogenic direct conversion of fibroblast. We revealed that the transduced cells with the five transcriptional factors were converted to myogenic lineage cells through a paraxial mesoderm-like stage. The expression level of myogenic-related genes of the transduced cells gradually increased as the passage increased. The induced myogenic lineage cells differentiated into muscle fibers in virto and in vivo. The current study revealed that the five transcription factors generated myogenic lineage cells from fibroblast via a paraxial mesoderm stage. The induced myogenic lineage cells may have a potential being applied as cell source for degenerative muscle disease.
Collapse
Affiliation(s)
- Meeyul Hwang
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University , Daegu, Republic of Korea
| | - Eun-Joo Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University , Daegu, Republic of Korea
| | - Myung-Jin Chung
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University , Daegu, Republic of Korea.,Stem Cell Therapeutic Research Center, Kyungpook National University , Daegu, Republic of Korea
| | - SunYoung Park
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University , Daegu, Republic of Korea.,Stem Cell Therapeutic Research Center, Kyungpook National University , Daegu, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University , Daegu, Republic of Korea.,Stem Cell Therapeutic Research Center, Kyungpook National University , Daegu, Republic of Korea
| |
Collapse
|
43
|
Helmbacher F, Stricker S. Tissue cross talks governing limb muscle development and regeneration. Semin Cell Dev Biol 2020; 104:14-30. [PMID: 32517852 DOI: 10.1016/j.semcdb.2020.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022]
Abstract
For decades, limb development has been a paradigm of three-dimensional patterning. Moreover, as the limb muscles and the other tissues of the limb's musculoskeletal system arise from distinct developmental sources, it has been a prime example of integrative morphogenesis and cross-tissue communication. As the limbs grow, all components of the musculoskeletal system (muscles, tendons, connective tissue, nerves) coordinate their growth and differentiation, ultimately giving rise to a functional unit capable of executing elaborate movement. While the molecular mechanisms governing global three-dimensional patterning and formation of the skeletal structures of the limbs has been a matter of intense research, patterning of the soft tissues is less understood. Here, we review the development of limb muscles with an emphasis on their interaction with other tissue types and the instructive roles these tissues play. Furthermore, we discuss the role of adult correlates of these embryonic accessory tissues in muscle regeneration.
Collapse
Affiliation(s)
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany.
| |
Collapse
|
44
|
Yang J, Antin P, Berx G, Blanpain C, Brabletz T, Bronner M, Campbell K, Cano A, Casanova J, Christofori G, Dedhar S, Derynck R, Ford HL, Fuxe J, García de Herreros A, Goodall GJ, Hadjantonakis AK, Huang RYJ, Kalcheim C, Kalluri R, Kang Y, Khew-Goodall Y, Levine H, Liu J, Longmore GD, Mani SA, Massagué J, Mayor R, McClay D, Mostov KE, Newgreen DF, Nieto MA, Puisieux A, Runyan R, Savagner P, Stanger B, Stemmler MP, Takahashi Y, Takeichi M, Theveneau E, Thiery JP, Thompson EW, Weinberg RA, Williams ED, Xing J, Zhou BP, Sheng G. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2020; 21:341-352. [PMID: 32300252 PMCID: PMC7250738 DOI: 10.1038/s41580-020-0237-9] [Citation(s) in RCA: 1141] [Impact Index Per Article: 285.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
Epithelial–mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by ‘the EMT International Association’ (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT. In this Consensus Statement, the authors (on behalf of the EMT International Association) propose guidelines to define epithelial–mesenchymal transition, its phenotypic plasticity and the associated multiple intermediate epithelial–mesenchymal cell states. Clarification of nomenclature and definitions will help reduce misinterpretation of research data generated in different experimental model systems and promote cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Jing Yang
- Departments of Pharmacology and Pediatrics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| | - Parker Antin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Geert Berx
- Molecular and Cellular Oncology Lab, Department of Biomedical Molecular Biology, Ghent University, Cancer Research Institute Ghent (CRIG), VIB Center for Inflammation Research, Ghent, Belgium
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kyra Campbell
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield, UK
| | - Amparo Cano
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPAZ & Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Jordi Casanova
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology/Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain
| | | | - Shoukat Dedhar
- Department of Biochemistry and Molecular Biology, University of British Columbia and British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Rik Derynck
- Departments of Cell and Tissue Biology, and Anatomy, University of California at San Francisco, San Francisco, CA, USA
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jonas Fuxe
- Department of Laboratory Medicine (LABMED), Division of Pathology, Karolinska University Hospital and Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Antonio García de Herreros
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) and Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Gregory J Goodall
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruby Y J Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute for medical Research Israel-Canada and the Safra Center for Neurosciences, Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, MD Anderson Cancer Center, Houston, TX, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, an Alliance of SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Herbert Levine
- Department of Physics, Northeastern University, Boston, MA, USA
| | - Jinsong Liu
- Department of Anatomic Pathology, The Division of Pathology and Laboratory Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory D Longmore
- Department of Medicine (Oncology) and Department of Cell Biology and Physiology, ICCE Institute, Washington University, St. Louis, MO, USA
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK
| | - David McClay
- Department of Biology, Duke University, Durham, NC, USA
| | - Keith E Mostov
- Departments of Anatomy and Biochemistry/Biophysics, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Donald F Newgreen
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - M Angela Nieto
- Instituto de Neurociencias (CSIC-UMH) Avda Ramon y Cajal s/n, Sant Joan d´Alacant, Spain
| | - Alain Puisieux
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France.,Institut Curie, PSL Research University, Paris, France
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Pierre Savagner
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Ben Stanger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Japan
| | | | - Eric Theveneau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean Paul Thiery
- Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Guangzhou, China
| | - Erik W Thompson
- School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Department of Biology, MIT Ludwig Center for Molecular Oncology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Woolloongabba, QLD, Australia
| | - Jianhua Xing
- Department of Computational and Systems Biology and UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry and UK Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.
| | | |
Collapse
|
45
|
Nelemans BKA, Schmitz M, Tahir H, Merks RMH, Smit TH. Somite Division and New Boundary Formation by Mechanical Strain. iScience 2020; 23:100976. [PMID: 32222696 PMCID: PMC7109633 DOI: 10.1016/j.isci.2020.100976] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/30/2020] [Accepted: 03/09/2020] [Indexed: 02/04/2023] Open
Abstract
Somitogenesis, the primary segmentation of the vertebrate embryo, is associated with oscillating genes that interact with a wave of cell differentiation. The necessity of cell-matrix adherence and embryonic tension, however, suggests that mechanical cues are also involved. To explicitly investigate this, we applied surplus axial strain to live chick embryos. Despite substantial deformations, the embryos developed normally and somite formation rate was unaffected. Surprisingly, however, we observed slow cellular reorganizations of the most elongated somites into two or more well-shaped daughter somites. In what appeared to be a regular process of boundary formation, somites divided and fibronectin was deposited in between. Cell counts and morphology indicated that cells from the somitocoel underwent mesenchymal-epithelial transition; this was supported by a Cellular Potts model of somite division. Thus, although somitogenesis appeared to be extremely robust, we observed new boundary formation in existing somites and conclude that mechanical strain can be morphologically instructive. Live chick embryos develop normally under substantial axial strain (>50%) Mature somites divide into daughter somites, and fibronectin is deposited in between Mesenchymal cells from the somitocoel transition into epithelial border cells Mechanical strain can induce border formation and thus affect morphogenesis
Collapse
Affiliation(s)
- Ben K A Nelemans
- Department of Orthopaedic Surgery, Amsterdam University Medical Centres, Amsterdam Movement Sciences, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Manuel Schmitz
- Department of Orthopaedic Surgery, Amsterdam University Medical Centres, Amsterdam Movement Sciences, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Hannan Tahir
- Department of Orthopaedic Surgery, Amsterdam University Medical Centres, Amsterdam Movement Sciences, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Centrum Wiskunde & Informatica, Science Park 123, 1098 XG Amsterdam, the Netherlands
| | - Roeland M H Merks
- Centrum Wiskunde & Informatica, Science Park 123, 1098 XG Amsterdam, the Netherlands; Mathematical Institute Leiden, Leiden University, Niels Bohrweg 1, 2333 CA Leiden, the Netherlands
| | - Theodoor H Smit
- Department of Orthopaedic Surgery, Amsterdam University Medical Centres, Amsterdam Movement Sciences, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam University Medical Centres, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands.
| |
Collapse
|
46
|
Ahmad K, Shaikh S, Ahmad SS, Lee EJ, Choi I. Cross-Talk Between Extracellular Matrix and Skeletal Muscle: Implications for Myopathies. Front Pharmacol 2020; 11:142. [PMID: 32184725 PMCID: PMC7058629 DOI: 10.3389/fphar.2020.00142] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle (SM) comprises around 40% of total body weight and is among the most important plastic tissues, as it supports skeletal development, controls body temperature, and manages glucose levels. Extracellular matrix (ECM) maintains the integrity of SM, enables biochemical signaling, provides structural support, and plays a vital role during myogenesis. Several human diseases are coupled with dysfunctions of the ECM, and several ECM components are involved in disease pathologies that affect almost all organ systems. Thus, mutations in ECM genes that encode proteins and their transmembrane receptors can result in diverse SM diseases, a large proportion of which are types of fibrosis and muscular dystrophy. In this review, we present major ECM components of SMs related to muscle-associated diseases, and discuss two major ECM myopathies, namely, collagen myopathy and laminin myopathies, and their therapeutic managements. A comprehensive understanding of the mechanisms underlying these ECM-related myopathies would undoubtedly aid the discovery of novel treatments for these devastating diseases.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
47
|
Mechanism of muscle–tendon–bone complex development in the head. Anat Sci Int 2020; 95:165-173. [DOI: 10.1007/s12565-019-00523-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 12/24/2019] [Indexed: 12/11/2022]
|
48
|
Lewandowski D, Dubińska-Magiera M, Migocka-Patrzałek M, Niedbalska-Tarnowska J, Haczkiewicz-Leśniak K, Dzięgiel P, Daczewska M. Everybody wants to move-Evolutionary implications of trunk muscle differentiation in vertebrate species. Semin Cell Dev Biol 2019; 104:3-13. [PMID: 31759871 DOI: 10.1016/j.semcdb.2019.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
In our review we have completed current knowledge on myotomal myogenesis in model and non-model vertebrate species (fishes, amphibians, reptiles, birds and mammals) at morphological and molecular levels. Data obtained from these studies reveal distinct similarities and differences between amniote and anamniote species. Based on the available data, we decided to present evolutionary implications in vertebrate trunk muscle development. Despite the fact that in all vertebrates muscle fibres are multinucleated, the pathways leading to them vary between vertebrate taxa. In fishes during early myogenesis myoblasts differentiate into multinucleated lamellae or multinucleate myotubes. In amphibians, myoblasts fuse to form multinucleated myotubes or, bypassing fusion, directly differentiate into mononucleated myotubes. Furthermore, mononucleated myotubes were also observed during primary myogenesis in amniotes. The mononucleated state of myogenic cells could be considered as an old phylogenetic, plesiomorphic feature, whereas direct multinuclearity of myotubes has a synapomorphic character. On the other hand, the explanation of this phenomenon could also be linked to the environmental conditions in which animals develop. The similarities observed in vertebrate myogenesis might result from a conservative myogenic programme governed by the Pax3/Pax7 and myogenic regulatory factor (MRF) network, whereas differences in anamniotes and amniotes are established by spatiotemporal pattern expression of MRFs during muscle differentiation and/or environmental conditions.
Collapse
Affiliation(s)
- Damian Lewandowski
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wrocław, Poland.
| | - Magda Dubińska-Magiera
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wrocław, Poland
| | - Marta Migocka-Patrzałek
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wrocław, Poland
| | - Joanna Niedbalska-Tarnowska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wrocław, Poland; Laboratory of Molecular and Cellular Immunology, Department of Tumor Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland
| | | | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a, 50-368 Wrocław, Poland; Department of Physiotherapy, University School of Physical Education, Paderewskiego 35, 51-612 Wrocław, Poland
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wrocław, Poland
| |
Collapse
|
49
|
Yang Z, Song C, Jiang R, Huang Y, Lan X, Lei C, Chen H. Micro-Ribonucleic Acid-216a Regulates Bovine Primary Muscle Cells Proliferation and Differentiation via Targeting SMAD Nuclear Interacting Protein-1 and Smad7. Front Genet 2019; 10:1112. [PMID: 31798627 PMCID: PMC6865218 DOI: 10.3389/fgene.2019.01112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs), belonging to a class of evolutionarily conserved small noncoding RNA of ∼22 nucleotides, are widely involved in skeletal muscle growth and development by regulating gene expression at the post-transcriptional level. While the expression feature and underlying function of miR-216a in mammal skeletal muscle development, especially in cattle, remains to be further elucidated. The aim of this study was to investigate the function and mechanism of miR-216a during bovine primary muscle cells proliferation and differentiation. Herein, we found that the expression level of miR-216a both presented a downward trend during the proliferation and differentiation phases, which suggested that it might have a potential role in the development of bovine skeletal muscle. Functionally, during the cells proliferation phase, overexpression of miR-216a inhibited the expression of proliferation-related genes, reduced the cell proliferation status, and resulted in cells G1 phase arrest. In cells differentiation stages, overexpression of miR-216a suppressed myogenic maker genes mRNA, protein, and myotube formation. Mechanistically, we found that SNIP1 and smad7 were the directly targets of miR-216a in regulating bovine primary muscle cells proliferation and differentiation, respectively. Altogether, these findings suggested that miR-216a functions as a suppressive miRNA in development of bovine primary muscle cells via targeting SNIP1 and smad7.
Collapse
Affiliation(s)
- Zhaoxin Yang
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chengchuang Song
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rui Jiang
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yongzhen Huang
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xianyong Lan
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hong Chen
- Shaanxi Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
50
|
Matsutani K, Ikegami K, Aoyama H. An in vitro model of region-specific rib formation in chick axial skeleton: Intercellular interaction between somite and lateral plate cells. Mech Dev 2019; 159:103568. [PMID: 31493459 DOI: 10.1016/j.mod.2019.103568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 07/22/2019] [Accepted: 08/30/2019] [Indexed: 11/30/2022]
Abstract
The axial skeleton is divided into different regions based on its morphological features. In particular, in birds and mammals, ribs are present only in the thoracic region. The axial skeleton is derived from a series of somites. In the thoracic region of the axial skeleton, descendants of somites coherently penetrate into the somatic mesoderm to form ribs. In regions other than the thoracic, descendants of somites do not penetrate the somatic lateral plate mesoderm. We performed live-cell time-lapse imaging to investigate the difference in the migration of a somite cell after contact with the somatic lateral plate mesoderm obtained from different regions of anterior-posterior axis in vitro on cytophilic narrow paths. We found that a thoracic somite cell continues to migrate after contact with the thoracic somatic lateral plate mesoderm, whereas it ceases migration after contact with the lumbar somatic lateral plate mesoderm. This suggests that cell-cell interaction works as an important guidance cue that regulates migration of somite cells. We surmise that the thoracic somatic lateral plate mesoderm exhibits region-specific competence to allow penetration of somite cells, whereas the lumbosacral somatic lateral plate mesoderm repels somite cells by contact inhibition of locomotion. The differences in the behavior of the somatic lateral plate mesoderm toward somite cells may confirm the distinction between different regions of the axial skeleton.
Collapse
Affiliation(s)
- Kaoru Matsutani
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Koji Ikegami
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Hirohiko Aoyama
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; Department of Medical Science and Technology, Faculty of Health Sciences, Hiroshima International University, 555-36 Kurosegakuendai, Higashihiroshima City, Hiroshima 739-2695, Japan.
| |
Collapse
|