1
|
Lu C, Kang T, Zhang J, Yang K, Liu Y, Song K, Lin Q, Dixit D, Gimple RC, Zhang Q, Shi Z, Fan X, Wu Q, Li D, Shan D, Gao J, Gu D, You H, Li Y, Yang J, Zhao L, Qiu Z, Yang H, Zhao N, Gao W, Tao W, Lu Y, Chen Y, Ji J, Zhu Z, Kang C, Man J, Agnihotri S, Wang Q, Lin F, Qian X, Mack SC, Hu Z, Li C, Taylor MD, Liu N, Zhang N, Lu M, You Y, Rich JN, Zhang W, Wang X. Combined targeting of glioblastoma stem cells of different cellular states disrupts malignant progression. Nat Commun 2025; 16:2974. [PMID: 40140646 PMCID: PMC11947120 DOI: 10.1038/s41467-025-58366-5] [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: 03/26/2024] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor with intra-tumoral hierarchy of glioblastoma stem cells (GSCs). The heterogeneity of GSCs within GBM inevitably leads to treatment resistance and tumor recurrence. Molecular mechanisms of different cellular state GSCs remain unclear. Here, we find that classical (CL) and mesenchymal (MES) GSCs are enriched in reactive immune region and high CL-MES signature informs poor prognosis in GBM. Through integrated analyses of GSCs RNA sequencing and single-cell RNA sequencing datasets, we identify specific GSCs targets, including MEOX2 for the CL GSCs and SRGN for the MES GSCs. MEOX2-NOTCH and SRGN-NFκB axes play important roles in promoting proliferation and maintaining stemness and subtype signatures of CL and MES GSCs, respectively. In the tumor microenvironment, MEOX2 and SRGN mediate the resistance of CL and MES GSCs to macrophage phagocytosis. Using genetic and pharmacologic approaches, we identify FDA-approved drugs targeting MEOX2 and SRGN. Combined CL and MES GSCs targeting demonstrates enhanced efficacy, both in vitro and in vivo. Our results highlighted a therapeutic strategy for the elimination of heterogeneous GSCs populations through combinatorial targeting of MEOX2 and SRGN in GSCs.
Collapse
Affiliation(s)
- Chenfei Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Kang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Yang Liu
- Department of Pharmacology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Kefan Song
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiankun Lin
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Deobrat Dixit
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ryan C Gimple
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Qian Zhang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiulian Wu
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Daqi Li
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danyang Shan
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiancheng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danling Gu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao You
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangqing Li
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junlei Yang
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linjie Zhao
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Zhixin Qiu
- Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Gao
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Tao
- College of Biomedicine and Health & College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yingmei Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Chen
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Ji
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhe Zhu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sameer Agnihotri
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Lin
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Stephen C Mack
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhibin Hu
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chaojun Li
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Michael D Taylor
- Department of Pediatrics- Hematology/Oncology and Neurosurgery, Texas Children's Cancer Center, Hematology-Oncology Section, Baylor College of Medicine, Houston, Texas, USA
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ming Lu
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jeremy N Rich
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xiuxing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Cell Biology, National Health Commission Key Laboratory of Antibody Techniques, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
Rostamzadeh Mahdabi E, Esmailizadeh A, Han J, Wang M. Comparative Analysis of Runs of Homozygosity Islands in Indigenous and Commercial Chickens Revealed Candidate Loci for Disease Resistance and Production Traits. Vet Med Sci 2025; 11:e70074. [PMID: 39655377 PMCID: PMC11629026 DOI: 10.1002/vms3.70074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 08/08/2024] [Accepted: 09/20/2024] [Indexed: 12/13/2024] Open
Abstract
Runs of homozygosity (ROH) are contiguous stretches of identical genomic regions inherited from both parents. Assessment of ROH in livestock species contributes significantly to our understanding of genetic health, population genetic structure, selective pressure and conservation efforts. In this study, whole genome re-sequencing data from 140 birds of 10 Iranian indigenous chicken ecotypes, 3 commercial chicken breeds and 1 red junglefowl (RJF) population were used to investigate their population genetic structure, ROH characteristics (length and frequency) and genomic inbreeding coefficients (FROH). Additionally, we examined ROH islands for selection footprints in the indigenous chicken populations. Our results revealed distinct genetic backgrounds, among which the White Leghorn breed exhibited the greatest genetic distance from other populations, while the gamecock populations formed a separate cluster. We observed significant differences in ROH characteristics, in which the commercial breeds showed a higher number of ROH compared to indigenous chickens and red junglefowls. Short ROH ranging from 0.1 to 1 Mb were dominant among the populations. The Arian line had the highest mean length of ROH, while the White Leghorn breed showed the highest number of ROH. Among indigenous chickens, the Lari-Afghani ecotype exhibited the highest FROH, but the Sari inherited the richest genetic diversity. Interestingly, GGA16 carried no ROH in the red junglefowls, whereas GGA22 had the highest FROH across all populations, except in the Isfahan ecotype. We also identified ROH islands associated with genetic adaptations in indigenous ecotypes. These islands harboured immune-related genes contributing to disease resistance (TLR2, TICAM1, IL22RA1, NOS2, CCL20 and IFNLR1), heat tolerance and oxidative stress response (NFKB1, HSF4, OSGIN1 and BDNF), and muscle development, lipid metabolism and reproduction (MEOX2, CEBPB, CDS2 and GnRH-I). Overall, this study highlights the genetic potential of indigenous chickens to survive and adapt to their respective environments.
Collapse
Affiliation(s)
| | - Ali Esmailizadeh
- Department of Animal ScienceFaculty of AgricultureShahid Bahonar University of KermanKermanIran
- Key Laboratory of Genetic Evolution & Animal ModelsState Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Jianlin Han
- CAAS‐ILRI Joint Laboratory on Livestock and Forage Genetic ResourcesInstitute of Animal ScienceChinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Ming‐Shan Wang
- Key Laboratory of Genetic Evolution & Animal ModelsState Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| |
Collapse
|
3
|
Lipp SN, Jacobson KR, Colling HA, Tuttle TG, Miles DT, McCreery KP, Calve S. Mechanical loading is required for initiation of extracellular matrix deposition at the developing murine myotendinous junction. Matrix Biol 2023; 116:28-48. [PMID: 36709857 PMCID: PMC10218368 DOI: 10.1016/j.matbio.2023.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
The myotendinous junction (MTJ) contributes to the generation of motion by connecting muscle to tendon. At the adult MTJ, a specialized extracellular matrix (ECM) is thought to contribute to the mechanical integrity of the muscle-tendon interface, but the factors that influence MTJ formation during mammalian development are unclear. Here, we combined 3D imaging and proteomics with murine models in which muscle contractility and patterning are disrupted to resolve morphological and compositional changes in the ECM during MTJ development. We found that MTJ-specific ECM deposition can be initiated via static loading due to growth; however, it required cyclic loading to develop a mature morphology. Furthermore, the MTJ can mature without the tendon terminating into cartilage. Based on these results, we describe a model wherein MTJ development depends on mechanical loading but not insertion into an enthesis.
Collapse
Affiliation(s)
- Sarah N Lipp
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, United States; The Indiana University Medical Scientist/Engineer Training Program, Indianapolis, IN 46202, United States
| | - Kathryn R Jacobson
- Purdue University Interdisciplinary Life Science Program, 155 S. Grant Street, West Lafayette, IN 47907, United States
| | - Haley A Colling
- Department of Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder CO, 80309, United States
| | - Tyler G Tuttle
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309, United States
| | - Dalton T Miles
- Chemical and Biological Engineering, University of Colorado Boulder, 596 UCB, CO 80309, United States
| | - Kaitlin P McCreery
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309, United States
| | - Sarah Calve
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, United States; Purdue University Interdisciplinary Life Science Program, 155 S. Grant Street, West Lafayette, IN 47907, United States; Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309, United States.
| |
Collapse
|
4
|
Schulreich SM, Salamanca-Díaz DA, Zieger E, Calcino AD, Wanninger A. A mosaic of conserved and novel modes of gene expression and morphogenesis in mesoderm and muscle formation of a larval bivalve. ORG DIVERS EVOL 2022; 22:893-913. [PMID: 36398106 PMCID: PMC9649484 DOI: 10.1007/s13127-022-00569-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/26/2022] [Indexed: 10/17/2022]
Abstract
The mesoderm gives rise to several key morphological features of bilaterian animals including endoskeletal elements and the musculature. A number of regulatory genes involved in mesoderm and/or muscle formation (e.g., Brachyury (Bra), even-skipped (eve), Mox, myosin II heavy chain (mhc)) have been identified chiefly from chordates and the ecdysozoans Drosophila and Caenorhabditis elegans, but data for non-model protostomes, especially those belonging to the ecdysozoan sister clade, Lophotrochozoa (e.g., flatworms, annelids, mollusks), are only beginning to emerge. Within the lophotrochozoans, Mollusca constitutes the most speciose and diverse phylum. Interestingly, however, information on the morphological and molecular underpinnings of key ontogenetic processes such as mesoderm formation and myogenesis remains scarce even for prominent molluscan sublineages such as the bivalves. Here, we investigated myogenesis and developmental expression of Bra, eve, Mox, and mhc in the quagga mussel Dreissena rostriformis, an invasive freshwater bivalve and an emerging model in invertebrate evodevo. We found that all four genes are expressed during mesoderm formation, but some show additional, individual sites of expression during ontogeny. While Mox and mhc are involved in early myogenesis, eve is also expressed in the embryonic shell field and Bra is additionally present in the foregut. Comparative analysis suggests that Mox has an ancestral role in mesoderm and possibly muscle formation in bilaterians, while Bra and eve are conserved regulators of mesoderm development of nephrozoans (protostomes and deuterostomes). The fully developed Dreissena veliger larva shows a highly complex muscular architecture, supporting a muscular ground pattern of autobranch bivalve larvae that includes at least a velum muscle ring, three or four pairs of velum retractors, one or two pairs of larval retractors, two pairs of foot retractors, a pedal plexus, possibly two pairs of mantle retractors, and the muscles of the pallial line, as well as an anterior and a posterior adductor. As is typical for their molluscan kin, remodelling and loss of prominent larval features such as the velum musculature and various retractor systems appear to be also common in bivalves. Supplementary information The online version contains supplementary material available at 10.1007/s13127-022-00569-5.
Collapse
Affiliation(s)
- Stephan M. Schulreich
- Unit for Integrative Zoology, Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - David A. Salamanca-Díaz
- Unit for Integrative Zoology, Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Elisabeth Zieger
- Unit for Integrative Zoology, Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Andrew D. Calcino
- Unit for Integrative Zoology, Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Andreas Wanninger
- Unit for Integrative Zoology, Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| |
Collapse
|
5
|
Kokotović T, Lenartowicz EM, Langeslag M, Ciotu CI, Fell CW, Scaramuzza A, Fischer MJM, Kress M, Penninger JM, Nagy V. Transcription factor mesenchyme homeobox protein 2 (MEOX2) modulates nociceptor function. FEBS J 2022; 289:3457-3476. [PMID: 35029322 PMCID: PMC9306780 DOI: 10.1111/febs.16347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/30/2021] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
Abstract
Mesenchyme homeobox protein 2 (MEOX2) is a transcription factor involved in mesoderm differentiation, including development of bones, muscles, vasculature and dermatomes. We have previously identified dysregulation of MEOX2 in fibroblasts from Congenital Insensitivity to Pain patients, and confirmed that btn, the Drosophila homologue of MEOX2, plays a role in nocifensive responses to noxious heat stimuli. To determine the importance of MEOX2 in the mammalian peripheral nervous system, we used a Meox2 heterozygous (Meox2+/−) mouse model to characterise its function in the sensory nervous system, and more specifically, in nociception. MEOX2 is expressed in the mouse dorsal root ganglia (DRG) and spinal cord, and localises in the nuclei of a subset of sensory neurons. Functional studies of the mouse model, including behavioural, cellular and electrophysiological analyses, showed altered nociception encompassing impaired action potential initiation upon depolarisation. Mechanistically, we noted decreased expression of Scn9a and Scn11a genes encoding Nav1.7 and Nav1.9 voltage‐gated sodium channels respectively, that are crucial in subthreshold amplification and action potential initiation in nociceptors. Further transcriptomic analyses of Meox2+/− DRG revealed downregulation of a specific subset of genes including those previously associated with pain perception, such as PENK and NPY. Based on these observations, we propose a novel role of MEOX2 in primary afferent nociceptor neurons for the maintenance of a transcriptional programme required for proper perception of acute and inflammatory noxious stimuli.
Collapse
Affiliation(s)
- Tomislav Kokotović
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases Vienna Austria
- CeMM‐Research Center for Molecular Medicine of the Austrian Academy of Sciences Vienna Austria
- Department of Neurology Medical University of Vienna Austria
| | | | - Michiel Langeslag
- Department of Physiology and Medical Physics Institute of Physiology Medical University of Innsbruck Austria
- Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI) University of Innsbruck Austria
- Department of Pharmacology Medical University of Innsbruck Austria
| | - Cosmin I. Ciotu
- Institute of Physiology Medical University of Vienna Austria
| | - Christopher W. Fell
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases Vienna Austria
- CeMM‐Research Center for Molecular Medicine of the Austrian Academy of Sciences Vienna Austria
- Department of Neurology Medical University of Vienna Austria
| | - Angelica Scaramuzza
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases Vienna Austria
| | | | - Michaela Kress
- Department of Physiology and Medical Physics Institute of Physiology Medical University of Innsbruck Austria
| | - Josef M. Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences VBC – Vienna BioCenter Campus Vienna Austria
- Department of Medical Genetics Life Science Institute University of British Columbia Vancouver Canada
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases Vienna Austria
- CeMM‐Research Center for Molecular Medicine of the Austrian Academy of Sciences Vienna Austria
- Department of Neurology Medical University of Vienna Austria
| |
Collapse
|
6
|
Petrosino G, Ponte G, Volpe M, Zarrella I, Ansaloni F, Langella C, Di Cristina G, Finaurini S, Russo MT, Basu S, Musacchia F, Ristoratore F, Pavlinic D, Benes V, Ferrante MI, Albertin C, Simakov O, Gustincich S, Fiorito G, Sanges R. Identification of LINE retrotransposons and long non-coding RNAs expressed in the octopus brain. BMC Biol 2022; 20:116. [PMID: 35581640 PMCID: PMC9115989 DOI: 10.1186/s12915-022-01303-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/21/2022] [Indexed: 01/07/2023] Open
Abstract
Background Transposable elements (TEs) widely contribute to the evolution of genomes allowing genomic innovations, generating germinal and somatic heterogeneity, and giving birth to long non-coding RNAs (lncRNAs). These features have been associated to the evolution, functioning, and complexity of the nervous system at such a level that somatic retrotransposition of long interspersed element (LINE) L1 has been proposed to be associated to human cognition. Among invertebrates, octopuses are fascinating animals whose nervous system reaches a high level of complexity achieving sophisticated cognitive abilities. The sequencing of the genome of the Octopus bimaculoides revealed a striking expansion of TEs which were proposed to have contributed to the evolution of its complex nervous system. We recently found a similar expansion also in the genome of Octopus vulgaris. However, a specific search for the existence and the transcription of full-length transpositionally competent TEs has not been performed in this genus. Results Here, we report the identification of LINE elements competent for retrotransposition in Octopus vulgaris and Octopus bimaculoides and show evidence suggesting that they might be transcribed and determine germline and somatic polymorphisms especially in the brain. Transcription and translation measured for one of these elements resulted in specific signals in neurons belonging to areas associated with behavioral plasticity. We also report the transcription of thousands of lncRNAs and the pervasive inclusion of TE fragments in the transcriptomes of both Octopus species, further testifying the crucial activity of TEs in the evolution of the octopus genomes. Conclusions The neural transcriptome of the octopus shows the transcription of thousands of putative lncRNAs and of a full-length LINE element belonging to the RTE class. We speculate that a convergent evolutionary process involving retrotransposons activity in the brain has been important for the evolution of sophisticated cognitive abilities in this genus. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01303-5.
Collapse
Affiliation(s)
- Giuseppe Petrosino
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy.,Institute of Molecular Biology (IMB), Mainz, Germany
| | - Giovanna Ponte
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | - Massimiliano Volpe
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy.,Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152, Genova, Italy.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ilaria Zarrella
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | - Federico Ansaloni
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152, Genova, Italy
| | - Concetta Langella
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | - Giulia Di Cristina
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy.,Institute of Zoology, University of Cologne, Cologne, Germany
| | - Sara Finaurini
- Neurobiology Sector, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy
| | - Monia T Russo
- Department of Integrative Marine Ecology, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | - Swaraj Basu
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy.,Strand Life Sciences, Bengaluru, India
| | - Francesco Musacchia
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | - Filomena Ristoratore
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | - Dinko Pavlinic
- Scientific Core Facilities & Technologies, GeneCore, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117, Heidelberg, Germany.,Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Vladimir Benes
- Scientific Core Facilities & Technologies, GeneCore, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Maria I Ferrante
- Department of Integrative Marine Ecology, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy
| | | | - Oleg Simakov
- Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 9040495, Japan.,Department of Molecular Evolution and Development, Wien University, Althanstraße 14 (UZA I), 1090, Wien, Austria
| | - Stefano Gustincich
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152, Genova, Italy.,Neurobiology Sector, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy
| | - Graziano Fiorito
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy.
| | - Remo Sanges
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, SZN, 80121, Naples, Italy. .,Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152, Genova, Italy. .,Neurobiology Sector, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy.
| |
Collapse
|
7
|
Cai Y, Liu Y, Sun Y, Ren Y. Mesenchyme homeobox 2 has a cancer-inhibiting function in breast carcinoma via affection of the PI3K/AKT/mTOR and ERK1/2 pathways. Biochem Biophys Res Commun 2022; 593:20-27. [DOI: 10.1016/j.bbrc.2022.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/22/2022]
|
8
|
Li J, Glover JD, Zhang H, Peng M, Tan J, Mallick CB, Hou D, Yang Y, Wu S, Liu Y, Peng Q, Zheng SC, Crosse EI, Medvinsky A, Anderson RA, Brown H, Yuan Z, Zhou S, Xu Y, Kemp JP, Ho YYW, Loesch DZ, Wang L, Li Y, Tang S, Wu X, Walters RG, Lin K, Meng R, Lv J, Chernus JM, Neiswanger K, Feingold E, Evans DM, Medland SE, Martin NG, Weinberg SM, Marazita ML, Chen G, Chen Z, Zhou Y, Cheeseman M, Wang L, Jin L, Headon DJ, Wang S. Limb development genes underlie variation in human fingerprint patterns. Cell 2022; 185:95-112.e18. [PMID: 34995520 PMCID: PMC8740935 DOI: 10.1016/j.cell.2021.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/20/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Fingerprints are of long-standing practical and cultural interest, but little is known about the mechanisms that underlie their variation. Using genome-wide scans in Han Chinese cohorts, we identified 18 loci associated with fingerprint type across the digits, including a genetic basis for the long-recognized "pattern-block" correlations among the middle three digits. In particular, we identified a variant near EVI1 that alters regulatory activity and established a role for EVI1 in dermatoglyph patterning in mice. Dynamic EVI1 expression during human development supports its role in shaping the limbs and digits, rather than influencing skin patterning directly. Trans-ethnic meta-analysis identified 43 fingerprint-associated loci, with nearby genes being strongly enriched for general limb development pathways. We also found that fingerprint patterns were genetically correlated with hand proportions. Taken together, these findings support the key role of limb development genes in influencing the outcome of fingerprint patterning.
Collapse
Affiliation(s)
- Jinxi Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - James D Glover
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Haiguo Zhang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Meifang Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Jingze Tan
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Chandana Basu Mallick
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK; Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Dan Hou
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Yajun Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Sijie Wu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Yu Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Shijie C Zheng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Edie I Crosse
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Helen Brown
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Ziyu Yuan
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu 225326, PRC
| | - Shen Zhou
- Shanghai Foreign Language School, Shanghai 200083, PRC
| | - Yanqing Xu
- Forest Ridge School of the Sacred Heart, Bellevue, WA 98006, USA
| | - John P Kemp
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| | - Yvonne Y W Ho
- QIMR Berghofer Medical Rese Institute, Brisbane, QLD, Australia
| | - Danuta Z Loesch
- Psychology Department, La Trobe University, Melbourne, VIC, Australia
| | | | | | | | - Xiaoli Wu
- WeGene, Shenzhen, Guangdong 518040, PRC
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ruogu Meng
- Center for Data Science in Health and Medicine, Peking University, Beijing 100191, PRC
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, PRC
| | - Jonathan M Chernus
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Katherine Neiswanger
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Eleanor Feingold
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David M Evans
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Sarah E Medland
- QIMR Berghofer Medical Rese Institute, Brisbane, QLD, Australia
| | | | - Seth M Weinberg
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Anthropology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mary L Marazita
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA; Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gang Chen
- WeGene, Shenzhen, Guangdong 518040, PRC
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Yong Zhou
- Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PRC
| | - Michael Cheeseman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Lan Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai 200438, PRC.
| | - Denis J Headon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, PRC.
| |
Collapse
|
9
|
Schönrock A, Heinzelmann E, Steffl B, Demirdizen E, Narayanan A, Krunic D, Bähr M, Park JW, Schmidt C, Özduman K, Pamir MN, Wick W, Bestvater F, Weichenhan D, Plass C, Taranda J, Mall M, Turcan Ş. OUP accepted manuscript. Neuro Oncol 2022; 24:1911-1924. [PMID: 35468210 PMCID: PMC9629421 DOI: 10.1093/neuonc/noac110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive tumor that frequently exhibits gain of chromosome 7, loss of chromosome 10, and aberrantly activated receptor tyrosine kinase signaling pathways. Previously, we identified Mesenchyme Homeobox 2 (MEOX2), a gene located on chromosome 7, as an upregulated transcription factor in GBM. Overexpressed transcription factors can be involved in driving GBM. Here, we aimed to address the role of MEOX2 in GBM. METHODS Patient-derived GBM tumorspheres were used to constitutively knockdown or overexpress MEOX2 and subjected to in vitro assays including western blot to assess ERK phosphorylation. Cerebral organoid models were used to investigate the role of MEOX2 in growth initiation. Intracranial mouse implantation models were used to assess the tumorigenic potential of MEOX2. RNA-sequencing, ACT-seq, and CUT&Tag were used to identify MEOX2 target genes. RESULTS MEOX2 enhanced ERK signaling through a feed-forward mechanism. We identified Ser155 as a putative ERK-dependent phosphorylation site upstream of the homeobox-domain of MEOX2. S155A substitution had a major effect on MEOX2 protein levels and altered its subnuclear localization. MEOX2 overexpression cooperated with p53 and PTEN loss in cerebral organoid models of human malignant gliomas to induce cell proliferation. Using high-throughput genomics, we identified putative transcriptional target genes of MEOX2 in patient-derived GBM tumorsphere models and a fresh frozen GBM tumor. CONCLUSIONS We identified MEOX2 as an oncogenic transcription regulator in GBM. MEOX2 increases proliferation in cerebral organoid models of GBM and feeds into ERK signaling that represents a core signaling pathway in GBM.
Collapse
Affiliation(s)
| | | | | | | | - Ashwin Narayanan
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Damir Krunic
- Core Facility Unit Light Microscopy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marion Bähr
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Claudia Schmidt
- Core Facility Unit Light Microscopy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Koray Özduman
- Department of Neurosurgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - M Necmettin Pamir
- Department of Neurosurgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Bestvater
- Core Facility Unit Light Microscopy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julian Taranda
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Moritz Mall
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Şevin Turcan
- Corresponding Author: Şevin Turcan, PhD, Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, INF 460, Heidelberg, Germany ()
| |
Collapse
|
10
|
Diaz RE, Taylor-Diaz EA, Trainor PA, Diogo R, Molnar JL. Comparative development of limb musculature in phylogenetically and ecologically divergent lizards. Dev Dyn 2021; 251:1576-1612. [PMID: 34927301 DOI: 10.1002/dvdy.447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Squamate reptiles (lizards, snakes, and amphisbaenians) exhibit incredible diversity in their locomotion, behavior, morphology, and ecological breadth. Although they often are used as models of locomotor diversity, surprisingly little attention has been given to muscle development in squamate reptiles. In fact, the most detailed examination was conducted almost 80 years ago and solely focused on the proximal limb regions. Herein, we present forelimb and hindlimb muscle morphogenesis data for three lizard species with different locomotion and feeding strategies: the desert grassland whiptail lizard, the central bearded dragon, and the veiled chameleon. This study fills critical gaps in our understanding of muscle morphogenesis in squamate reptiles and presents a comparative and temporospatial analysis of muscle development. RESULTS Our results reveal a conserved pattern of early muscle development among lizards with different adult morphologies and ecologies. The variations that exist are concentrated in distal regions, particularly the specialized autopodia of chameleons, where differentiation of muscles associated with the digits is delayed. CONCLUSIONS The chameleon autopod provides an example of major evolutionary modifications to the skeleton with only minor disruption of the conserved order and pattern of limb muscle development. This robustness of muscle patterning facilitates the evolution of extreme yet functional phenotypes.
Collapse
Affiliation(s)
- Raul E Diaz
- Department of Biological Sciences, California State University, Los Angeles, California, USA.,Department of Herpetology, Natural History Museum of Los Angeles County, Los Angeles, California, USA
| | - Elizabeth A Taylor-Diaz
- Department of Biological Sciences, California State University, Los Angeles, California, USA
| | - Paul A Trainor
- Investigator, Stowers Institute for Medical Research, Kansas City, Missouri, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Rui Diogo
- Department of Anatomy, Howard University College of Medicine, Washington, District of Columbia, USA
| | - Julia L Molnar
- Department of Anatomy, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, New York, USA
| |
Collapse
|
11
|
Peralta-Arrieta I, Trejo-Villegas OA, Armas-López L, Ceja-Rangel HA, Ordóñez-Luna MDC, Pineda-Villegas P, González-López MA, Ortiz-Quintero B, Mendoza-Milla C, Zatarain-Barrón ZL, Arrieta O, Zúñiga J, Ávila-Moreno F. Failure to EGFR-TKI-based therapy and tumoural progression are promoted by MEOX2/GLI1-mediated epigenetic regulation of EGFR in the human lung cancer. Eur J Cancer 2021; 160:189-205. [PMID: 34844838 DOI: 10.1016/j.ejca.2021.10.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/26/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mesenchyme homeobox-2 (MEOX2)-mediated regulation of glioma-associated oncogene-1 (GLI1) has been associated with poor overall survival, conferring chemoresistance in lung cancer. However, the role of MEOX2/GLI1 in resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs)-based therapy remains unexplored in human lung cancer. METHODS Functional assays using genetic silencing strategy by short hairpin RNAs, as well as cytotoxic (tetrazolium dye MTT) and clonogenic assays, were performed to evaluate MEOX2/GLI1-induced malignancy capacity in lung cancer cells. Further analysis performed includes western blot, qPCR and ChIP-qPCR assays to identify whether MEOX2/GLI1 promote EGFR/AKT/ERK activation, as well as EGFR overexpression through epigenetic mechanisms. Finally, preclinical tumour progression in vivo and progression-free disease interval analyses in patients treated with EGFR-TKI were included. RESULTS Overexpressed MEOX2/GLI1 in both EGFR wild-type and EGFR/KRAS-mutated lung cancer cells were detected and involved in the activation/expression of EGFR/AKT/ERK biomarkers. In addition, MEOX2/GLI1 was shown to be involved in the increased proliferation of tumour cells and resistance capacity to cisplatin, EGFR-TKIs (erlotinib and AZD9291 'osimertinib'), AZD8542-SMO, and AZD6244-MEKK1/2. In addition, we identified that MEOX2/GLI1 promote lung tumour cells progression in vivo and are clinically associated with poorer progression-free disease intervals. Finally, both MEOX2 and GLI1 were detected to be epigenetically involved in EGFR expression by reducing both repressive markers polycomb-EZH2 and histone H3K27me3, but, particularly, increasing an activated histone profile H3K27Ac/H3K4me3 at EGFR-gene enhancer-promoter sequences that probably representing a novel EGFR-TKI-based therapy resistance mechanism. CONCLUSION MEOX2/GLI1 promote resistance to cisplatin and EGFR-TKI-based therapy in lung cancer cells, modulating EGFR/AKT/ERK signalling pathway activation, as well as inducing an aberrant epigenetic modulation of the EGFR-gene expression in human lung cancer.
Collapse
Affiliation(s)
- Irlanda Peralta-Arrieta
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Octavio A Trejo-Villegas
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Leonel Armas-López
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Hugo A Ceja-Rangel
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - María Del Carmen Ordóñez-Luna
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Priscila Pineda-Villegas
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico.
| | - Marco A González-López
- Unidad Funcional de Oncología Torácica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Blanca Ortiz-Quintero
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico.
| | - Criselda Mendoza-Milla
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico.
| | - Zyanya L Zatarain-Barrón
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Ciudad de México, Mexico.
| | - Oscar Arrieta
- Unidad Funcional de Oncología Torácica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Joaquín Zúñiga
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Ciudad de México, Mexico.
| | - Federico Ávila-Moreno
- Universidad Nacional Autónoma de México (UNAM), Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores (FES) Iztacala, Tlalnepantla de Baz, 54090, Estado de México, Mexico; Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calz de Tlalpan, 14080, Ciudad de México, Mexico.
| |
Collapse
|
12
|
Rostamzadeh Mahdabi E, Esmailizadeh A, Ayatollahi Mehrgardi A, Asadi Fozi M. A genome-wide scan to identify signatures of selection in two Iranian indigenous chicken ecotypes. Genet Sel Evol 2021; 53:72. [PMID: 34503452 PMCID: PMC8428137 DOI: 10.1186/s12711-021-00664-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background Various regions of the chicken genome have been under natural and artificial selection for thousands of years. The substantial diversity that exits among chickens from different geographic regions provides an excellent opportunity to investigate the genomic regions under selection which, in turn, will increase our knowledge about the mechanisms that underlie chicken diversity and adaptation. Several statistics have been developed to detect genomic regions that are under selection. In this study, we applied approaches based on differences in allele or haplotype frequencies (FST and hapFLK, respectively) between populations, differences in long stretches of consecutive homozygous sequences (ROH), and differences in allele frequencies within populations (composite likelihood ratio (CLR)) to identify inter- and intra-populations traces of selection in two Iranian indigenous chicken ecotypes, the Lari fighting chicken and the Khazak or creeper (short-leg) chicken. Results Using whole-genome resequencing data of 32 individuals from the two chicken ecotypes, approximately 11.9 million single nucleotide polymorphisms (SNPs) were detected and used in genomic analyses after quality processing. Examination of the distribution of ROH in the two populations indicated short to long ROH, ranging from 0.3 to 5.4 Mb. We found 90 genes that were detected by at least two of the four applied methods. Gene annotation of the detected putative regions under selection revealed candidate genes associated with growth (DCN, MEOX2 and CACNB1), reproduction (ESR1 and CALCR), disease resistance (S1PR1, ALPK1 and MHC-B), behavior pattern (AGMO, GNAO1 and PSEN1), and morphological traits (IHH and NHEJ1). Conclusions Our findings show that these two phenotypically different indigenous chicken populations have been under selection for reproduction, immune, behavioral, and morphology traits. The results illustrate that selection can play an important role in shaping signatures of differentiation across the genomic landscape of two chicken populations. Supplementary Information The online version contains supplementary material available at 10.1186/s12711-021-00664-9.
Collapse
Affiliation(s)
- Elaheh Rostamzadeh Mahdabi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran
| | - Ahmad Ayatollahi Mehrgardi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran
| | - Masood Asadi Fozi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran.
| |
Collapse
|
13
|
Sachslehner A, Zieger E, Calcino A, Wanninger A. HES and Mox genes are expressed during early mesoderm formation in a mollusk with putative ancestral features. Sci Rep 2021; 11:18030. [PMID: 34504115 PMCID: PMC8429573 DOI: 10.1038/s41598-021-96711-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022] Open
Abstract
The mesoderm is considered the youngest of the three germ layers. Although its morphogenesis has been studied in some metazoans, the molecular components underlying this process remain obscure for numerous phyla including the highly diverse Mollusca. Here, expression of Hairy and enhancer of split (HES), Mox, and myosin heavy chain (MHC) was investigated in Acanthochitona fascicularis, a representative of Polyplacophora with putative ancestral molluscan features. While AfaMHC is expressed throughout myogenesis, AfaMox1 is only expressed during early stages of mesodermal band formation and in the ventrolateral muscle, an autapomorphy of the polyplacophoran trochophore. Comparing our findings to previously published data across Metazoa reveals Mox expression in the mesoderm in numerous bilaterians including gastropods, polychaetes, and brachiopods. It is also involved in myogenesis in molluscs, annelids, tunicates, and craniates, suggesting a dual role of Mox in mesoderm and muscle formation in the last common bilaterian ancestor. AfaHESC2 is expressed in the ectoderm of the polyplacophoran gastrula and later in the mesodermal bands and in putative neural tissue, whereas AfaHESC7 is expressed in the trochoblasts of the gastrula and during foregut formation. This confirms the high developmental variability of HES gene expression and demonstrates that Mox and HES genes are pleiotropic.
Collapse
Affiliation(s)
- Attila Sachslehner
- Department of Evolutionary Biology, Unit for Integrative Zoology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Elisabeth Zieger
- Department of Evolutionary Biology, Unit for Integrative Zoology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Andrew Calcino
- Department of Evolutionary Biology, Unit for Integrative Zoology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Andreas Wanninger
- Department of Evolutionary Biology, Unit for Integrative Zoology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria.
| |
Collapse
|
14
|
Wang Z, Yang H, Zhang R, Luo B, Xu B, Zhu Z, Lin P. MEOX2 serves as a novel biomarker associated with macrophage infiltration in oesophageal squamous cell carcinoma and other digestive system carcinomas. Autoimmunity 2021; 54:373-383. [PMID: 34160343 DOI: 10.1080/08916934.2021.1919880] [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]
Abstract
BACKGROUND Oesophageal squamous cell carcinoma (ESCC) is a malignant tumour of the digestive system that is associated with high morbidity and mortality rates worldwide. With the increased use of immunotherapy in cancer treatment, there is an urgent need to elucidate the immune-related mechanisms in ESCC and other digestive system carcinomas. METHODS In our study, single-sample gene set enrichment analysis (ssGSEA) was first performed to analyse the expression profile downloaded from the NCBI Gene Expression Omnibus (GEO) database. Then, via a series of bioinformatic analyses, including the Mann-Whitney test, weighted gene co-expression network analysis (WGCNA), functional enrichment analysis and differentially expressed genes (DEGs) analysis, we identified target immunocytes and related genes. Finally, we validated the results with the TIMER database. RESULTS Our analyses showed that the numbers of infiltrating macrophages were obviously higher in advanced stages in ESCC compared with other types of immunocytes. MEOX2 was identified as a biomarker correlated with macrophage infiltration in ESCC and other types of digestive system carcinomas. And MEOX2 expression was strongly associated with the mRNA expression of colony-stimulating factor 1 (CSF-1) and CSF-1 receptor (CSF-1R) in these kinds of carcinomas. CONCLUSION We speculated that MEOX2 could facilitate macrophage infiltration via CSF-1/CSF-1R signalling in ESCC and other kinds of digestive system carcinomas, and MEOX2 might serve as a novel target in prospective tumour immunotherapies.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| | - Han Yang
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| | - Rusi Zhang
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| | - Bin Luo
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| | - Bingchen Xu
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| | - Zhihua Zhu
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| | - Peng Lin
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, People's Republic of China
| |
Collapse
|
15
|
Andrikou C, Hejnol A. FGF signaling acts on different levels of mesoderm development within Spiralia. Development 2021; 148:264929. [PMID: 33999997 PMCID: PMC8180254 DOI: 10.1242/dev.196089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/08/2021] [Indexed: 01/23/2023]
Abstract
FGF signaling is involved in mesoderm induction in members of deuterostomes (e.g. tunicates, hemichordates), but not in flies and nematodes, in which it has a role in mesoderm patterning and migration. However, we need comparable studies in other protostome taxa in order to decipher whether this mesoderm-inducing function of FGF extends beyond the lineage of deuterostomes. Here, we investigated the role of FGF signaling in mesoderm development in three species of lophophorates, a clade within the protostome group Spiralia. Our gene expression analyses show that the mesodermal molecular patterning is conserved between brachiopods and phoronids, but the spatial and temporal recruitment of transcription factors differs significantly. Moreover, the use of the inhibitor SU5402 demonstrates that FGF signaling is involved in different steps of mesoderm development, as well as in morphogenetic movements of gastrulation and axial elongation. Our findings suggest that the mesoderm-inducing role of FGF extends beyond the group of deuterostomes.
Collapse
Affiliation(s)
- Carmen Andrikou
- University of Bergen, Department of Biological Sciences, Thormøhlensgate 55, 5006 Bergen, Norway.,Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006 Bergen, Norway
| | - Andreas Hejnol
- University of Bergen, Department of Biological Sciences, Thormøhlensgate 55, 5006 Bergen, Norway.,Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006 Bergen, Norway
| |
Collapse
|
16
|
Gasparyan M, Lo MC, Jiang H, Lin CC, Sun D. Combined p53- and PTEN-deficiency activates expression of mesenchyme homeobox 1 (MEOX1) required for growth of triple-negative breast cancer. J Biol Chem 2020; 295:12188-12202. [PMID: 32467227 PMCID: PMC7443492 DOI: 10.1074/jbc.ra119.010710] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 05/02/2020] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive cancer subtype for which effective therapies are unavailable. TNBC has a high frequency of tumor protein p53 (Tp53/p53)- and phosphatase and tensin homolog (PTEN) deficiencies, and combined p53- and PTEN-deficiency is associated with poor prognosis and poor response to anticancer therapies. In this study, we discovered that combined p53- and PTEN-deficiency in TNBC activates expression of the transcription factor mesenchyme homeobox 1 (MEOX1). We found that MEOX1 is expressed only in TNBC cells with frequent deficiencies in p53 and PTEN, and that its expression is undetectable in luminal A, luminal B, and HER2+ subtypes, as well as in normal breast cells with wild-type (WT) p53 and PTEN. Notably, siRNA knockdown of both p53 and PTEN activated MEOX1 expression in breast cancer cells, whereas individual knockdowns of either p53 or PTEN had only minimal effects on MEOX1 expression. MEOX1 knockdown abolished cell proliferation of p53- and PTEN-deficient TNBC in vitro and inhibited tumor growth in vivo, but had no effect on the proliferation of luminal and HER2+ cancer cells and normal breast cells. RNA-Seq and immunoblotting analyses showed that MEOX1 knockdown decreased expression of tyrosine kinase 2 (TYK2), signal transducer and activator of transcription 5B (STAT5B), and STAT6 in p53- and PTEN-deficient TNBC cells. These results reveal the effects of combined p53- and PTEN-deficiency on MEOX1 expression and TNBC cell proliferation, suggesting that MEOX1 may serve as a potential therapeutic target for managing p53- and PTEN-deficient TNBC.
Collapse
Affiliation(s)
- Mari Gasparyan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Miao-Chia Lo
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Chang-Ching Lin
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA; Chemical Biology Program, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
17
|
Understanding paraxial mesoderm development and sclerotome specification for skeletal repair. Exp Mol Med 2020; 52:1166-1177. [PMID: 32788657 PMCID: PMC8080658 DOI: 10.1038/s12276-020-0482-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022] Open
Abstract
Pluripotent stem cells (PSCs) are attractive regenerative therapy tools for skeletal tissues. However, a deep understanding of skeletal development is required in order to model this development with PSCs, and for the application of PSCs in clinical settings. Skeletal tissues originate from three types of cell populations: the paraxial mesoderm, lateral plate mesoderm, and neural crest. The paraxial mesoderm gives rise to the sclerotome mainly through somitogenesis. In this process, key developmental processes, including initiation of the segmentation clock, formation of the determination front, and the mesenchymal–epithelial transition, are sequentially coordinated. The sclerotome further forms vertebral columns and contributes to various other tissues, such as tendons, vessels (including the dorsal aorta), and even meninges. To understand the molecular mechanisms underlying these developmental processes, extensive studies have been conducted. These studies have demonstrated that a gradient of activities involving multiple signaling pathways specify the embryonic axis and induce cell-type-specific master transcription factors in a spatiotemporal manner. Moreover, applying the knowledge of mesoderm development, researchers have attempted to recapitulate the in vivo development processes in in vitro settings, using mouse and human PSCs. In this review, we summarize the state-of-the-art understanding of mesoderm development and in vitro modeling of mesoderm development using PSCs. We also discuss future perspectives on the use of PSCs to generate skeletal tissues for basic research and clinical applications. A deeper understanding of skeletal tissue development and improvements in tissue engineering will help pluripotent stem cell (PSC) therapies to reach their full potential for skeletal repair. The paraxial mesoderm, an embryonic germ layer, is crucial to the formation of healthy axial skeleton. Shoichiro Tani at the University of Tokyo, Japan, and co-workers reviewed current understanding of paraxial mesoderm development and studies involving in vitro PSC skeletal modeling. The formation of the paraxial mesoderm and associated connective tissues comprises multiple stages, and studies in vertebrate embryos have uncovered critical signaling pathways and cellular components important to PSC modeling. Although many individual cellular components can now be modeled, it remains challenging to recreate three-dimensional skeletal tissues. Such an achievement would facilitate a functioning model of bone metabolism, the next step in achieving skeletal regeneration.
Collapse
|
18
|
Yin H, He H, Cao X, Shen X, Han S, Cui C, Zhao J, Wei Y, Chen Y, Xia L, Wang Y, Li D, Zhu Q. MiR-148a-3p Regulates Skeletal Muscle Satellite Cell Differentiation and Apoptosis via the PI3K/AKT Signaling Pathway by Targeting Meox2. Front Genet 2020; 11:512. [PMID: 32582277 PMCID: PMC7287179 DOI: 10.3389/fgene.2020.00512] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
As bioinformatic approaches have been developed, it has been demonstrated that microRNAs (miRNAs) are involved in the formation of muscles and play important roles in regulation of muscle cell proliferation and differentiation. Previously, it has been demonstrated that miR-148a-3p is one of the most abundant miRNAs in chicken skeletal muscle. Here, we build on that work and demonstrate that miR-148a-3p is important in the control of differentiation of chicken skeletal muscle satellite cells (SMSCs). Elevated expression of miR-148a-3p significantly promoted differentiation and inhibited apoptosis of SMSCs but did not affect proliferation. Furthermore, it was observed that the mesenchyme homeobox 2 (Meox2) is a target gene of miR-148a-3p and that miR-148a-3p can down-regulate expression of Meox2, which promote differentiation of SMSCs and suppress apoptosis. Furthermore, miR-148a-3p overexpression encouraged activation of the PI3K/AKT signaling pathway, which could be recovered by overexpression of Meox2. Overall, these findings suggest that microRNA-148a-3p is a potent promoter of myogenesis via direct targeting of Meox2 and increase of the PI3K/AKT signaling pathway in chicken SMSCs.
Collapse
Affiliation(s)
- Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Haorong He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xinao Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiaoxu Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shunshun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Can Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jing Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yuanhang Wei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yuqi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Lu Xia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
19
|
Skuplik I, Cobb J. Animal Models for Understanding Human Skeletal Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:157-188. [DOI: 10.1007/978-981-15-2389-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
20
|
Buchanan RA, Foley KE, Pepper KW, Reagan AM, Keezer KJ, Hewes AA, Diemler CA, Preuss C, Soto I, John SWM, Howell GR. Meox2 Haploinsufficiency Accelerates Axonal Degeneration in DBA/2J Glaucoma. Invest Ophthalmol Vis Sci 2019; 60:3283-3296. [PMID: 31369031 PMCID: PMC6676925 DOI: 10.1167/iovs.18-26126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Glaucoma is a complex disease with major risk factors including advancing age and increased intraocular pressure (IOP). Dissecting these earliest events will likely identify new avenues for therapeutics. Previously, we performed transcriptional profiling in DBA/2J (D2) mice, a widely used mouse model relevant to glaucoma. Here, we use these data to identify and test regulators of early gene expression changes in DBA/2J glaucoma. Methods Upstream regulator analysis (URA) in Ingenuity Pathway Analysis was performed to identify potential master regulators of differentially expressed genes. The function of one putative regulator, mesenchyme homeobox 2 (Meox2), was tested using a combination of genetic, biochemical, and immunofluorescence approaches. Results URA identified Meox2 as a potential regulator of early gene expression changes in the optic nerve head (ONH) of DBA/2J mice. Meox2 haploinsufficiency did not affect the characteristic diseases of the iris or IOP elevation seen in DBA/2J mice but did cause a significant increase in the numbers of eyes with axon damage compared to controls. While young mice appeared normal, aged Meox2 haploinsufficient DBA/2J mice showed a 44% reduction in MEOX2 protein levels. This correlated with modulation of age- and disease-specific vascular and myeloid alterations. Conclusions Our data support a model whereby Meox2 controls IOP-dependent vascular remodeling and neuroinflammation to promote axon survival. Promoting these earliest responses prior to IOP elevation may be a viable neuroprotective strategy to delay or prevent human glaucoma.
Collapse
Affiliation(s)
| | - Kate E Foley
- The Jackson Laboratory, Bar Harbor, Maine, United States.,Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States
| | | | | | - Kelly J Keezer
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | - Amanda A Hewes
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | - Cory A Diemler
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | | | - Ileana Soto
- The Jackson Laboratory, Bar Harbor, Maine, United States.,Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States.,Department of Biomedical and Translational Sciences, Rowan University, Glassboro, New Jersey, United States
| | - Simon W M John
- The Jackson Laboratory, Bar Harbor, Maine, United States.,Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States.,The Howard Hughes Medical Institute, Bar Harbor, Maine, United States.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, United States
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, Maine, United States.,Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, United States
| |
Collapse
|
21
|
Xenopus SOX5 enhances myogenic transcription indirectly through transrepression. Dev Biol 2018; 442:262-275. [PMID: 30071218 DOI: 10.1016/j.ydbio.2018.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/16/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
Abstract
In anamniotes, somite compartimentalization in the lateral somitic domain leads simultaneously to myotome and dermomyotome formation. In the myotome, Xenopus Sox5 is co-expressed with Myod1 in the course of myogenic differentiation. Here, we studied the function of Sox5 using a Myod1-induced myogenic transcription assay in pluripotent cells of animal caps. We found that Sox5 enhances myogenic transcription of muscle markers Des, Actc1, Ckm and MyhE3. The use of chimeric transactivating or transrepressive Sox5 proteins indicates that Sox5 acts as a transrepressor and indirectly stimulates myogenic transcription except for the slow muscle-specific genes Myh7L, Myh7S, Myl2 and Tnnc1. We showed that this role is shared by Sox6, which is structurally similar to Sox5, both belonging to the SoxD subfamily of transcription factors. Moreover, Sox5 can antagonize the inhibitory function of Meox2 on myogenic differentiation. Meox2 which is a dermomyotome marker, represses myogenic transcription in Myod-induced myogenic transcription assay and in Nodal5-induced mesoderm from animal cap assay. The inhibitory function of Meox2 and the pro-myogenic function of Sox5 were confirmed during Xenopus normal development by the use of translation-blocking oligomorpholinos and dexamethasone inducible chimeric Sox5 and Meox2 proteins. We have therefore identified a new function for SoxD proteins in muscle cells, which can indirectly enhance myogenic transcription through transrepression, in addition to the previously identified function as a direct repressor of slow muscle-specific genes.
Collapse
|
22
|
Adachi N, Pascual-Anaya J, Hirai T, Higuchi S, Kuroda S, Kuratani S. Stepwise participation of HGF/MET signaling in the development of migratory muscle precursors during vertebrate evolution. ZOOLOGICAL LETTERS 2018; 4:18. [PMID: 29946484 PMCID: PMC6004694 DOI: 10.1186/s40851-018-0094-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/11/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND The skeletal musculature of gnathostomes, which is derived from embryonic somites, consists of epaxial and hypaxial portions. Some hypaxial muscles, such as tongue and limb muscles, undergo de-epithelialization and migration during development. Delamination and migration of these myoblasts, or migratory muscle precursors (MMPs), is generally thought to be regulated by hepatocyte growth factor (HGF) and receptor tyrosine kinase (MET) signaling. However, the prevalence of this mechanism and the expression patterns of the genes involved in MMP development across different vertebrate species remain elusive. RESULTS We performed a comparative analysis of Hgf and Met gene expression in several vertebrates, including mouse, chicken, dogfish (Scyliorhinus torazame), and lamprey (Lethenteron camtschaticum). While both Hgf and Met were expressed during development in the mouse tongue muscle, and in limb muscle formation in the mouse and chicken, we found no clear evidence for the involvement of HGF/MET signaling in MMP development in shark or lamprey embryos. CONCLUSIONS Our results indicate that the expressions and functions of both Hgf and Met genes do not represent shared features of vertebrate MMPs, suggesting a stepwise participation of HGF/MET signaling in MMP development during vertebrate evolution.
Collapse
Affiliation(s)
- Noritaka Adachi
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Present address: Aix-Marseille Université, CNRS, IBDM UMR 7288, 13288 Marseille, France
| | - Juan Pascual-Anaya
- Laboratory for Evolutionary Morphology, RIKEN Cluster for Pioneering Research, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
| | - Tamami Hirai
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Laboratory for Evolutionary Morphology, RIKEN Cluster for Pioneering Research, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
| | - Shinnosuke Higuchi
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Laboratory for Evolutionary Morphology, RIKEN Cluster for Pioneering Research, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Department of Biology, Graduate School of Science, Kobe University, Kobe, 657-8501 Japan
| | - Shunya Kuroda
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Laboratory for Evolutionary Morphology, RIKEN Cluster for Pioneering Research, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Department of Biology, Graduate School of Science, Kobe University, Kobe, 657-8501 Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
- Laboratory for Evolutionary Morphology, RIKEN Cluster for Pioneering Research, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047 Japan
| |
Collapse
|
23
|
Li Y, Jiang Q, Ding Z, Liu G, Yu P, Jiang G, Yu Z, Yang C, Qian J, Jiang H, Zou Y. Identification of a Common Different Gene Expression Signature in Ischemic Cardiomyopathy. Genes (Basel) 2018; 9:genes9010056. [PMID: 29361784 PMCID: PMC5793207 DOI: 10.3390/genes9010056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 01/25/2023] Open
Abstract
The molecular mechanisms underlying the development of ischemic cardiomyopathy (ICM) remain poorly understood. Gene expression profiling is helpful to discover the molecular changes taking place in ICM. The aim of this study was to identify the genes that are significantly changed during the development of heart failure caused by ICM. The differentially expressed genes (DEGs) were identified from 162 control samples and 227 ICM patients. PANTHER was used to perform gene ontology (GO), and Reactome for pathway enrichment analysis. A protein–protein interaction network was established using STRING and Cytoscape. A further validation was performed by real-time polymerase chain reaction (RT-PCR). A total of 255 common DEGs was found. Gene ontology, pathway enrichment, and protein–protein interaction analysis showed that nucleic acid-binding proteins, enzymes, and transcription factors accounted for a great part of the DEGs, while immune system signaling and cytokine signaling displayed the most significant changes. Furthermore, seven hub genes and nine transcription factors were identified. Interestingly, the top five upregulated DEGs were located on chromosome Y, and four of the top five downregulated DEGs were involved in immune and inflammation signaling. Further, the top DEGs were validated by RT-PCR in human samples. Our study explored the possible molecular mechanisms of heart failure caused by ischemic heart disease.
Collapse
Affiliation(s)
- Yana Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Qiu Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Guijian Liu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Peng Yu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Guoliang Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Ziqing Yu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Chunjie Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Hong Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Shanghai Institute of clinical bioinformatics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
- Institutes of Biomedical Sciences, Fudan University, 130 Dong'an Road, Shanghai 200032, China.
| |
Collapse
|
24
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
25
|
Kitazawa M, Tamura M, Kaneko-Ishino T, Ishino F. Severe damage to the placental fetal capillary network causes mid- to late fetal lethality and reduction in placental size in Peg11/Rtl1 KO mice. Genes Cells 2017; 22:174-188. [PMID: 28111885 DOI: 10.1111/gtc.12465] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/06/2016] [Indexed: 11/29/2022]
Abstract
Paternally expressed 11/Retrotransposon-like 1 (Peg11/Rtl1) knockout (KO) mice show mid- to late fetal lethality or late fetal growth retardation associated with frequent neonatal lethality. The lethal phenotype is largely dependent on genetic background and becomes more severe with each succeeding generation in the course of backcross experiments to C57BL/6 (B6). We previously suggested that these lethal and growth phenotypes in the fetal stages were due to severe defects in placental fetal capillaries in the labyrinth layer. In this study, we re-examined KO fetuses and placentas and confirmed that the severe clogging of fetal capillaries was associated with KO fetuses showing mid-fetal lethality with internal bleeding. Importantly, the basal region of the fetal capillary network was specifically damaged, also leading to poor expansion of the labyrinth layer and placental size reduction in the later stage. An apparent down-regulation of transmembrane protein 100 (Tmem100), mesenchyme homeobox 2 (Meox2) and lymphatic vessel endothelial hyaluronan receptor 1 (Lyve1) expression were observed in earlier stage placentas even before apparent size reduction became, suggesting that these genes are involved in the maintenance of fetal capillaries associated with Peg11/Rtl1 during development.
Collapse
Affiliation(s)
- Moe Kitazawa
- Department of Epigenetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype Analysis, The Japan Mouse Clinic, RIKEN BioResource Center (BRC), 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Tomoko Kaneko-Ishino
- School of Health Sciences, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
26
|
|
27
|
Daubas P, Duval N, Bajard L, Langa Vives F, Robert B, Mankoo BS, Buckingham M. Fine-tuning the onset of myogenesis by homeobox proteins that interact with the Myf5 limb enhancer. Biol Open 2015; 4:1614-24. [PMID: 26538636 PMCID: PMC4736032 DOI: 10.1242/bio.014068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Skeletal myogenesis in vertebrates is initiated at different sites of skeletal muscle formation during development, by activation of specific control elements of the myogenic regulatory genes. In the mouse embryo, Myf5 is the first myogenic determination gene to be expressed and its spatiotemporal regulation requires multiple enhancer sequences, extending over 120 kb upstream of the Mrf4-Myf5 locus. An enhancer, located at −57/−58 kb from Myf5, is responsible for its activation in myogenic cells derived from the hypaxial domain of the somite, that will form limb muscles. Pax3 and Six1/4 transcription factors are essential activators of this enhancer, acting on a 145-bp core element. Myogenic progenitor cells that will form the future muscle masses of the limbs express the factors necessary for Myf5 activation when they delaminate from the hypaxial dermomyotome and migrate into the forelimb bud, however they do not activate Myf5 and the myogenic programme until they have populated the prospective muscle masses. We show that Msx1 and Meox2 homeodomain-containing transcription factors bind in vitro and in vivo to specific sites in the 145-bp element, and are implicated in fine-tuning activation of Myf5 in the forelimb. Msx1, when bound between Pax and Six sites, prevents the binding of these key activators, thus inhibiting transcription of Myf5 and consequent premature myogenic differentiation. Meox2 is required for Myf5 activation at the onset of myogenesis via direct binding to other homeodomain sites in this sequence. Thus, these homeodomain factors, acting in addition to Pax3 and Six1/4, fine-tune the entry of progenitor cells into myogenesis at early stages of forelimb development. Summary: Homeodomain factors Msx1 and Meox2, acting in addition to Pax3 and Six1/4, fine-tune the entry of progenitor cells into myogenesis at early stages of forelimb development via modulation of limb enhancer gene Myf5.
Collapse
Affiliation(s)
- Philippe Daubas
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Nathalie Duval
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Lola Bajard
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | | | - Benoît Robert
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Baljinder S Mankoo
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Margaret Buckingham
- CNRS URA 2578, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
| |
Collapse
|
28
|
Endo T. Molecular mechanisms of skeletal muscle development, regeneration, and osteogenic conversion. Bone 2015; 80:2-13. [PMID: 26453493 DOI: 10.1016/j.bone.2015.02.028] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 02/18/2015] [Accepted: 02/28/2015] [Indexed: 12/21/2022]
Abstract
Both skeletal muscle and bone are of mesodermal origin and derived from somites during embryonic development. Somites differentiate into the dorsal dermomyotome and the ventral sclerotome, which give rise to skeletal muscle and bone, respectively. Extracellular signaling molecules, such as Wnt and Shh, secreted from the surrounding environment, determine the developmental fate of skeletal muscle. Dermomyotome cells are specified as trunk muscle progenitor cells by transcription factor networks involving Pax3. These progenitor cells delaminate and migrate to form the myotome, where they are determined as myoblasts that differentiate into myotubes or myofibers. The MyoD family of transcription factors plays pivotal roles in myogenic determination and differentiation. Adult skeletal muscle regenerates upon exercise, muscle injury, or degeneration. Satellite cells are muscle-resident stem cells and play essential roles in muscle growth and regeneration. Muscle regeneration recapitulates the process of muscle development in many aspects. In certain muscle diseases, ectopic calcification or heterotopic ossification, as well as fibrosis and adipogenesis, occurs in skeletal muscle. Muscle-resident mesenchymal progenitor cells, which may be derived from vascular endothelial cells, are responsible for the ectopic osteogenesis, fibrogenesis, and adipogenesis. The small GTPase M-Ras is likely to participate in the ectopic calcification and ossification, as well as in osteogenesis during development. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
29
|
NDR Kinases Are Essential for Somitogenesis and Cardiac Looping during Mouse Embryonic Development. PLoS One 2015; 10:e0136566. [PMID: 26305214 PMCID: PMC4549247 DOI: 10.1371/journal.pone.0136566] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/05/2015] [Indexed: 01/07/2023] Open
Abstract
Studies of mammalian tissue culture cells indicate that the conserved and distinct NDR isoforms, NDR1 and NDR2, play essential cell biological roles. However, mice lacking either Ndr1 or Ndr2 alone develop normally. Here, we studied the physiological consequences of inactivating both NDR1 and NDR2 in mice, showing that the lack of both Ndr1/Ndr2 (called Ndr1/2-double null mutants) causes embryonic lethality. In support of compensatory roles for NDR1 and NDR2, total protein and activating phosphorylation levels of the remaining NDR isoform were elevated in mice lacking either Ndr1 or Ndr2. Mice retaining one single wild-type Ndr allele were viable and fertile. Ndr1/2-double null embryos displayed multiple phenotypes causing a developmental delay from embryonic day E8.5 onwards. While NDR kinases are not required for notochord formation, the somites of Ndr1/2-double null embryos were smaller, irregularly shaped and unevenly spaced along the anterior-posterior axis. Genes implicated in somitogenesis were down-regulated and the normally symmetric expression of Lunatic fringe, a component of the Notch pathway, showed a left-right bias in the last forming somite in 50% of all Ndr1/2-double null embryos. In addition, Ndr1/2-double null embryos developed a heart defect that manifests itself as pericardial edemas, obstructed heart tubes and arrest of cardiac looping. The resulting cardiac insufficiency is the likely cause of the lethality of Ndr1/2-double null embryos around E10. Taken together, we show that NDR kinases compensate for each other in vivo in mouse embryos, explaining why mice deficient for either Ndr1 or Ndr2 are viable. Ndr1/2-double null embryos show defects in somitogenesis and cardiac looping, which reveals their essential functions and shows that the NDR kinases are critically required during the early phase of organogenesis.
Collapse
|
30
|
Passamaneck YJ, Hejnol A, Martindale MQ. Mesodermal gene expression during the embryonic and larval development of the articulate brachiopod Terebratalia transversa. EvoDevo 2015; 6:10. [PMID: 25897375 PMCID: PMC4404124 DOI: 10.1186/s13227-015-0004-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Background Brachiopods undergo radial cleavage, which is distinct from the stereotyped development of closely related spiralian taxa. The mesoderm has been inferred to derive from the archenteron walls following gastrulation, and the primary mesoderm derivative in the larva is a complex musculature. To investigate the specification and differentiation of the mesoderm in the articulate brachiopod Terebratalia transversa, we have identified orthologs of genes involved in mesoderm development in other taxa and investigated their spatial and temporal expression during the embryonic and larval development of T. transversa. Results Orthologs of 17 developmental regulatory genes with roles in the development of the mesoderm in other bilaterian animals were found to be expressed in the developing mesoderm of T. transversa. Five genes, Tt.twist, Tt.GATA456, Tt.dachshund, Tt.mPrx, and Tt.NK1, were found to have expression throughout the archenteron wall at the radial gastrula stage, shortly after the initiation of gastrulation. Three additional genes, Tt.Pax1/9, Tt.MyoD, and Tt.Six1/2, showed expression at this stage in only a portion of the archenteron wall. Tt.eya, Tt.FoxC, Tt.FoxF, Tt.Mox, Tt.paraxis, Tt.Limpet, and Tt.Mef2 all showed initial mesodermal expression during later gastrula or early larval stages. At the late larval stage, Tt.dachshund, Tt.Limpet, and Tt.Mef2 showed expression in nearly all mesoderm cells, while all other genes were localized to specific regions of the mesoderm. Tt.FoxD and Tt.noggin both showed expression in the ventral mesoderm at the larval stages, with gastrula expression patterns in the archenteron roof and blastopore lip, respectively. Conclusions Expression analyses support conserved roles for developmental regulators in the specification and differentiation of the mesoderm during the development of T. transversa. Expression of multiple mesodermal factors in the archenteron wall during gastrulation supports previous morphological observations that this region gives rise to larval mesoderm. Localized expression domains during gastrulation and larval development evidence early regionalization of the mesoderm and provide a basis for hypotheses regarding the molecular regulation underlying the complex system of musculature observed in the larva. Electronic supplementary material The online version of this article (doi:10.1186/s13227-015-0004-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yale J Passamaneck
- Kewalo Marine Laboratory, PBRC, University of Hawaii, 41 Ahui Street, Honolulu, HI 96813 USA ; The Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL 32080 USA
| | - Andreas Hejnol
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate, 55, 5008 Bergen, Norway
| | - Mark Q Martindale
- The Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL 32080 USA
| |
Collapse
|
31
|
Abstract
The effect of strain background on gene function in growth and development has been well documented. However, it has not been extensively reported whether the strain background affects the gene expression pattern. Here, we found that the expression of homeobox gene Meox-2 and FGF receptor 1 gene Fgfr1 during mouse palate development is strain-dependent. On the C57B6 inbred background, Meox-2 is expressed in the palatal outgrowth on Embryonic Day 11.5 (E11.5); the expression shifts posteriorly and is restricted to the back of palate on E14.5. On the Swiss Webster outbred background, Meox-2 expression covers both anterior and posterior regions with the same intensity from E12.5 to E14.5. On the Black Swiss background, Meox-2 expression also covers the entire palate A-P axis, but is much weaker in the anterior region on E14.5. Fgfr1 also displays distinct expression patterns in the palatal outgrowth on E11.5 in these three strains. On the Black Swiss outbred background, the expression is restricted to the anterior palatal outgrowth. In marked contrast, the expression in the Swiss Webster outbred strain is located exclusively in the posterior palate outgrowth on E11.5, whereas in the C57B6 inbred strain, the expression is undetectable in the palatal outgrowth on E11.5.
Collapse
Affiliation(s)
- Jiu-Zhen Jin
- Department of Molecular, Cellular and Craniofacial Biology and Birth Defects Center, University of Louisville School of Dentistry, Louisville, KY 40202, USA
- Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jixiang Ding
- Department of Molecular, Cellular and Craniofacial Biology and Birth Defects Center, University of Louisville School of Dentistry, Louisville, KY 40202, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-502-852-2455; Fax: +1-502-852-4702
| |
Collapse
|
32
|
Zhong Z, Zhao H, Mayo J, Chai Y. Different requirements for Wnt signaling in tongue myogenic subpopulations. J Dent Res 2015; 94:421-9. [PMID: 25576472 DOI: 10.1177/0022034514566030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The tongue is a muscular organ that is essential in vertebrates for important functions, such as food intake and communication. Little is known about regulation of myogenic progenitors during tongue development when compared with the limb or trunk region. In this study, we investigated the relationship between different myogenic subpopulations and the function of canonical Wnt signaling in regulating these subpopulations. We found that Myf5- and MyoD-expressing myogenic subpopulations exist during embryonic tongue myogenesis. In the Myf5-expressing myogenic progenitors, there is a cell-autonomous requirement for canonical Wnt signaling for cell migration and differentiation. In contrast, the MyoD-expressing subpopulation does not require canonical Wnt signaling during tongue myogenesis. Taken together, our results demonstrate that canonical Wnt signaling differentially regulates the Myf5- and MyoD-expressing subpopulations during tongue myogenesis.
Collapse
Affiliation(s)
- Z Zhong
- Department of Orthodontics, School of Stomatology, Peking University, Beijing, China Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - H Zhao
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - J Mayo
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Y Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
33
|
Coppiello G, Collantes M, Sirerol-Piquer MS, Vandenwijngaert S, Schoors S, Swinnen M, Vandersmissen I, Herijgers P, Topal B, van Loon J, Goffin J, Prósper F, Carmeliet P, García-Verdugo JM, Janssens S, Peñuelas I, Aranguren XL, Luttun A. Meox2/Tcf15 heterodimers program the heart capillary endothelium for cardiac fatty acid uptake. Circulation 2015; 131:815-26. [PMID: 25561514 DOI: 10.1161/circulationaha.114.013721] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Microvascular endothelium in different organs is specialized to fulfill the particular needs of parenchymal cells. However, specific information about heart capillary endothelial cells (ECs) is lacking. METHODS AND RESULTS Using microarray profiling on freshly isolated ECs from heart, brain, and liver, we revealed a genetic signature for microvascular heart ECs and identified Meox2/Tcf15 heterodimers as novel transcriptional determinants. This signature was largely shared with skeletal muscle and adipose tissue endothelium and was enriched in genes encoding fatty acid (FA) transport-related proteins. Using gain- and loss-of-function approaches, we showed that Meox2/Tcf15 mediate FA uptake in heart ECs, in part, by driving endothelial CD36 and lipoprotein lipase expression and facilitate FA transport across heart ECs. Combined Meox2 and Tcf15 haplodeficiency impaired FA uptake in heart ECs and reduced FA transfer to cardiomyocytes. In the long term, this combined haplodeficiency resulted in impaired cardiac contractility. CONCLUSIONS Our findings highlight a regulatory role for ECs in FA transfer to the heart parenchyma and unveil 2 of its intrinsic regulators. Our insights could be used to develop new strategies based on endothelial Meox2/Tcf15 targeting to modulate FA transfer to the heart and remedy cardiac dysfunction resulting from altered energy substrate usage.
Collapse
Affiliation(s)
- Giulia Coppiello
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Maria Collantes
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - María Salomé Sirerol-Piquer
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Sara Vandenwijngaert
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Sandra Schoors
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Melissa Swinnen
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Ine Vandersmissen
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Paul Herijgers
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Baki Topal
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Johannes van Loon
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Jan Goffin
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Felipe Prósper
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Peter Carmeliet
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Jose Manuel García-Verdugo
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Stefan Janssens
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Iván Peñuelas
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Xabier L Aranguren
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium
| | - Aernout Luttun
- From Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology (G.C., I.V., X.L.A., A.L.), Department of Cardiovascular Sciences, Cardiology Unit (S.V., M.S., S.J.), Laboratory of Angiogenesis & Neurovascular link, Vesalius Research Center, VIB/Department of Oncology (S.S., P.C.), and Department of Cardiovascular Sciences, Experimental Cardiac Surgery Unit (P.H.), KULeuven, Belgium; Department of Nuclear Medicine, Clínica Universidad de Navarra/MicroPET Research Unit CIMA-CUN (M.C., I.P.), and Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research (F.P., X.L.A), University of Navarra, Pamplona, Spain; Laboratory of Comparative Neurobiology, Instituto Cavanilles, University of Valencia, CIBERNED, Spain (M.S.S.-P., J.M.G.-V.); and Departments of Abdominal Surgery (B.T.) and Neurosurgery (J.v.L., J.G.), University Hospitals Leuven/KULeuven, Belgium.
| |
Collapse
|
34
|
Kopan R, Chen S, Little M. Nephron progenitor cells: shifting the balance of self-renewal and differentiation. Curr Top Dev Biol 2014; 107:293-331. [PMID: 24439811 DOI: 10.1016/b978-0-12-416022-4.00011-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Within the developing mammalian kidney, several populations of progenitors form the discrete cellular components of the final organ. Fate mapping experiments revealed the cap mesenchyme (CM) to be the progenitor population for all nephron epithelial cells, whereas the neighboring stromal mesenchyme gives rise to mesangial, pericytic, renin-producing and interstitial cells. The collecting ducts are derived from a population of progenitors at the ureteric bud (UB) tip and a proportion of the endothelium is also derived from a dedicated mesenchymal progenitor. The stroma, CM, and UB interact to create spatially defined niches at the periphery of the developing organ. While the UB tip population persist, the CM represents a transient progenitor population that is exhausted to set the final organ size. The timing of CM exhaustion, and hence the final organ structure, is sensitive to disruptions such as premature birth. Here we will discuss our current understanding of the molecular processes allowing these populations to balance cell survival, self-renewal, support of branching, and maintain capacity to commit to differentiation.
Collapse
Affiliation(s)
- Raphael Kopan
- Department of Developmental Biology, Washington University, St. Louis, Missouri, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.
| | - Shuang Chen
- Department of Developmental Biology, Washington University, St. Louis, Missouri, USA
| | - Melissa Little
- Department of Developmental Biology, Washington University, St. Louis, Missouri, USA; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
35
|
Gurgul A, Pawlina K, Frys-Żurek M, Bugno-Poniewierska M. Identification of differential selection traces in two Polish cattle breeds. Anim Sci J 2014; 86:17-24. [PMID: 25124517 DOI: 10.1111/asj.12242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/13/2014] [Indexed: 11/30/2022]
Abstract
Genetic improvement of animals based on artificial selection is leading to changes in the frequency of genes related to desirable production traits. The changes are reflected by the neutral, intergenic single nucleotide polymorphims (SNPs) being in long-range linkage disequilibrium with functional polymorphisms. Genome-wide SNP analysis tools designed for cattle, allow for scanning divergences in allelic frequencies between distinct breeds and thus for identification of genomic regions which were divergently selected in breeds' histories. In this study, by using Bovine SNP50 assay, we attempted to identify genomic regions showing the highest differences in allele frequencies between two distinct cattle breeds - preserved, unselected Polish Red breed and highly selected Holstein cattle. Our study revealed 19 genomic regions encompassing 55 protein-coding genes and numerous quantitative trait loci which potentially may underlie some of the phenotypic traits distinguishing the breeds.
Collapse
Affiliation(s)
- Artur Gurgul
- Laboratory of Genomics, National Research Institute of Animal Production, Balice, Poland
| | | | | | | |
Collapse
|
36
|
Stettler O, Moya KL. Distinct roles of homeoproteins in brain topographic mapping and in neural circuit formation. Semin Cell Dev Biol 2014; 35:165-72. [PMID: 25042849 DOI: 10.1016/j.semcdb.2014.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/07/2014] [Indexed: 01/02/2023]
Abstract
The construction of the brain is a highly regulated process, requiring coordination of various cellular and molecular mechanisms that together ensure the stability of the cerebrum architecture and functions. The mature brain is an organ that performs complex computational operations using specific sensory information from the outside world and this requires precise organization within sensory networks and a separation of sensory modalities during development. We review here the role of homeoproteins in the arealization of the brain according to sensorimotor functions, the micropartition of its cytoarchitecture, and the maturation of its sensory circuitry. One of the most interesting observation about homeoproteins in recent years concerns their ability to act both in a cell-autonomous and non-cell-autonomous manner. The highlights in the present review collectively show how these two modes of action of homeoproteins confer various functions in shaping cortical maps.
Collapse
Affiliation(s)
- Olivier Stettler
- Laboratoire CRRET EAC 7149, Université Paris-Est Créteil, 61, Av. du Général de Gaulle, 94010 Créteil Cedex, France.
| | - Kenneth L Moya
- Collège de France, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, 11 place Marcelin Berthelot, 75005 Paris, France; Labex Memolife, PSL Research University, France
| |
Collapse
|
37
|
Beck-Cormier S, Escande M, Souilhol C, Vandormael-Pournin S, Sourice S, Pilet P, Babinet C, Cohen-Tannoudji M. Notchless is required for axial skeleton formation in mice. PLoS One 2014; 9:e98507. [PMID: 24875805 PMCID: PMC4038589 DOI: 10.1371/journal.pone.0098507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 05/03/2014] [Indexed: 01/19/2023] Open
Abstract
Maintenance of cell survival is essential for proper embryonic development. In the mouse, Notchless homolog 1 (Drosophila) (Nle1) is instrumental for survival of cells of the inner cell mass upon implantation. Here, we analyze the function of Nle1 after implantation using the Meox2tm1(cre)Sor mouse that expresses the Cre recombinase specifically in the epiblast at E5.5. First, we find that NLE1 function is required in epiblast cells, as Nle1-deficient cells are rapidly eliminated. In this report, we also show that the Meox2Cre transgene is active in specific tissues during organogenesis. In particular, we detect high Cre expression in the vertebral column, ribs, limbs and tailbud. We took advantage of this dynamic expression profile to analyze the effects of inducing mosaic deletion of Nle1 in the embryo. We show that Nle1 deletion in this context, results in severe developmental anomalies leading to lethality at birth. Mutant embryos display multiple developmental defects in particular during axial skeletal formation. We also provide evidence that axial defects are due to an increase in apoptotic cell death in the somite at E9.5. These data demonstrate an essential role for Nle1 during organogenesis and in particular during axial development.
Collapse
Affiliation(s)
- Sarah Beck-Cormier
- Mouse Functional Genetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique, URA 2578, Institut Pasteur, Paris, France
- * E-mail:
| | - Marie Escande
- Mouse Functional Genetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
| | - Céline Souilhol
- Mouse Functional Genetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
| | - Sandrine Vandormael-Pournin
- Mouse Functional Genetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique, URA 2578, Institut Pasteur, Paris, France
| | - Sophie Sourice
- Institut National de la Santé et de la recherche Médicale, U791, LIOAD, STEP group “Skeletal Tissue Engineering and Physiopathology”, Nantes, France
| | - Paul Pilet
- Institut National de la Santé et de la recherche Médicale, U791, LIOAD, STEP group “Skeletal Tissue Engineering and Physiopathology”, Nantes, France
| | - Charles Babinet
- Mouse Functional Genetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique, URA 2578, Institut Pasteur, Paris, France
| | - Michel Cohen-Tannoudji
- Mouse Functional Genetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique, URA 2578, Institut Pasteur, Paris, France
| |
Collapse
|
38
|
Gene regulatory networks and transcriptional mechanisms that control myogenesis. Dev Cell 2014; 28:225-38. [PMID: 24525185 DOI: 10.1016/j.devcel.2013.12.020] [Citation(s) in RCA: 439] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/24/2013] [Accepted: 12/27/2013] [Indexed: 12/11/2022]
Abstract
We discuss the upstream regulators of myogenesis that lead to the activation of myogenic determination genes and subsequent differentiation, focusing on the mouse model. Key upstream genes, such as Pax3 and Pax7, Six1 and Six4, or Pitx2, participate in gene regulatory networks at different sites of skeletal muscle formation. MicroRNAs also intervene, with emerging evidence for the role of other noncoding RNAs. Myogenic determination and subsequent differentiation depend on members of the MyoD family. We discuss new insights into mechanisms underlying the transcriptional activity of these factors.
Collapse
|
39
|
Cunnington RH, Northcott JM, Ghavami S, Filomeno KL, Jahan F, Kavosh MS, Davies JJL, Wigle JT, Dixon IMC. The Ski-Zeb2-Meox2 pathway provides a novel mechanism for regulation of the cardiac myofibroblast phenotype. J Cell Sci 2013; 127:40-9. [PMID: 24155330 DOI: 10.1242/jcs.126722] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiac fibrosis is linked to fibroblast-to-myofibroblast phenoconversion and proliferation but the mechanisms underlying this are poorly understood. Ski is a negative regulator of TGF-β-Smad signaling in myofibroblasts, and might redirect the myofibroblast phenotype back to fibroblasts. Meox2 could alter TGF-β-mediated cellular processes and is repressed by Zeb2. Here, we investigated whether Ski diminishes the myofibroblast phenotype by de-repressing Meox2 expression and function through repression of Zeb2 expression. We show that expression of Meox1 and Meox2 mRNA and Meox2 protein is reduced during phenoconversion of fibroblasts to myofibroblasts. Overexpression of Meox2 shifts the myofibroblasts into fibroblasts, whereas the Meox2 DNA-binding mutant has no effect on myofibroblast phenotype. Overexpression of Ski partially restores Meox2 mRNA expression levels to those in cardiac fibroblasts. Expression of Zeb2 increased during phenoconversion and Ski overexpression reduces Zeb2 expression in first-passage myofibroblasts. Furthermore, expression of Meox2 is decreased in scar following myocardial infarction, whereas Zeb2 protein expression increases in the infarct scar. Thus Ski modulates the cardiac myofibroblast phenotype and function through suppression of Zeb2 by upregulating the expression of Meox2. This cascade might regulate cardiac myofibroblast phenotype and presents therapeutic options for treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Ryan H Cunnington
- Department of Physiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bayrakli F, Guclu B, Yakicier C, Balaban H, Kartal U, Erguner B, Sagiroglu MS, Yuksel S, Ozturk AR, Kazanci B, Ozum U, Kars HZ. Mutation in MEOX1 gene causes a recessive Klippel-Feil syndrome subtype. BMC Genet 2013; 14:95. [PMID: 24073994 PMCID: PMC3849745 DOI: 10.1186/1471-2156-14-95] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/26/2013] [Indexed: 11/23/2022] Open
Abstract
Background Klippel-Feil syndrome (KFS) is characterized by the developmental failure of the cervical spine and has two dominantly inherited subtypes. Affected individuals who are the children of a consanguineous marriage are extremely rare in the medical literature, but the gene responsible for this recessive trait subtype of KFS has recently been reported. Results We identified a family with the KFS phenotype in which their parents have a consanguineous marriage. Radiological examinations revealed that they carry fusion defects and numerical abnormalities in the cervical spine, scoliosis, malformations of the cranial base, and Sprengel’s deformity. We applied whole genome linkage and whole-exome sequencing analysis to identify the chromosomal locus and gene mutated in this family. Whole genome linkage analysis revealed a significant linkage to chromosome 17q12-q33 with a LOD score of 4.2. Exome sequencing identified the G > A p.Q84X mutation in the MEOX1 gene, which is segregated based on pedigree status. Homozygous MEOX1 mutations have reportedly caused a similar phenotype in knockout mice. Conclusions Here, we report a truncating mutation in the MEOX1 gene in a KFS family with an autosomal recessive trait. Together with another recently reported study and the knockout mouse model, our results suggest that mutations in MEOX1 cause a recessive KFS phenotype in humans.
Collapse
Affiliation(s)
- Fatih Bayrakli
- Department of Neurosurgery, Cumhuriyet University School of Medicine, Kampus, Merkez, 58140, Sivas, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Anderson C, Williams VC, Moyon B, Daubas P, Tajbakhsh S, Buckingham ME, Shiroishi T, Hughes SM, Borycki AG. Sonic hedgehog acts cell-autonomously on muscle precursor cells to generate limb muscle diversity. Genes Dev 2012; 26:2103-17. [PMID: 22987640 DOI: 10.1101/gad.187807.112] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
How muscle diversity is generated in the vertebrate body is poorly understood. In the limb, dorsal and ventral muscle masses constitute the first myogenic diversification, as each gives rise to distinct muscles. Myogenesis initiates after muscle precursor cells (MPCs) have migrated from the somites to the limb bud and populated the prospective muscle masses. Here, we show that Sonic hedgehog (Shh) from the zone of polarizing activity (ZPA) drives myogenesis specifically within the ventral muscle mass. Shh directly induces ventral MPCs to initiate Myf5 transcription and myogenesis through essential Gli-binding sites located in the Myf5 limb enhancer. In the absence of Shh signaling, myogenesis is delayed, MPCs fail to migrate distally, and ventral paw muscles fail to form. Thus, Shh production in the limb ZPA is essential for the spatiotemporal control of myogenesis and coordinates muscle and skeletal development by acting directly to regulate the formation of specific ventral muscles.
Collapse
Affiliation(s)
- Claire Anderson
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Retinoic acid enhances skeletal myogenesis in human embryonic stem cells by expanding the premyogenic progenitor population. Stem Cell Rev Rep 2012; 8:482-93. [PMID: 21735106 DOI: 10.1007/s12015-011-9284-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human embryonic stem cells (hESCs) are a potential source of material for cell therapy of muscle diseases. To date, it has proven difficult to generate skeletal muscle from hESCs in high yields and within a reasonable timeframe. Further, a hESC-derived Pax3/7-positive skeletal muscle progenitor population has not yet been described. Previous studies have shown that Pax3/7-positive progenitor cells can repopulate the satellite cell niche, indicating the importance of this population for therapy. We sought to optimize the differentiation of hESCs into skeletal muscle in order to characterize myogenesis at a molecular level and shorten the time course. We treated hESCs with retinoic acid (RA) and found an enhancement of skeletal myogenesis, and the expression of the myogenic regulatory factors (MRFs) MyoD and myogenin by day 25. Furthermore, we found that RA treatment expanded the muscle progenitor pool, which occurred as a distinct Pax3(+ve) population prior to MRF expression. Non-skeletal muscle tissue types were not significantly affected. Therefore, we have identified a differentiation pathway in hESCs that provides a skeletal muscle progenitor population which can undergo myogenesis more efficiently. We propose that RA could fit into a directed culture method for deriving skeletal muscle from hESCs.
Collapse
|
43
|
Yusuf F, Brand-Saberi B. Myogenesis and muscle regeneration. Histochem Cell Biol 2012; 138:187-99. [DOI: 10.1007/s00418-012-0972-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2012] [Indexed: 12/27/2022]
|
44
|
Cole NJ, Currie P. Shaping muscle bioarchitecture for the fin to limb transition. BIOARCHITECTURE 2012; 2:98-103. [PMID: 22880150 PMCID: PMC3414388 DOI: 10.4161/bioa.20969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Our recent paper examined how pelvic fins and their musculature form developmentally and how these mechanisms have evolved within the vertebrate lineage, a process fundamental to the tetrapod transition. The transition from the water onto the land is among one of the most well studied steps in the evolutionary history of vertebrates, yet the genetic basis of this evolutionary transition is little studied and ill-defined. The advent of these terrestrial species resulted in a shift in locomotor strategies from the rhythmic undulating muscles of the fish body to a reliance upon powerful weight bearing muscles of the limbs to generate movement. We demonstrated that the pelvic fin muscles of bony fish are generated by a mechanism that has features of both of limb/fin muscle formation in tetrapods and primitive cartilaginous fish. We hypothesize that the adoption of the fully derived mode of hindlimb muscle formation, was a further modification of the mode of development deployed to generate pelvic fin muscles, a shift in overall muscle bioarchitecture we believe was critical to the success of the tetrapod transition.
Collapse
|
45
|
Milligan JN, Jolly ER. Identification and characterization of a Mef2 transcriptional activator in schistosome parasites. PLoS Negl Trop Dis 2012; 6:e1443. [PMID: 22235355 PMCID: PMC3250504 DOI: 10.1371/journal.pntd.0001443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 11/08/2011] [Indexed: 11/17/2022] Open
Abstract
Myocyte enhancer factor 2 protein (Mef2) is an evolutionarily conserved activator of transcription that is critical to induce and control complex processes in myogenesis and neurogenesis in vertebrates and insects, and osteogenesis in vertebrates. In Drosophila, Mef2 null mutants are unable to produce differentiated muscle cells, and in vertebrates, Mef2 mutants are embryonic lethal. Schistosome worms are responsible for over 200 million cases of schistosomiasis globally, but little is known about early development of schistosome parasites after infecting a vertebrate host. Understanding basic schistosome development could be crucial to delineating potential drug targets. Here, we identify and characterize Mef2 from the schistosome worm Schistosoma mansoni (SmMef2). We initially identified SmMef2 as a homolog to the yeast Mef2 homolog, Resistance to Lethality of MKK1P386 overexpression (Rlm1), and we show that SmMef2 is homologous to conserved Mef2 family proteins. Using a genetics approach, we demonstrate that SmMef2 is a transactivator that can induce transcription of four separate heterologous reporter genes by yeast one-hybrid analysis. We also show that Mef2 is expressed during several stages of schistosome development by quantitative PCR and that it can bind to conserved Mef2 DNA consensus binding sequences. Schistosome parasites infect more than 200 million people worldwide and cause human schistosomiasis. Free-swimming schistosome larvae are highly mobile and invade and penetrate the host's skin to perpetuate their lifecycle in their human host, growing from 90–215 micrometers in length as a schistosomulum to a 7–20 millimeter long adult worm. Few molecular pathways have been identified in schistosome worms that are important for parasite early development. The myocyte enhancer factor protein 2 is a major regulator of muscle and nerve development in mammals and insects and is highly conserved from bread yeast to vertebrates. Here we identify and characterize the Mef2 activator from parasitic schistosome worms, the first described in any parasitic worm, and delineation of its function may be important to further understanding the basic biology of schistosome early development. Additionally, since schistosomes developed early evolutionarily, an investigation of schistosome Mef2 regulatory mechanisms could lead to a greater understanding of the development of early muscle and neurogenic development in animals.
Collapse
Affiliation(s)
- John N Milligan
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
46
|
Makarenkova HP, Meech R. Barx homeobox family in muscle development and regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 297:117-73. [PMID: 22608559 DOI: 10.1016/b978-0-12-394308-8.00004-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Homeobox transcription factors are key intrinsic regulators of myogenesis. In studies spanning several years, we have characterized the homeobox factor Barx2 as a novel marker for muscle progenitor cells and an important regulator of muscle growth and repair. In this review, we place the expression and function of Barx2 and its paralogue Barx1 in context with other muscle-expressed homeobox factors in both embryonic and adult myogenesis. We also describe the structure and regulation of Barx genes and possible gene/disease associations. The functional domains of Barx proteins, their molecular interactions, and cellular functions are presented with particular emphasis on control of genes and processes involved in myogenic differentiation. Finally, we describe the patterns of Barx gene expression in vivo and the phenotypes of various Barx gene perturbation models including null mice. We focus on the Barx2 null mouse model, which has demonstrated the critical roles of Barx2 in postnatal myogenesis including muscle maintenance during aging, and regeneration of acute and chronic muscle injury.
Collapse
Affiliation(s)
- Helen P Makarenkova
- The Neurobiology Department, Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
47
|
Sambasivan R, Kuratani S, Tajbakhsh S. An eye on the head: the development and evolution of craniofacial muscles. Development 2011; 138:2401-15. [DOI: 10.1242/dev.040972] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Skeletal muscles exert diverse functions, enabling both crushing with great force and movement with exquisite precision. A remarkably distinct repertoire of genes and ontological features characterise this tissue, and recent evidence has shown that skeletal muscles of the head, the craniofacial muscles, are evolutionarily, morphologically and molecularly distinct from those of the trunk. Here, we review the molecular basis of craniofacial muscle development and discuss how this process is different to trunk and limb muscle development. Through evolutionary comparisons of primitive chordates (such as amphioxus) and jawless vertebrates (such as lampreys) with jawed vertebrates, we also provide some clues as to how this dichotomy arose.
Collapse
Affiliation(s)
- Ramkumar Sambasivan
- Institut Pasteur, Stem Cells and Development, Paris, F-75015, France
- CNRS URA 2578, 25 rue du Dr Roux, Paris, F-75015, France
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells and Development, Paris, F-75015, France
- CNRS URA 2578, 25 rue du Dr Roux, Paris, F-75015, France
| |
Collapse
|
48
|
Zhao X, Mo D, Li A, Gong W, Xiao S, Zhang Y, Qin L, Niu Y, Guo Y, Liu X, Cong P, He Z, Wang C, Li J, Chen Y. Comparative analyses by sequencing of transcriptomes during skeletal muscle development between pig breeds differing in muscle growth rate and fatness. PLoS One 2011; 6:e19774. [PMID: 21637832 PMCID: PMC3102668 DOI: 10.1371/journal.pone.0019774] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 04/05/2011] [Indexed: 01/25/2023] Open
Abstract
Understanding the dynamics of muscle transcriptome during development and between breeds differing in muscle growth is necessary to uncover the complex mechanism underlying muscle development. Herein, we present the first transcriptome-wide longissimus dorsi muscle development research concerning Lantang (LT, obese) and Landrace (LR, lean) pig breeds during 10 time-points from 35 days-post-coitus (dpc) to 180 days-post-natum (dpn) using Solexa/Illumina's Genome Analyzer. The data demonstrated that myogenesis was almost completed before 77 dpc, but the muscle phenotypes were still changed from 77 dpc to 28 dpn. Comparative analysis of the two breeds suggested that myogenesis started earlier but progressed more slowly in LT than in LR, the stages ranging from 49 dpc to 77 dpc are critical for formation of different muscle phenotypes. 595 differentially expressed myogenesis genes were identified, and their roles in myogenesis were discussed. Furthermore, GSK3B, IKBKB, ACVR1, ITGA and STMN1 might contribute to later myogenesis and more muscle fibers in LR than LT. Some myogenesis inhibitors (ID1, ID2, CABIN1, MSTN, SMAD4, CTNNA1, NOTCH2, GPC3 and HMOX1) were higher expressed in LT than in LR, which might contribute to more slow muscle differentiation in LT than in LR. We also identified several genes which might contribute to intramuscular adipose differentiation. Most important, we further proposed a novel model in which MyoD and MEF2A controls the balance between intramuscular adipogenesis and myogenesis by regulating CEBP family; Myf5 and MEF2C are essential during the whole myogenesis process while MEF2D affects muscle growth and maturation. The MRFs and MEF2 families are also critical for the phenotypic differences between the two pig breeds. Overall, this study contributes to elucidating the mechanism underlying muscle development, which could provide valuable information for pig meat quality improvement. The raw data have been submitted to Gene Expression Omnibus (GEO) under series GSE25406.
Collapse
Affiliation(s)
- Xiao Zhao
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Delin Mo
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Anning Li
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Wen Gong
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shuqi Xiao
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yue Zhang
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Limei Qin
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yuna Niu
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yunxue Guo
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Peiqing Cong
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zuyong He
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chong Wang
- College of Animal Science, South China of Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Jiaqi Li
- College of Animal Science, South China of Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
- * E-mail:
| |
Collapse
|
49
|
Hasson P. "Soft" tissue patterning: muscles and tendons of the limb take their form. Dev Dyn 2011; 240:1100-7. [PMID: 21438070 DOI: 10.1002/dvdy.22608] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2011] [Indexed: 12/18/2022] Open
Abstract
The musculoskeletal system grants our bodies a vast range of movements. Because it is mainly composed of easily identifiable components, it serves as an ideal model to study patterning of the specific tissues that make up the organ. Surprisingly, although critical for the function of the musculoskeletal system, understanding of the embryonic processes that regulate muscle and tendon patterning is very limited. The recent identification of specific markers and the reagents stemming from them has revealed some of the molecular events regulating patterning of these soft tissues. This review will focus on some of the current work, with an emphasis on the roles of the muscle connective tissue, and discuss several key points that addressing them will advance our understanding of these patterning events.
Collapse
Affiliation(s)
- Peleg Hasson
- Department of Anatomy and Cell Biology, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Bat Galim, Haifa, Israel.
| |
Collapse
|
50
|
Mok GF, Sweetman D. Many routes to the same destination: lessons from skeletal muscle development. Reproduction 2011; 141:301-12. [PMID: 21183656 DOI: 10.1530/rep-10-0394] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The development and differentiation of vertebrate skeletal muscle provide an important paradigm to understand the inductive signals and molecular events controlling differentiation of specific cell types. Recent findings show that a core transcriptional network, initiated by the myogenic regulatory factors (MRFs; MYF5, MYOD, myogenin and MRF4), is activated by separate populations of cells in embryos in response to various signalling pathways. This review will highlight how cells from multiple distinct starting points can converge on a common set of regulators to generate skeletal muscle.
Collapse
Affiliation(s)
- Gi Fay Mok
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, UK
| | | |
Collapse
|