1
|
Godwin JS, Michel JM, Libardi CA, Kavazis AN, Fry CS, Frugé AD, McCashland M, Vechetti IJ, McCarthy JJ, Mobley CB, Roberts MD. Resistance exercise and mechanical overload upregulate vimentin for skeletal muscle remodeling. Am J Physiol Cell Physiol 2025; 328:C1509-C1525. [PMID: 40178318 DOI: 10.1152/ajpcell.01028.2024] [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: 12/20/2024] [Revised: 01/10/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025]
Abstract
We adopted a proteomic and follow-through approach to investigate how mechanical overload (MOV) potentially affects novel targets in skeletal muscle, and how a perturbation in this response could potentially affect the adaptive response. First, we determined that 10 wk of resistance training in 15 college-aged females increased sarcolemmal-associated protein content (+10.1%, P < 0.05). Sarcolemmal protein isolates were then queried using mass spectrometry-based proteomics, ∼10% (38/387) of proteins putatively associated with the sarcolemma or extracellular matrix (ECM) were upregulated (>1.5-fold, P < 0.05), and one target (intermediate filament vimentin; VIM) warranted further investigation due to its correlation to myofiber hypertrophy (r = 0.652, P = 0.009). VIM expression was then examined in 4-mo-old C57BL/6J mice following 10 and 20 days of plantaris MOV via synergist ablation. Relative to Sham (control) mice, VIM mRNA and protein content was significantly higher in MOV mice, and immunohistochemistry indicated that VIM predominantly resided in the ECM. MOV experiments were replicated in Pax7-DTA (satellite cell depleted) mice, which reduced VIM in the ECM by ∼74%. A third MOV experiment was performed in C57BL/6 mice intramuscularly injected with either AAV9-scrambled (control) or AAV9-VIM-shRNA. Although VIM-shRNA mice possessed lower VIM in the ECM (∼45%), plantaris masses in response to MOV were similar between groups. However, VIM-shRNA mice possessed smaller and more centrally nucleated MyHCemb-positive fibers in response to MOV. In summary, skeletal muscle VIM appears to be enriched in the ECM following MOV, satellite cells may regulate its expression, and a disruption in expression during MOV leads to an excessive regenerative phenotype.NEW & NOTEWORTHY Our highly integrative approach suggests that skeletal muscle vimentin seems to function as a mechanosensitive protein that becomes enriched in the extracellular matrix following MOV. Satellite cells may play a role in regulating their expression, and an exaggerated regenerative response occurs when vimentin expression becomes dysregulated during mechanical overload. Although these data implicate vimentin in aiding with tissue remodeling following MOV, more data are needed to determine the functional ramifications of VIM response deficiencies.
Collapse
Affiliation(s)
- Joshua S Godwin
- Nutrabolt Applied and Molecular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - J Max Michel
- Nutrabolt Applied and Molecular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of Sao Carlos, Sao Carlos, Brazil
| | - Andreas N Kavazis
- Nutrabolt Applied and Molecular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Christopher S Fry
- Department of Athletic Training & Clinical Nutrition, University of Kentucky, Lexington, Kentucky, United States
| | - Andrew D Frugé
- Nutrabolt Applied and Molecular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
- College of Nursing, Auburn University, Auburn, Alabama, United States
| | - Mariah McCashland
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, Nebraska, United States
| | - Ivan J Vechetti
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, Nebraska, United States
| | - John J McCarthy
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - C Brooks Mobley
- Nutrabolt Applied and Molecular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Michael D Roberts
- Nutrabolt Applied and Molecular Physiology Laboratory, School of Kinesiology, Auburn University, Auburn, Alabama, United States
| |
Collapse
|
2
|
Samardak K, Bâcle J, Moriel-Carretero M. Behind the stoNE wall: A fervent activity for nuclear lipids. Biochimie 2024; 227:53-84. [PMID: 39111564 DOI: 10.1016/j.biochi.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/27/2024]
Abstract
The four main types of biomolecules are nucleic acids, proteins, carbohydrates and lipids. The knowledge about their respective interactions is as important as the individual understanding of each of them. However, while, for example, the interaction of proteins with the other three groups is extensively studied, that of nucleic acids and lipids is, in comparison, very poorly explored. An iconic paradigm of physical (and likely functional) proximity between DNA and lipids is the case of the genomic DNA in eukaryotes: enclosed within the nucleus by two concentric lipid bilayers, the wealth of implications of this interaction, for example in genome stability, remains underassessed. Nuclear lipid-related phenotypes have been observed for 50 years, yet in most cases kept as mere anecdotical descriptions. In this review, we will bring together the evidence connecting lipids with both the nuclear envelope and the nucleoplasm, and will make critical analyses of these descriptions. Our exploration establishes a scenario in which lipids irrefutably play a role in nuclear homeostasis.
Collapse
Affiliation(s)
- Kseniya Samardak
- Centre de Recherche en Biologie cellulaire de Montpellier (CRBM) UMR5237, Université de Montpellier, Centre National de La Recherche Scientifique, 34293 Montpellier Cedex 5, France
| | - Janélie Bâcle
- Centre de Recherche en Biologie cellulaire de Montpellier (CRBM) UMR5237, Université de Montpellier, Centre National de La Recherche Scientifique, 34293 Montpellier Cedex 5, France
| | - María Moriel-Carretero
- Centre de Recherche en Biologie cellulaire de Montpellier (CRBM) UMR5237, Université de Montpellier, Centre National de La Recherche Scientifique, 34293 Montpellier Cedex 5, France.
| |
Collapse
|
3
|
Wada E, Susumu N, Kaya M, Hayashi YK. Characteristics of nuclear architectural abnormalities of myotubes differentiated from Lmna H222P/H222P skeletal muscle cells. In Vitro Cell Dev Biol Anim 2024; 60:781-792. [PMID: 38724872 DOI: 10.1007/s11626-024-00915-1] [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: 02/13/2024] [Accepted: 04/22/2024] [Indexed: 08/03/2024]
Abstract
The presence of nuclear architectural abnormalities is a hallmark of the nuclear envelopathies, which are a group of diseases caused by mutations in genes encoding nuclear envelope proteins. Mutations in the lamin A/C gene cause several diseases, named laminopathies, including muscular dystrophies, progeria syndromes, and lipodystrophy. A mouse model carrying with the LmnaH222P/H222P mutation (H222P) was shown to develop severe cardiomyopathy but only mild skeletal myopathy, although abnormal nuclei were observed in their striated muscle. In this report, we analyzed the abnormal-shaped nuclei in myoblasts and myotubes isolated from skeletal muscle of H222P mice, and evaluated the expression of nuclear envelope proteins in these abnormal myonuclei. Primary skeletal muscle cells from H222P mice proliferated and efficiently differentiated into myotubes in vitro, similarly to those from wild-type mice. During cell proliferation, few abnormal-shaped nuclei were detected; however, numerous markedly abnormal myonuclei were observed in myotubes from H222P mice on days 5 and 7 of differentiation. Time-lapse observation demonstrated that myonuclei with a normal shape maintained their normal shape, whereas abnormal-shaped myonuclei remained abnormal for at least 48 h during differentiation. Among the abnormal-shaped myonuclei, 65% had a bleb with a string structure, and 35% were severely deformed. The area and nuclear contents of the nuclear blebs were relatively stable, whereas the myocytes with nuclear blebs were actively fused within primary myotubes. Although myonuclei were markedly deformed, the deposition of DNA damage marker (γH2AX) or apoptotic marker staining was rarely observed. Localizations of lamin A/C and emerin were maintained within the blebs, strings, and severely deformed regions of myonuclei; however, lamin B1, nesprin-1, and a nuclear pore complex protein were absent in these abnormal regions. These results demonstrate that nuclear membranes from H222P skeletal muscle cells do not rupture and are resistant to DNA damage, despite these marked morphological changes.
Collapse
Affiliation(s)
- Eiji Wada
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Nao Susumu
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Motoshi Kaya
- Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yukiko K Hayashi
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
4
|
Yang J, Dong X, Wen H, Li Y, Wang X, Yan S, Zuo C, Lyu L, Zhang K, Qi X. FGFs function in regulating myoblasts differentiation in spotted sea bass (Lateolabrax maculatus). Gen Comp Endocrinol 2024; 347:114426. [PMID: 38103843 DOI: 10.1016/j.ygcen.2023.114426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Fibroblast growth factors (FGFs) are a family of structurally related peptides that regulate processes such as cell proliferation, differentiation, and damage repair. In our previous study, fibroblast growth factor receptor 4 (fgfr4) was detected in the most significant quantitative trait loci (QTL), when identified of QTLs and genetic markers for growth-related traits in spotted sea bass. However, knowledge of the function of fgfr4 is lacking, even the legends to activate the receptor is unknown in fish. To remedy this problem, in the present study, a total of 33 fgfs were identified from the genomic and transcriptomic databases of spotted sea bass, of which 10 were expressed in the myoblasts. According to the expression pattern during myoblasts proliferation and differentiation, fgf6a, fgf6b and fgf18 were selected for further prokaryotic expression and purification. The recombinant proteins FGF6a, FGF6b and FGF18 were found to inhibit myoblast differentiation. Overall, our results provide a theoretical basis for the molecular mechanisms of growth regulation in economic fish such as spotted sea bass.
Collapse
Affiliation(s)
- Jing Yang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Ximeng Dong
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Xiaojie Wang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Shaojing Yan
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Chenpeng Zuo
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Likang Lyu
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Kaiqiang Zhang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao 266003.
| |
Collapse
|
5
|
Hakim F, Kazemiraad C, Akbari-Birgani S, Abdollahpour D, Mohammadi S. Caspase-9-mediated cleavage of vimentin attenuates the aggressiveness of leukemic NB4 cells. Mol Cell Biochem 2023; 478:2435-2444. [PMID: 36807844 DOI: 10.1007/s11010-023-04671-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/30/2023] [Indexed: 02/21/2023]
Abstract
Vimentin is a main type 3 intermediate filament protein. It seems that abnormal expression of vimentin is contributed to the appearance of the aggressive feature of cancer cells. So that it has been reported that malignancy and epithelial-mesenchymal transition in solid tumors, and poor clinical outcomes in patients with lymphocytic leukemia and acute myelocytic leukemia have been associated with the high expression of vimentin. Vimentin is a non-caspase substrate of caspase-9 although its cleavage by caspase-9 in biological processes has not been reported. In the present study, we sought to understand whether vimentin cleavage mediated by caspase-9 could reverse the malignancy in leukemic cells. Herein, to address the issue, we investigated vimentin changes in differentiation and took advantage of the inducible caspase-9 (iC9)/AP1903 system in human leukemic NB4 cells. Following the transfection and treatment of the cells using the iC9/AP1903 system, vimentin expression, cleavage, and subsequently, the cell invasion and the relevant markers such as CD44 and MMP-9 were evaluated. Our results revealed the downregulation and cleavage of vimentin which attenuates the malignant phenotype of the NB4 cells. Considering the favorable effect of this strategy in keeping down the malignant features of the leukemic cells, the effect of the iC9/AP1903 system in combination with all-trans-retinoic acid (ATRA) treatment was evaluated. The obtained data prove that iC9/AP1903 significantly makes the leukemic cells more sensitive to ATRA.
Collapse
Affiliation(s)
- Fatemeh Hakim
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), 45137-66731, Zanjan, Iran
| | - Cyrus Kazemiraad
- Laboratory for Functional and Metabolic Imaging, Institute of Physics, Swiss Federal Institute of Technology (EPFL), Station6, 1015, Lausanne, Switzerland
| | - Shiva Akbari-Birgani
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), 45137-66731, Zanjan, Iran.
- Research Center for Basic Sciences and Modern Technologies (RBST), Institute for Advanced Studies in Basic Sciences (IASBS), 45137-66731, Zanjan, Iran.
| | - Daryoush Abdollahpour
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), 45137-66731, Zanjan, Iran
- Optics Research Center, Institute for Advanced Studies in Basic Sciences (IASBS), 45137-66731, Zanjan, Iran
| | - Saeed Mohammadi
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Stiekema M, Houben F, Verheyen F, Borgers M, Menzel J, Meschkat M, van Zandvoort MAMJ, Ramaekers FCS, Broers JLV. The Role of Lamins in the Nucleoplasmic Reticulum, a Pleiomorphic Organelle That Enhances Nucleo-Cytoplasmic Interplay. Front Cell Dev Biol 2022; 10:914286. [PMID: 35784476 PMCID: PMC9243388 DOI: 10.3389/fcell.2022.914286] [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: 04/06/2022] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
Invaginations of the nuclear membrane occur in different shapes, sizes, and compositions. Part of these pleiomorphic invaginations make up the nucleoplasmic reticulum (NR), while others are merely nuclear folds. We define the NR as tubular invaginations consisting of either both the inner and outer nuclear membrane, or only the inner nuclear membrane. Specifically, invaginations of both the inner and outer nuclear membrane are also called type II NR, while those of only the inner nuclear membrane are defined as type I NR. The formation and structure of the NR is determined by proteins associated to the nuclear membrane, which induce a high membrane curvature leading to tubular invaginations. Here we review and discuss the current knowledge of nuclear invaginations and the NR in particular. An increase in tubular invaginations of the nuclear envelope is associated with several pathologies, such as laminopathies, cancer, (reversible) heart failure, and Alzheimer’s disease. Furthermore, viruses can induce both type I and II NR. In laminopathies, the amount of A-type lamins throughout the nucleus is generally decreased or the organization of lamins or lamin-associated proteins is disturbed. Also, lamin overexpression or modulation of lamin farnesylation status impacts NR formation, confirming the importance of lamin processing in NR formation. Virus infections reorganize the nuclear lamina via (de)phosphorylation of lamins, leading to an uneven thickness of the nuclear lamina and in turn lobulation of the nuclear membrane and the formation of invaginations of the inner nuclear membrane. Since most studies on the NR have been performed with cell cultures, we present additional proof for the existence of these structures in vivo, focusing on a variety of differentiated cardiovascular and hematopoietic cells. Furthermore, we substantiate the knowledge of the lamin composition of the NR by super-resolution images of the lamin A/C and B1 organization. Finally, we further highlight the essential role of lamins in NR formation by demonstrating that (over)expression of lamins can induce aberrant NR structures.
Collapse
Affiliation(s)
- Merel Stiekema
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Frederik Houben
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Healthcare, PXL University College, Hasselt, Belgium
| | - Fons Verheyen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marcel Borgers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | | | - Marc A. M. J. van Zandvoort
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research IMCAR, RWTH Aachen University, Aachen, Germany
| | - Frans C. S. Ramaekers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jos L. V. Broers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands
- *Correspondence: Jos L. V. Broers,
| |
Collapse
|
7
|
Echarri A. A Multisensory Network Drives Nuclear Mechanoadaptation. Biomolecules 2022; 12:biom12030404. [PMID: 35327596 PMCID: PMC8945967 DOI: 10.3390/biom12030404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 12/03/2022] Open
Abstract
Cells have adapted to mechanical forces early in evolution and have developed multiple mechanisms ensuring sensing of, and adaptation to, the diversity of forces operating outside and within organisms. The nucleus must necessarily adapt to all types of mechanical signals, as its functions are essential for virtually all cell processes, many of which are tuned by mechanical cues. To sense forces, the nucleus is physically connected with the cytoskeleton, which senses and transmits forces generated outside and inside the cell. The nuclear LINC complex bridges the cytoskeleton and the nuclear lamina to transmit mechanical information up to the chromatin. This system creates a force-sensing macromolecular complex that, however, is not sufficient to regulate all nuclear mechanoadaptation processes. Within the nucleus, additional mechanosensitive structures, including the nuclear envelope and the nuclear pore complex, function to regulate nuclear mechanoadaptation. Similarly, extra nuclear mechanosensitive systems based on plasma membrane dynamics, mechanotransduce information to the nucleus. Thus, the nucleus has the intrinsic structural components needed to receive and interpret mechanical inputs, but also rely on extra nuclear mechano-sensors that activate nuclear regulators in response to force. Thus, a network of mechanosensitive cell structures ensures that the nucleus has a tunable response to mechanical cues.
Collapse
Affiliation(s)
- Asier Echarri
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Mechanoadaptation and Caveolae Biology Laboratory, Areas of Cell & Developmental Biology, Calle Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| |
Collapse
|
8
|
Mehrabi M, Morris TA, Cang Z, Nguyen CHH, Sha Y, Asad MN, Khachikyan N, Greene TL, Becker DM, Nie Q, Zaragoza MV, Grosberg A. A Study of Gene Expression, Structure, and Contractility of iPSC-Derived Cardiac Myocytes from a Family with Heart Disease due to LMNA Mutation. Ann Biomed Eng 2021; 49:3524-3539. [PMID: 34585335 PMCID: PMC8671287 DOI: 10.1007/s10439-021-02850-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Genetic mutations to the Lamin A/C gene (LMNA) can cause heart disease, but the mechanisms making cardiac tissues uniquely vulnerable to the mutations remain largely unknown. Further, patients with LMNA mutations have highly variable presentation of heart disease progression and type. In vitro patient-specific experiments could provide a powerful platform for studying this phenomenon, but the use of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) introduces heterogeneity in maturity and function thus complicating the interpretation of the results of any single experiment. We hypothesized that integrating single cell RNA sequencing (scRNA-seq) with analysis of the tissue architecture and contractile function would elucidate some of the probable mechanisms. To test this, we investigated five iPSC-CM lines, three controls and two patients with a (c.357-2A>G) mutation. The patient iPSC-CM tissues had significantly weaker stress generation potential than control iPSC-CM tissues demonstrating the viability of our in vitro approach. Through scRNA-seq, differentially expressed genes between control and patient lines were identified. Some of these genes, linked to quantitative structural and functional changes, were cardiac specific, explaining the targeted nature of the disease progression seen in patients. The results of this work demonstrate the utility of combining in vitro tools in exploring heart disease mechanics.
Collapse
Affiliation(s)
- Mehrsa Mehrabi
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Tessa A Morris
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA.,Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA
| | - Zixuan Cang
- Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA.,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Cecilia H H Nguyen
- Genetics & Genomics Division, Department of Pediatrics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Yutong Sha
- Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Mira N Asad
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Nyree Khachikyan
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Taylor L Greene
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Danielle M Becker
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Qing Nie
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA.,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Michael V Zaragoza
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.,Genetics & Genomics Division, Department of Pediatrics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Anna Grosberg
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA. .,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA. .,Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA. .,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA. .,Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA. .,The Henry Samueli School of Engineering, University of California, Irvine, 2418 Engineering Hall, Irvine, CA, 92697, USA.
| |
Collapse
|
9
|
CRISPR/Cas9 small promoter deletion in H19 lncRNA is associated with altered cell morphology and proliferation. Sci Rep 2021; 11:18380. [PMID: 34526543 PMCID: PMC8443613 DOI: 10.1038/s41598-021-97058-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023] Open
Abstract
The imprinted H19 long non-coding RNA, a knowing oncofetal gene, presents a controversial role during the carcinogenesis process since its tumor suppressor or oncogenic activity is not completely elucidated. Since H19 lncRNA is involved in many biological pathways related to tumorigenesis, we sought to develop a non-cancer lineage with CRISPR-Cas9-mediated H19 knockdown (H19-) and observe the changes in a cellular context. To edit the promoter region of H19, two RNA guides were designed, and the murine C2C12 myoblast cells were transfected. H19 deletion was determined by DNA sequencing and gene expression by qPCR. We observed a small deletion (~ 60 bp) in the promoter region that presented four predicted transcription binding sites. The deletion reduced H19 expression (30%) and resulted in increased proliferative activity, altered morphological patterns including cell size and intracellular granularity, without changes in viability. The increased proliferation rate in the H19- cell seems to facilitate chromosomal abnormalities. The H19- myoblast presented characteristics similar to cancer cells, therefore the H19 lncRNA may be an important gene during the initiation of the tumorigenic process. Due to CRISPR/Cas9 permanent edition, the C2C12 H19- knockdown cells allows functional studies of H19 roles in tumorigenesis, prognosis, metastases, as well as drug resistance and targeted therapy.
Collapse
|
10
|
Feliksiak K, Witko T, Solarz D, Guzik M, Rajfur Z. Vimentin Association with Nuclear Grooves in Normal MEF 3T3 Cells. Int J Mol Sci 2020; 21:E7478. [PMID: 33050497 PMCID: PMC7590159 DOI: 10.3390/ijms21207478] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/25/2022] Open
Abstract
Vimentin, an intermediate filament protein present in leukocytes, blood vessel endothelial cells, and multiple mesenchymal cells, such as mouse embryonic fibroblasts (MEF 3T3), is crucial for various cellular processes, as well as for maintaining the integrity and durability (stability) of the cell cytoskeleton. Vimentin intermediate filaments (VIFs) adhere tightly to the nucleus and spread to the lamellipodium and tail of the cell, serving as a connector between the nucleus, and the cell's edges, especially in terms of transferring mechanical signals throughout the cell. How these signals are transmitted exactly remains under investigation. In the presented work, we propose that vimentin is involved in that transition by influencing the shape of the nucleus through the formation of nuclear blebs and grooves, as demonstrated by microscopic observations of healthy MEF (3T3) cells. Grooved, or "coffee beans" nuclei, have, to date, been noticed in several healthy cells; however, these structures are especially frequent in cancer cells-they serve as a significant marker for recognition of multiple cancers. We observed 288 MEF3T3 cells cultured on polyhydroxyoctanoate (PHO), polylactide (PLA), and glass, and we identified grooves, coaligned with vimentin fibers in the nuclei of 47% of cells cultured on PHO, 50% of cells on glass, and 59% of cells growing on PLA. We also observed nuclear blebs and associated their occurrence with the type of substrate used for cell culture. We propose that the higher rate of blebs in the nuclei of cells, cultured on PLA, is related to the microenvironmental features of the substrate, pH in particular.
Collapse
Affiliation(s)
- Karolina Feliksiak
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland; (K.F.); (D.S.)
| | - Tomasz Witko
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Kraków, Poland;
| | - Daria Solarz
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland; (K.F.); (D.S.)
| | - Maciej Guzik
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Kraków, Poland;
| | - Zenon Rajfur
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland; (K.F.); (D.S.)
| |
Collapse
|
11
|
Freifeld L, Odstrcil I, Förster D, Ramirez A, Gagnon JA, Randlett O, Costa EK, Asano S, Celiker OT, Gao R, Martin-Alarcon DA, Reginato P, Dick C, Chen L, Schoppik D, Engert F, Baier H, Boyden ES. Expansion microscopy of zebrafish for neuroscience and developmental biology studies. Proc Natl Acad Sci U S A 2017; 114:E10799-E10808. [PMID: 29162696 PMCID: PMC5740639 DOI: 10.1073/pnas.1706281114] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Expansion microscopy (ExM) allows scalable imaging of preserved 3D biological specimens with nanoscale resolution on fast diffraction-limited microscopes. Here, we explore the utility of ExM in the larval and embryonic zebrafish, an important model organism for the study of neuroscience and development. Regarding neuroscience, we found that ExM enabled the tracing of fine processes of radial glia, which are not resolvable with diffraction-limited microscopy. ExM further resolved putative synaptic connections, as well as molecular differences between densely packed synapses. Finally, ExM could resolve subsynaptic protein organization, such as ring-like structures composed of glycine receptors. Regarding development, we used ExM to characterize the shapes of nuclear invaginations and channels, and to visualize cytoskeletal proteins nearby. We detected nuclear invagination channels at late prophase and telophase, potentially suggesting roles for such channels in cell division. Thus, ExM of the larval and embryonic zebrafish may enable systematic studies of how molecular components are configured in multiple contexts of interest to neuroscience and developmental biology.
Collapse
Affiliation(s)
- Limor Freifeld
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
| | - Iris Odstrcil
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Dominique Förster
- Department Genes-Circuits-Behavior, Max Planck Institute of Neurobiology, Martinsried 82152, Germany
| | - Alyson Ramirez
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - James A Gagnon
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Owen Randlett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Emma K Costa
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139
| | - Shoh Asano
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
| | - Orhan T Celiker
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139
| | - Ruixuan Gao
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139
| | | | - Paul Reginato
- Department of Biological Engineering, MIT, Cambridge, MA 02139
- Department of Genetics, Harvard Medical School, Cambridge, MA 02138
| | - Cortni Dick
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
| | - Linlin Chen
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
- Neuroscience Program, Wellesley College, Wellesley, MA 02481
| | - David Schoppik
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016
- Neuroscience Institute, New York University School of Medicine, New York NY 10016
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Herwig Baier
- Department Genes-Circuits-Behavior, Max Planck Institute of Neurobiology, Martinsried 82152, Germany
| | - Edward S Boyden
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139;
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139
- Center for Neurobiological Engineering, MIT, Cambridge, MA 02139
| |
Collapse
|
12
|
Core JQ, Mehrabi M, Robinson ZR, Ochs AR, McCarthy LA, Zaragoza MV, Grosberg A. Age of heart disease presentation and dysmorphic nuclei in patients with LMNA mutations. PLoS One 2017; 12:e0188256. [PMID: 29149195 PMCID: PMC5693421 DOI: 10.1371/journal.pone.0188256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/05/2017] [Indexed: 01/24/2023] Open
Abstract
Nuclear shape defects are a distinguishing characteristic in laminopathies, cancers, and other pathologies. Correlating these defects to the symptoms, mechanisms, and progression of disease requires unbiased, quantitative, and high-throughput means of quantifying nuclear morphology. To accomplish this, we developed a method of automatically segmenting fluorescently stained nuclei in 2D microscopy images and then classifying them as normal or dysmorphic based on three geometric features of the nucleus using a package of Matlab codes. As a test case, cultured skin-fibroblast nuclei of individuals possessing LMNA splice-site mutation (c.357-2A>G), LMNA nonsense mutation (c.736 C>T, pQ246X) in exon 4, LMNA missense mutation (c.1003C>T, pR335W) in exon 6, Hutchinson-Gilford Progeria Syndrome, and no LMNA mutations were analyzed. For each cell type, the percentage of dysmorphic nuclei, and other morphological features such as average nuclear area and average eccentricity were obtained. Compared to blind observers, our procedure implemented in Matlab codes possessed similar accuracy to manual counting of dysmorphic nuclei while being significantly more consistent. The automatic quantification of nuclear defects revealed a correlation between in vitro results and age of patients for initial symptom onset. Our results demonstrate the method’s utility in experimental studies of diseases affecting nuclear shape through automated, unbiased, and accurate identification of dysmorphic nuclei.
Collapse
Affiliation(s)
- Jason Q. Core
- Departments of Biomedical Engineering, University of California, Irvine, CA, United States of America
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, United States of America
| | - Mehrsa Mehrabi
- Departments of Biomedical Engineering, University of California, Irvine, CA, United States of America
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, United States of America
| | - Zachery R. Robinson
- Departments of Biomedical Engineering, University of California, Irvine, CA, United States of America
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, United States of America
| | - Alexander R. Ochs
- Departments of Biomedical Engineering, University of California, Irvine, CA, United States of America
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, United States of America
| | - Linda A. McCarthy
- Departments of Biomedical Engineering, University of California, Irvine, CA, United States of America
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, United States of America
| | - Michael V. Zaragoza
- Pediatrics–Genetics & Genomics Division–School of Medicine, University of California, Irvine, CA, United States of America
- Biological Chemistry–School of Medicine, University of California, Irvine, CA, United States of America
| | - Anna Grosberg
- Departments of Biomedical Engineering, University of California, Irvine, CA, United States of America
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, United States of America
- Chemical Engineering and Materials Science, University of California, Irvine, CA, United States of America
- * E-mail:
| |
Collapse
|
13
|
Skinner BM, Johnson EEP. Nuclear morphologies: their diversity and functional relevance. Chromosoma 2017; 126:195-212. [PMID: 27631793 PMCID: PMC5371643 DOI: 10.1007/s00412-016-0614-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Studies of chromosome and genome biology often focus on condensed chromatin in the form of chromosomes and neglect the non-dividing cells. Even when interphase nuclei are considered, they are often then treated as interchangeable round objects. However, different cell types can have very different nuclear shapes, and these shapes have impacts on cellular function; indeed, many pathologies are linked with alterations to nuclear shape. In this review, we describe some of the nuclear morphologies beyond the spherical and ovoid. Many of the leukocytes of the immune system have lobed nuclei, which aid their flexibility and migration; smooth muscle cells have a spindle shaped nucleus, which must deform during muscle contractions; spermatozoa have highly condensed nuclei which adopt varied shapes, potentially associated with swimming efficiency. Nuclei are not passive passengers within the cell. There are clear effects of nuclear shape on the transcriptional activity of the cell. Recent work has shown that regulation of gene expression can be influenced by nuclear morphology, and that cells can drastically remodel their chromatin during differentiation. The link between the nucleoskeleton and the cytoskeleton at the nuclear envelope provides a mechanism for transmission of mechanical forces into the nucleus, directly affecting chromatin compaction and organisation.
Collapse
Affiliation(s)
- Benjamin M Skinner
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
| | - Emma E P Johnson
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| |
Collapse
|
14
|
Ansseau E, Eidahl JO, Lancelot C, Tassin A, Matteotti C, Yip C, Liu J, Leroy B, Hubeau C, Gerbaux C, Cloet S, Wauters A, Zorbo S, Meyer P, Pirson I, Laoudj-Chenivesse D, Wattiez R, Harper SQ, Belayew A, Coppée F. Homologous Transcription Factors DUX4 and DUX4c Associate with Cytoplasmic Proteins during Muscle Differentiation. PLoS One 2016; 11:e0146893. [PMID: 26816005 PMCID: PMC4729438 DOI: 10.1371/journal.pone.0146893] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 12/24/2015] [Indexed: 12/26/2022] Open
Abstract
Hundreds of double homeobox (DUX) genes map within 3.3-kb repeated elements dispersed in the human genome and encode DNA-binding proteins. Among these, we identified DUX4, a potent transcription factor that causes facioscapulohumeral muscular dystrophy (FSHD). In the present study, we performed yeast two-hybrid screens and protein co-purifications with HaloTag-DUX fusions or GST-DUX4 pull-down to identify protein partners of DUX4, DUX4c (which is identical to DUX4 except for the end of the carboxyl terminal domain) and DUX1 (which is limited to the double homeodomain). Unexpectedly, we identified and validated (by co-immunoprecipitation, GST pull-down, co-immunofluorescence and in situ Proximal Ligation Assay) the interaction of DUX4, DUX4c and DUX1 with type III intermediate filament protein desmin in the cytoplasm and at the nuclear periphery. Desmin filaments link adjacent sarcomere at the Z-discs, connect them to sarcolemma proteins and interact with mitochondria. These intermediate filament also contact the nuclear lamina and contribute to positioning of the nuclei. Another Z-disc protein, LMCD1 that contains a LIM domain was also validated as a DUX4 partner. The functionality of DUX4 or DUX4c interactions with cytoplasmic proteins is underscored by the cytoplasmic detection of DUX4/DUX4c upon myoblast fusion. In addition, we identified and validated (by co-immunoprecipitation, co-immunofluorescence and in situ Proximal Ligation Assay) as DUX4/4c partners several RNA-binding proteins such as C1QBP, SRSF9, RBM3, FUS/TLS and SFPQ that are involved in mRNA splicing and translation. FUS and SFPQ are nuclear proteins, however their cytoplasmic translocation was reported in neuronal cells where they associated with ribonucleoparticles (RNPs). Several other validated or identified DUX4/DUX4c partners are also contained in mRNP granules, and the co-localizations with cytoplasmic DAPI-positive spots is in keeping with such an association. Large muscle RNPs were recently shown to exit the nucleus via a novel mechanism of nuclear envelope budding. Following DUX4 or DUX4c overexpression in muscle cell cultures, we observed their association with similar nuclear buds. In conclusion, our study demonstrated unexpected interactions of DUX4/4c with cytoplasmic proteins playing major roles during muscle differentiation. Further investigations are on-going to evaluate whether these interactions play roles during muscle regeneration as previously suggested for DUX4c.
Collapse
Affiliation(s)
- Eugénie Ansseau
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Jocelyn O. Eidahl
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Céline Lancelot
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Christel Matteotti
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Cassandre Yip
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Jian Liu
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Baptiste Leroy
- Laboratory of Proteomic and Microbiology, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Céline Hubeau
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Cécile Gerbaux
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Samuel Cloet
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Armelle Wauters
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Sabrina Zorbo
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Pierre Meyer
- Pediatric Department, CHRU Montpellier, Montpellier, France
| | - Isabelle Pirson
- I.R.I.B.H.M., Free University of Brussels, Brussels, Belgium
| | | | - Ruddy Wattiez
- Laboratory of Proteomic and Microbiology, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Scott Q. Harper
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
- Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Alexandra Belayew
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Frédérique Coppée
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
- * E-mail:
| |
Collapse
|
15
|
Nikonenko AG, Bozhok YM. Simple computational technique to quantify nuclear shape asymmetry. Cytometry A 2014; 87:309-14. [DOI: 10.1002/cyto.a.22612] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 10/17/2014] [Accepted: 12/02/2014] [Indexed: 12/18/2022]
Affiliation(s)
| | - Yuriy M. Bozhok
- Department of Functional Diagnostics; Institute of Endocrinology and Metabolism; Kiev Ukraine
| |
Collapse
|
16
|
Fernandez-Fuente M, Terracciano CM, Martin-Duque P, Brown SC, Vassaux G, Piercy RJ. Calcium homeostasis in myogenic differentiation factor 1 (MyoD)-transformed, virally-transduced, skin-derived equine myotubes. PLoS One 2014; 9:e105971. [PMID: 25148524 PMCID: PMC4141859 DOI: 10.1371/journal.pone.0105971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/30/2014] [Indexed: 11/19/2022] Open
Abstract
Dysfunctional skeletal muscle calcium homeostasis plays a central role in the pathophysiology of several human and animal skeletal muscle disorders, in particular, genetic disorders associated with ryanodine receptor 1 (RYR1) mutations, such as malignant hyperthermia, central core disease, multiminicore disease and certain centronuclear myopathies. In addition, aberrant skeletal muscle calcium handling is believed to play a pivotal role in the highly prevalent disorder of Thoroughbred racehorses, known as Recurrent Exertional Rhabdomyolysis. Traditionally, such defects were studied in human and equine subjects by examining the contractile responses of biopsied muscle strips exposed to caffeine, a potent RYR1 agonist. However, this test is not widely available and, due to its invasive nature, is potentially less suitable for valuable animals in training or in the human paediatric setting. Furthermore, increasingly, RYR1 gene polymorphisms (of unknown pathogenicity and significance) are being identified through next generation sequencing projects. Consequently, we have investigated a less invasive test that can be used to study calcium homeostasis in cultured, skin-derived fibroblasts that are converted to the muscle lineage by viral transduction with a MyoD (myogenic differentiation 1) transgene. Similar models have been utilised to examine calcium homeostasis in human patient cells, however, to date, there has been no detailed assessment of the cells’ calcium homeostasis, and in particular, the responses to agonists and antagonists of RYR1. Here we describe experiments conducted to assess calcium handling of the cells and examine responses to treatment with dantrolene, a drug commonly used for prophylaxis of recurrent exertional rhabdomyolysis in horses and malignant hyperthermia in humans.
Collapse
Affiliation(s)
- Marta Fernandez-Fuente
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London, United Kingdom
| | - Cesare M. Terracciano
- Laboratory of Cell Electrophysiology, Imperial College London, Myocardial Function, National Heart and Lung Institute, Hammersmith Hospital, London, United Kingdom
| | - Pilar Martin-Duque
- Universidad Francisco de Vitoria, Facultad de Ciencias Biosanitarias: Pozuelo de Alarcón (Madrid), Madrid, Spain
| | - Susan C. Brown
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Georges Vassaux
- Laboratoire TIRO, UMRE 4320, iBEB, DSV, Commissariat a’ l’Energie Atomique, Nice, France
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London, United Kingdom
- * E-mail:
| |
Collapse
|
17
|
Rambharose S, Ojewole E, Mackraj I, Govender T. Comparative buccal permeability enhancement of didanosine and tenofovir by potential multifunctional polymeric excipients and their effects on porcine buccal histology. Pharm Dev Technol 2013; 19:82-90. [DOI: 10.3109/10837450.2012.752505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Salova AV, Leontieva EA, Mozhenok TP, Kornilova ES, Krolenko SA, Belyaeva TN. Changes in localization of cellular vesicular apparatus during differentiation of myoblasts into myotubules in cell culture. ACTA ACUST UNITED AC 2011. [DOI: 10.1134/s1990519x11030096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Malhas A, Goulbourne C, Vaux DJ. The nucleoplasmic reticulum: form and function. Trends Cell Biol 2011; 21:362-73. [PMID: 21514163 DOI: 10.1016/j.tcb.2011.03.008] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/17/2011] [Accepted: 03/23/2011] [Indexed: 11/29/2022]
Abstract
The nuclear envelope (NE) physically separates nucleoplasm and cytoplasm, contributes to nuclear structural integrity, controls selective bidirectional transport of ions and macromolecular cargo, regulates gene expression, and acts as a mechanotransducer and a platform for signalling. It is noteworthy however that the NE is not simply a smooth-surfaced outer boundary but is interrupted by invaginations that reach deep within the nucleoplasm and could even traverse the nucleus completely. The existence of such a complex branched network of invaginations forming a nucleoplasmic reticulum (NR) provides sites that are capable of carrying out the 'conventional' NE functions deep within the nucleus in regions that would otherwise be remote from the nuclear periphery. In this review, we describe the structural features of NR in normal and pathological states and discuss the current understanding of their functional and possible pathological roles.
Collapse
Affiliation(s)
- Ashraf Malhas
- Sir William Dunn School of Pathology, Oxford OX1 3RE, UK
| | | | | |
Collapse
|
20
|
Polychronidou M, Grobhans J. Determining nuclear shape: the role of farnesylated nuclear membrane proteins. Nucleus 2011; 2:17-23. [PMID: 21647295 PMCID: PMC3104805 DOI: 10.4161/nucl.2.1.13992] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 01/18/2023] Open
Abstract
Changes in nuclear morphology are observed in diverse developmental processes as well as in pathological conditions. Modification of nuclear membrane and nuclear lamina protein levels results in altered nuclear shapes, as it has been demonstrated in experimental systems ranging from yeast to human cells. The important role of nuclear membrane components in regulating nuclear morphology is additionally highlighted by the abnormally shaped nuclei observed in diseases where nuclear lamina proteins are mutated. Even though the effect of nuclear envelope components on nuclear shape has been thoroughly described, not much is known about the molecular mechanisms that govern these events. In addition to the known role of intermediate filament formation by lamins, here we discuss several mechanisms that might alone or in combination participate in the regulation of nuclear shape observed upon modification of the levels of nuclear membrane and lamina proteins. Based on recent work with the two farnesylated nuclear membrane Drosophila proteins, kugelkern and lamin Dm0, we propose that the direct interaction of farnesylated nuclear membrane proteins with the phospholipid bilayer leads to nuclear envelope deformation. In addition to this mechanism, we suggest that the interaction of nuclear membrane and lamina proteins with cytoskeletal elements and chromatin, and modifications in lipid biosynthesis might also be involved in the formation of abnormally shaped nuclei.
Collapse
Affiliation(s)
- Maria Polychronidou
- Institut für Biochemie und Molekulare Zellbiologie, Georg-August-Universität Göttingen, Germany
| | | |
Collapse
|
21
|
|
22
|
Myhre JL, Pilgrim DB. Cellular Differentiation in Primary Cell Cultures from Single Zebrafish Embryos as a Model for the Study of Myogenesis. Zebrafish 2010; 7:255-66. [DOI: 10.1089/zeb.2010.0665] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- J. Layne Myhre
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - David B. Pilgrim
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
23
|
Langevin HM, Storch KN, Snapp RR, Bouffard NA, Badger GJ, Howe AK, Taatjes DJ. Tissue stretch induces nuclear remodeling in connective tissue fibroblasts. Histochem Cell Biol 2010; 133:405-15. [PMID: 20237796 PMCID: PMC2880391 DOI: 10.1007/s00418-010-0680-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2010] [Indexed: 01/14/2023]
Abstract
Studies in cultured cells have shown that nuclear shape is an important factor influencing nuclear function, and that mechanical forces applied to the cell can directly affect nuclear shape. In a previous study, we demonstrated that stretching of whole mouse subcutaneous tissue causes dynamic cytoskeletal remodeling with perinuclear redistribution of alpha-actin in fibroblasts within the tissue. We have further shown that the nuclei of these fibroblasts have deep invaginations containing alpha-actin. In the current study, we hypothesized that tissue stretch would cause nuclear remodeling with a reduced amount of nuclear invagination, measurable as a change in nuclear concavity. Subcutaneous areolar connective tissue samples were excised from 28 mice and randomized to either tissue stretch or no stretch for 30 min, then examined with histochemistry and confocal microscopy. In stretched tissue (vs. non-stretched), fibroblast nuclei had a larger cross-sectional area (P < 0.001), smaller thickness (P < 0.03) in the plane of the tissue, and smaller relative concavity (P < 0.005) indicating an increase in nuclear convexity. The stretch-induced loss of invaginations may have important influences on gene expression, RNA trafficking and/or cell differentiation.
Collapse
Affiliation(s)
- Helene M Langevin
- Department of Neurology, University of Vermont College of Medicine, 89 Beaumont Ave, Burlington, VT, 05405, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Surface and inner cell behaviour along skeletal muscle cell in vitro differentiation. Micron 2008; 39:843-51. [DOI: 10.1016/j.micron.2007.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 12/20/2007] [Accepted: 12/21/2007] [Indexed: 11/22/2022]
|
25
|
Brunet N, Tarabal O, Portero-Otín M, Oppenheim RW, Esquerda JE, Calderó J. Survival and death of mature avian motoneurons in organotypic slice culture: trophic requirements for survival and different types of degeneration. J Comp Neurol 2007; 501:669-90. [PMID: 17299760 DOI: 10.1002/cne.21157] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We have developed an organotypic culture technique that uses slices of chick embryo spinal cord, in which trophic requirements for long-term survival of mature motoneurons (MNs) were studied. Slices were obtained from E16 chick embryos and maintained for up to 28 days in vitro (DIV) in a basal medium. Under these conditions, most MNs died. To promote MN survival, 14 different trophic factors were assayed. Among these 14, glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor were the most effective. GDNF was able to promote MN survival for at least 28 DIV. K(+) depolarization or caspase inhibition prevented MN death but also induced degenerative-like changes in rescued MNs. Agents that elevate cAMP levels promoted the survival of a proportion of MNs for at least 7 DIV. Examination of dying MNs revealed that, in addition to cells exhibiting a caspase-3-dependent apoptotic pattern, some MNs died by a caspase-3-independent mechanism and displayed autophagic vacuoles, an extremely convoluted nucleus, and a close association with microglia. This organotypic spinal cord slice culture may provide a convenient model for testing conditions that promote survival of mature-like MNs that are affected in late-onset MN disease such as amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Núria Brunet
- Unitat de Neurobiologia Cel.lular, Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida and IRB Lleida, 25008 Lleida, Catalonia, Spain
| | | | | | | | | | | |
Collapse
|
26
|
Qin RF, Mao TQ, Gu XM, Hu KJ, Liu YP, Chen JW, Nie X. Regulation of skeletal muscle differentiation in fibroblasts by exogenous MyoD gene in vitro and in vivo. Mol Cell Biochem 2007; 302:233-9. [PMID: 17415623 DOI: 10.1007/s11010-007-9446-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 03/02/2007] [Indexed: 01/20/2023]
Abstract
MyoD of the myogenic regulatory factors (MRFs) family regulates the skeletal muscle differentiation program. In this study, stably transfected NIH3T3-derived cell lines were established, in which exogenous MyoD was expressed at high levels. Transcriptional activation of endogenous muscle regulatory gene and induction towards the skeletal muscle lineages were observed with phase-contrast microscopy when continuously cultured in vitro. Moreover, to determine their ability of myogenic formation in vivo, the transfected cells were implanted in nude mice subcutaneously for up to 10 weeks. The morphological characterization of inductive cells was observed using transmission electron microscope and histological staining. Myogenesis of fibroblasts incubated in the medium was activated by overexpression of MyoD, and the cells were accumulated and fused into multinucleated myotubes. Correlatively, RT-PCR and immunohistochemistry confirmed the increased expression of characteristic downstream molecule myogenin and mysion heavy chains during myogenic differentiation. Ecoptic myogenesis was found and remained stable phenotype when the transfected cells were seeded in vivo. Our results suggest that MyoD can be considered to be a determining factor of myogenic lineages, and it may play an important role in the cell therapy and cell-mediated gene therapy of the skeletal muscle.
Collapse
Affiliation(s)
- Rui-Feng Qin
- Department of Oral Maxillofacial Surgery, Qin Du Stomatological College, Fourth Military Medical University, Xian, P.R. China
| | | | | | | | | | | | | |
Collapse
|
27
|
Trelles-Sticken E, Adelfalk C, Loidl J, Scherthan H. Meiotic telomere clustering requires actin for its formation and cohesin for its resolution. J Cell Biol 2005; 170:213-23. [PMID: 16027219 PMCID: PMC2171397 DOI: 10.1083/jcb.200501042] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Accepted: 06/15/2005] [Indexed: 11/25/2022] Open
Abstract
In diploid organisms, meiosis reduces the chromosome number by half during the formation of haploid gametes. During meiotic prophase, telomeres transiently cluster at a limited sector of the nuclear envelope (bouquet stage) near the spindle pole body (SPB). Cohesin is a multisubunit complex that contributes to chromosome segregation in meiosis I and II divisions. In yeast meiosis, deficiency for Rec8 cohesin subunit induces telomere clustering to persist, whereas telomere cluster-SPB colocalization is defective. These defects are rescued by expressing the mitotic cohesin Scc1 in rec8delta meiosis, whereas bouquet-stage exit is independent of Cdc5 pololike kinase. An analysis of living Saccharomyces cerevisiae meiocytes revealed highly mobile telomeres from leptotene up to pachytene, with telomeres experiencing an actin- but not microtubule-dependent constraint of mobility during the bouquet stage. Our results suggest that cohesin is required for exit from actin polymerization-dependent telomere clustering and for linking the SPB to the telomere cluster in synaptic meiosis.
Collapse
|