1
|
Wang H, Li J, Yu K, Lu Y, Ma M, Li Y. The cellular localization and oncogenic or tumor suppressive effects of angiomiotin-like protein 2 in tumor and normal cells. IUBMB Life 2024; 76:764-779. [PMID: 38717123 DOI: 10.1002/iub.2830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/10/2024] [Indexed: 10/19/2024]
Abstract
Angiomiotin (AMOT) family comprises three members: AMOT, AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). AMOTL2 is widely expressed in endothelial cells, epithelial cells, and various cancer cells. Specifically, AMOTL2 predominantly localizes in the cytoplasm and nucleus in human normal cells, whereas associates with cell-cell junctions and actin cytoskeleton in non-human cells, and locates at cell junctions or within the recycling endosomes in cancer cells. AMOTL2 is implicated in regulation of tube formation, cell polarity, and shape, although the specific impact on tumorigenesis remains to be conclusively determined. It has been shown that AMOTL2 enhances tumor growth and metastasis in pancreatic, breast, and colon cancer, however inhibits cell proliferation and migration in lung, hepatocellular cancer, and glioblastoma. In addition to its role in cell shape and cytoskeletal dynamics through co-localization with F-actin, AMOTL2 modulates the transcription of Yes-associated protein (YAP) by binding to it, thereby affecting its phosphorylation and cellular sequestration. Furthermore, the stability and cellular localization of AMOTL2, influenced by its phosphorylation and ubiquitination mediated by specific proteins, affects its cellular function. Additionally, we observe that AMOTL2 is predominantly downregulated in some tumors, but significantly elevated in colorectal adenocarcinoma (COAD). Moreover, overall analysis, GSEA and ROC curve analysis indicate that AMOTL2 exerts as an oncogenic protein in COAD by modulating Wnt pathway, participating in synthesis of collagen formation, and interacting with extracellular matrix receptor. In addition, AMOTL2 potentially regulates the distribution of immune cells infiltration in COAD. In summary, AMOTL2 probably functions as an oncogene in COAD. Consequently, further in-depth mechanistic research is required to elucidate the precise roles of AMOTL2 in various cancers.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kexun Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengdi Ma
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Venghateri JB, Dassa B, Morgenstern D, Shreberk-Shaked M, Oren M, Geiger B. Deciphering the involvement of the Hippo pathway co-regulators, YAP/TAZ in invadopodia formation and matrix degradation. Cell Death Dis 2023; 14:290. [PMID: 37185904 PMCID: PMC10130049 DOI: 10.1038/s41419-023-05769-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023]
Abstract
Invadopodia are adhesive, actin-rich protrusions formed by metastatic cancer cells that degrade the extracellular matrix and facilitate invasion. They support the metastatic cascade by a spatially and temporally coordinated process whereby invading cells bind to the matrix, degrade it by specific metalloproteinases, and mechanically penetrate diverse tissue barriers by forming actin-rich extensions. However, despite the apparent involvement of invadopodia in the metastatic process, the molecular mechanisms that regulate invadopodia formation and function are still largely unclear. In this study, we have explored the involvement of the key Hippo pathway co-regulators, namely YAP, and TAZ, in invadopodia formation and matrix degradation. Toward that goal, we tested the effect of depletion of YAP, TAZ, or both on invadopodia formation and activity in multiple human cancer cell lines. We report that the knockdown of YAP and TAZ or their inhibition by verteporfin induces a significant elevation in matrix degradation and invadopodia formation in several cancer cell lines. Conversely, overexpression of these proteins strongly suppresses invadopodia formation and matrix degradation. Proteomic and transcriptomic profiling of MDA-MB-231 cells, following co-knockdown of YAP and TAZ, revealed a significant change in the levels of key invadopodia-associated proteins, including the crucial proteins Tks5 and MT1-MMP (MMP14). Collectively, our findings show that YAP and TAZ act as negative regulators of invadopodia formation in diverse cancer lines, most likely by reducing the levels of essential invadopodia components. Dissecting the molecular mechanisms of invadopodia formation in cancer invasion may eventually reveal novel targets for therapeutic applications against invasive cancer.
Collapse
Affiliation(s)
- Jubina Balan Venghateri
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Bareket Dassa
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | | | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Benjamin Geiger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Luderman LN, Michaels MT, Levic DS, Knapik EW. Zebrafish Erc1b mediates motor innervation and organization of craniofacial muscles in control of jaw movement. Dev Dyn 2023; 252:104-123. [PMID: 35708710 DOI: 10.1002/dvdy.511] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Movement of the lower jaw, a common behavior observed among vertebrates, is required for eating and processing food. This movement is controlled by signals sent from the trigeminal motor nerve through neuromuscular junctions (NMJs) to the masticatory muscles. Dysfunctional jaw movements contribute to craniomandibular disorders, yet the pathophysiology of these disorders is not well understood, as limited studies have been conducted on the molecular mechanisms of jaw movement. RESULTS Using erc1b/kimm533 genetic loss of function mutant, we evaluated lower jaw muscle organization and innervation by the cranial motor nerves in developing zebrafish. Using time-lapse confocal imaging of the erc1b mutant in a transgenic fluorescent reporter line, we found delayed trigeminal nerve growth and disrupted nerve branching architecture during muscle innervation. By automated 3D image analysis of NMJ distribution, we identified an increased number of small, disorganized NMJ clusters in erc1b mutant larvae compared to WT siblings. Using genetic replacement experiments, we determined the Rab GTPase binding domain of Erc1b is required for cranial motor nerve branching, but not NMJ organization or muscle attachment. CONCLUSIONS We identified Erc1b/ERC1 as a novel component of a genetic pathway contributing to muscle organization, trigeminal nerve outgrowth, and NMJ spatial distribution during development that is required for jaw movement.
Collapse
Affiliation(s)
- Lauryn N Luderman
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mackenzie T Michaels
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daniel S Levic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
- Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Ela W Knapik
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
4
|
Gong B, Zheng Y, Li J, Lei H, Liu K, Tang J, Peng Y. Luteolin activates M2 macrophages and suppresses M1 macrophages by upregulation of hsa_circ_0001326 in THP-1 derived macrophages. Bioengineered 2022; 13:5079-5090. [PMID: 35152837 PMCID: PMC8973855 DOI: 10.1080/21655979.2022.2036897] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Asthma is accompanied by inflammatory progression. Macrophages are a major type of cells to response inflammation caused by different type of factors by polarized into specific phenotypes. Luteolin and glycyrrhizic acid exert protect role in asthma; however, their role in THP-1 derived macrophages polarization whether through regulating the expression of hsa_circ_0001326 is still unknown. The effect of luteolin and glycyrrhizic acid on THP-1 derived macrophages polarization were evaluated using qRT-PCR, Western blotting, and ELISA assay. The function of hsa_circ_0001326 on macrophages polarization in luteolin treated THP-1 derived macrophages were assessed after silence of hsa_circ_0001326. And the expression of its’ potential downstream gene, including hsa-miR-136-5p and ubiquitin-specific protease 4 (USP4), were detected using qRT-PCR and Western blot analysis. Furthermore, the potential mechanism of hsa_circ_0001326 were validated using rescue experiment. Results showed that luteolin promoted M2 polarization and inhibited M1 polarization in THP-1 induced macrophages, but glycyrrhizic acid had no these effects. Hsa_circ_0001326 expression was upregulated in luteolin treat THP-1 derived macrophages. Silence of hsa_circ_0001326 reversed the function of luteolin on macrophages polarization. In addition, hsa_circ_0001326 attenuated the inhibition effect of luteolin on hsa-miR-136-5p expression, and the promotion effect on USP4 expression. Furthermore, hsa-miR-136-5p inhibitor reversed the effect of hsa_circ_0001326 on macrophages polarization and the USP4 expression. Taken together, luteolin activates M2 macrophages and suppresses M1 macrophages by upregulation of hsa_circ_0001326. Further mechanism maybe by regulating hsa_circ_0001326 downstream gene expression, including hsa-miR-136-5p and USP4, in THP-1 derived macrophages. These findings provide a new insight for macrophage polarization under stimulation of luteolin.
Collapse
Affiliation(s)
- Benxin Gong
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| | - Ying Zheng
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| | - Jiahua Li
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| | - Huafeng Lei
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| | - Kexin Liu
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| | - Jingyun Tang
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| | - Yanrong Peng
- University of Chinese Academy of Sciences, Shenzhen Hospital East Hospital Pediatrics, Shenzhen City, China
| |
Collapse
|
5
|
Noordstra I, van den Berg CM, Boot FWJ, Katrukha EA, Yu KL, Tas RP, Portegies S, Viergever BJ, de Graaff E, Hoogenraad CC, de Koning EJP, Carlotti F, Kapitein LC, Akhmanova A. Organization and dynamics of the cortical complexes controlling insulin secretion in β-cells. J Cell Sci 2022; 135:274234. [PMID: 35006275 PMCID: PMC8918791 DOI: 10.1242/jcs.259430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/21/2021] [Indexed: 11/20/2022] Open
Abstract
Insulin secretion in pancreatic β-cells is regulated by cortical complexes that are enriched at the sites of adhesion to extracellular matrix facing the vasculature. Many components of these complexes, including bassoon, RIM, ELKS and liprins, are shared with neuronal synapses. Here, we show that insulin secretion sites also contain the non-neuronal proteins LL5β (also known as PHLDB2) and KANK1, which, in migrating cells, organize exocytotic machinery in the vicinity of integrin-based adhesions. Depletion of LL5β or focal adhesion disassembly triggered by myosin II inhibition perturbed the clustering of secretory complexes and attenuated the first wave of insulin release. Although previous analyses in vitro and in neurons have suggested that secretory machinery might assemble through liquid–liquid phase separation, analysis of endogenously labeled ELKS in pancreatic islets indicated that its dynamics is inconsistent with such a scenario. Instead, fluorescence recovery after photobleaching and single-molecule imaging showed that ELKS turnover is driven by binding and unbinding to low-mobility scaffolds. Both the scaffold movements and ELKS exchange were stimulated by glucose treatment. Our findings help to explain how integrin-based adhesions control spatial organization of glucose-stimulated insulin release. Summary: Characterization of the composition of cortical complexes controlling insulin secretion, showing that their dynamics is inconsistent with assembly through liquid–liquid phase separation.
Collapse
Affiliation(s)
- Ivar Noordstra
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands.,Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Cyntha M van den Berg
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Fransje W J Boot
- Department of Internal Medicine, Nephrology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Eugene A Katrukha
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Ka Lou Yu
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Roderick P Tas
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Sybren Portegies
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Bastiaan J Viergever
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Esther de Graaff
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Nephrology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Nephrology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
6
|
Tks5 Regulates Synaptic Podosome Formation and Stabilization of the Postsynaptic Machinery at the Neuromuscular Junction. Int J Mol Sci 2021; 22:ijms222112051. [PMID: 34769479 PMCID: PMC8585010 DOI: 10.3390/ijms222112051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Currently, the etiology of many neuromuscular disorders remains unknown. Many of them are characterized by aberrations in the maturation of the neuromuscular junction (NMJ) postsynaptic machinery. Unfortunately, the molecular factors involved in this process are still largely unknown, which poses a great challenge for identifying potential therapeutic targets. Here, we identified Tks5 as a novel interactor of αdystrobrevin-1, which is a crucial component of the NMJ postsynaptic machinery. Tks5 has been previously shown in cancer cells to be an important regulator of actin-rich structures known as invadosomes. However, a role of this scaffold protein at a synapse has never been studied. We show that Tks5 is crucial for remodeling of the NMJ postsynaptic machinery by regulating the organization of structures similar to the invadosomes, known as synaptic podosomes. Additionally, it is involved in the maintenance of the integrity of acetylcholine receptor (AChR) clusters and regulation of their turnover. Lastly, our data indicate that these Tks5 functions may be mediated by its involvement in recruitment of actin filaments to the postsynaptic machinery. Collectively, we show for the first time that the Tks5 protein is involved in regulation of the postsynaptic machinery.
Collapse
|
7
|
Alvarez-Suarez P, Nowak N, Protasiuk-Filipunas A, Yamazaki H, Prószyński TJ, Gawor M. Drebrin Regulates Acetylcholine Receptor Clustering and Organization of Microtubules at the Postsynaptic Machinery. Int J Mol Sci 2021; 22:9387. [PMID: 34502296 PMCID: PMC8430516 DOI: 10.3390/ijms22179387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 01/07/2023] Open
Abstract
Proper muscle function depends on the neuromuscular junctions (NMJs), which mature postnatally to complex "pretzel-like" structures, allowing for effective synaptic transmission. Postsynaptic acetylcholine receptors (AChRs) at NMJs are anchored in the actin cytoskeleton and clustered by the scaffold protein rapsyn, recruiting various actin-organizing proteins. Mechanisms driving the maturation of the postsynaptic machinery and regulating rapsyn interactions with the cytoskeleton are still poorly understood. Drebrin is an actin and microtubule cross-linker essential for the functioning of the synapses in the brain, but its role at NMJs remains elusive. We used immunohistochemistry, RNA interference, drebrin inhibitor 3,5-bis-trifluoromethyl pyrazole (BTP2) and co-immunopreciptation to explore the role of this protein at the postsynaptic machinery. We identify drebrin as a postsynaptic protein colocalizing with the AChRs both in vitro and in vivo. We also show that drebrin is enriched at synaptic podosomes. Downregulation of drebrin or blocking its interaction with actin in cultured myotubes impairs the organization of AChR clusters and the cluster-associated microtubule network. Finally, we demonstrate that drebrin interacts with rapsyn and a drebrin interactor, plus-end-tracking protein EB3. Our results reveal an interplay between drebrin and cluster-stabilizing machinery involving rapsyn, actin cytoskeleton, and microtubules.
Collapse
Affiliation(s)
- Paloma Alvarez-Suarez
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Natalia Nowak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Anna Protasiuk-Filipunas
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Hiroyuki Yamazaki
- Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan;
| | - Tomasz J. Prószyński
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| | - Marta Gawor
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (P.A.-S.); (N.N.); (A.P.-F.); (T.J.P.)
| |
Collapse
|
8
|
Han X, Zhang C, Ma X, Yan X, Xiong B, Shen W, Yin S, Zhang H, Sun Q, Zhao Y. Muscarinic acetylcholine receptor M5 is involved in spermatogenesis through the modification of cell-cell junctions. Reproduction 2021; 162:47-59. [PMID: 33970124 PMCID: PMC8183636 DOI: 10.1530/rep-21-0079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023]
Abstract
Muscarinic acetylcholine receptor (mAChR) antagonists have been reported to decrease male fertility; however, the roles of mAChRs in spermatogenesis and the underlying mechanisms are not understood yet. During spermatogenesis, extensive remodeling between Sertoli cells and/or germ cells interfaces takes place to accommodate the transport of developing germ cells across the blood-testis barrier (BTB) and adluminal compartment. The cell–cell junctions play a vital role in the spermatogenesis process. This study used ICR male mice and spermatogonial cells (C18-4) and Sertoli cells (TM-4). shRNA of control or M5 gene was injected into 5-week-old ICR mice testes. Ten days post-viral grafting, mice were deeply anesthetized with pentobarbital and the testes were collected. One testicle was fresh frozen for RNA-seq analysis or Western blotting (WB). The second testicle was fixed for immunofluorescence staining (IHF). C18-4 or TM-4 cells were treated with shRNA of control or M5 gene. Then, the cells were collected for RNA-seq analysis, WB, or IHF. Knockdown of mAChR M5 disrupted mouse spermatogenesis and damaged the actin-based cytoskeleton and many types of junction proteins in both Sertoli cells and germ cells. M5 knockdown decreased Phldb2 expression in both germ cells and Sertoli cells which suggested that Phldb2 may be involved in cytoskeleton and cell–cell junction formation to regulate spermatogenesis. Our investigation has elucidated a novel role for mAChR M5 in the regulation of spermatogenesis through the interactions of Phldb2 and cell–cell junctions. M5 may be an attractive future therapeutic target in the treatment of male reproductive disorders.
Collapse
Affiliation(s)
- Xiao Han
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Cong Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Xiangping Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Xiaowei Yan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Bohui Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Shen Yin
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Qingyuan Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, People's Republic of China
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| |
Collapse
|
9
|
Petrany MJ, Swoboda CO, Sun C, Chetal K, Chen X, Weirauch MT, Salomonis N, Millay DP. Single-nucleus RNA-seq identifies transcriptional heterogeneity in multinucleated skeletal myofibers. Nat Commun 2020; 11:6374. [PMID: 33311464 PMCID: PMC7733460 DOI: 10.1038/s41467-020-20063-w] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
While the majority of cells contain a single nucleus, cell types such as trophoblasts, osteoclasts, and skeletal myofibers require multinucleation. One advantage of multinucleation can be the assignment of distinct functions to different nuclei, but comprehensive interrogation of transcriptional heterogeneity within multinucleated tissues has been challenging due to the presence of a shared cytoplasm. Here, we utilized single-nucleus RNA-sequencing (snRNA-seq) to determine the extent of transcriptional diversity within multinucleated skeletal myofibers. Nuclei from mouse skeletal muscle were profiled across the lifespan, which revealed the presence of distinct myonuclear populations emerging in postnatal development as well as aging muscle. Our datasets also provided a platform for discovery of genes associated with rare specialized regions of the muscle cell, including markers of the myotendinous junction and functionally validated factors expressed at the neuromuscular junction. These findings reveal that myonuclei within syncytial muscle fibers possess distinct transcriptional profiles that regulate muscle biology.
Collapse
Affiliation(s)
- Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Lin SS, Hsieh TL, Liou GG, Li TN, Lin HC, Chang CW, Wu HY, Yao CK, Liu YW. Dynamin-2 Regulates Postsynaptic Cytoskeleton Organization and Neuromuscular Junction Development. Cell Rep 2020; 33:108310. [DOI: 10.1016/j.celrep.2020.108310] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 11/30/2022] Open
|
11
|
Brzoska E, Kalkowski L, Kowalski K, Michalski P, Kowalczyk P, Mierzejewski B, Walczak P, Ciemerych MA, Janowski M. Muscular Contribution to Adolescent Idiopathic Scoliosis from the Perspective of Stem Cell-Based Regenerative Medicine. Stem Cells Dev 2020; 28:1059-1077. [PMID: 31170887 DOI: 10.1089/scd.2019.0073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a relatively frequent disease within a range 0.5%-5.0% of population, with higher frequency in females. While a resultant spinal deformity is usually medically benign condition, it produces far going psychosocial consequences, which warrant attention. The etiology of AIS is unknown and current therapeutic approaches are symptomatic only, and frequently inconvenient or invasive. Muscular contribution to AIS is widely recognized, although it did not translate to clinical routine as yet. Muscle asymmetry has been documented by pathological examinations as well as systemic muscle disorders frequently leading to scoliosis. It has been also reported numerous genetic, metabolic and radiological alterations in patients with AIS, which are linked to muscular and neuromuscular aspects. Therefore, muscles might be considered an attractive and still insufficiently exploited therapeutic target for AIS. Stem cell-based regenerative medicine is rapidly gaining momentum based on the tremendous progress in understanding of developmental biology. It comes also with a toolbox of various stem cells such as satellite cells or mesenchymal stem cells, which could be transplanted; also, the knowledge acquired in research on regenerative medicine can be applied to manipulation of endogenous stem cells to obtain desired therapeutic goals. Importantly, paravertebral muscles are located relatively superficially; therefore, they can be an easy target for minimally invasive approaches to treatment of AIS. It comes in pair with a fast progress in image guidance, which allows for precise delivery of therapeutic agents, including stem cells to various organs such as brain, muscles, and others. Summing up, it seems that there is a link between AIS, muscles, and stem cells, which might be worth of further investigations with a long-term goal of setting foundations for eventual bench-to-bedside translation.
Collapse
Affiliation(s)
- Edyta Brzoska
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Lukasz Kalkowski
- 2Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamil Kowalski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Pawel Michalski
- 3Spine Surgery Department, Institute of Mother and Child, Warsaw, Poland
| | - Pawel Kowalczyk
- 4Department of Neurosurgery, Children's Memorial Health Institute, Warsaw, Poland
| | - Bartosz Mierzejewski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Piotr Walczak
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria A Ciemerych
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Miroslaw Janowski
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Bernadzki KM, Daszczuk P, Rojek KO, Pęziński M, Gawor M, Pradhan BS, de Cicco T, Bijata M, Bijata K, Włodarczyk J, Prószyński TJ, Niewiadomski P. Arhgef5 Binds α-Dystrobrevin 1 and Regulates Neuromuscular Junction Integrity. Front Mol Neurosci 2020; 13:104. [PMID: 32587503 PMCID: PMC7299196 DOI: 10.3389/fnmol.2020.00104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/13/2020] [Indexed: 01/09/2023] Open
Abstract
The neuromuscular junctions (NMJs) connect muscle fibers with motor neurons and enable the coordinated contraction of skeletal muscles. The dystrophin-associated glycoprotein complex (DGC) is an essential component of the postsynaptic machinery of the NMJ and is important for the maintenance of NMJ structural integrity. To identify novel proteins that are important for NMJ organization, we performed a mass spectrometry-based screen for interactors of α-dystrobrevin 1 (aDB1), one of the components of the DGC. The guanidine nucleotide exchange factor (GEF) Arhgef5 was found to be one of the aDB1 binding partners that is recruited to Tyr-713 in a phospho-dependent manner. We show here that Arhgef5 localizes to the NMJ and that its genetic depletion in the muscle causes the fragmentation of the synapses in conditional knockout mice. Arhgef5 loss in vivo is associated with a reduction in the levels of active GTP-bound RhoA and Cdc42 GTPases, highlighting the importance of actin dynamics regulation for the maintenance of NMJ integrity.
Collapse
Affiliation(s)
- Krzysztof M Bernadzki
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Patrycja Daszczuk
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Katarzyna O Rojek
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland.,Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Marcin Pęziński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marta Gawor
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bhola S Pradhan
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Teresa de Cicco
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Monika Bijata
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Krystian Bijata
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Jakub Włodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Tomasz J Prószyński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland.,Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Paweł Niewiadomski
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Warsaw, Poland.,Laboratory of Molecular and Cellular Signaling, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
13
|
Han Z, Ruthel G, Dash S, Berry CT, Freedman BD, Harty RN, Shtanko O. Angiomotin regulates budding and spread of Ebola virus. J Biol Chem 2020; 295:8596-8601. [PMID: 32381509 DOI: 10.1074/jbc.ac120.013171] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/05/2020] [Indexed: 12/17/2022] Open
Abstract
The Ebola virus (EBOV) VP40 matrix protein (eVP40) orchestrates assembly and budding of virions in part by hijacking select WW-domain-bearing host proteins via its PPxY late (L)-domain motif. Angiomotin (Amot) is a multifunctional PPxY-containing adaptor protein that regulates angiogenesis, actin dynamics, and cell migration/motility. Amot also regulates the Hippo signaling pathway via interactions with the WW-domain-containing Hippo effector protein Yes-associated protein (YAP). In this report, we demonstrate that endogenous Amot is crucial for positively regulating egress of eVP40 virus-like particles (VLPs) and for egress and spread of authentic EBOV. Mechanistically, we show that ectopic YAP expression inhibits eVP40 VLP egress and that Amot co-expression rescues budding of eVP40 VLPs in a dose-dependent and PPxY-dependent manner. Moreover, results obtained with confocal and total internal reflection fluorescence microscopy suggested that Amot's role in actin organization and dynamics also contributes to promoting eVP40-mediated egress. In summary, these findings reveal a functional and competitive interplay between virus and host proteins involving the multifunctional PPxY-containing adaptor Amot, which regulates both the Hippo pathway and actin dynamics. We propose that our results have wide-ranging implications for understanding the biology and pathology of EBOV infections.
Collapse
Affiliation(s)
- Ziying Han
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shantoshini Dash
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Corbett T Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
14
|
Pęziński M, Daszczuk P, Pradhan BS, Lochmüller H, Prószyński TJ. An improved method for culturing myotubes on laminins for the robust clustering of postsynaptic machinery. Sci Rep 2020; 10:4524. [PMID: 32161296 PMCID: PMC7066178 DOI: 10.1038/s41598-020-61347-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 02/20/2020] [Indexed: 01/03/2023] Open
Abstract
Motor neurons form specialized synapses with skeletal muscle fibers, called neuromuscular junctions (NMJs). Cultured myotubes are used as a simplified in vitro system to study the postsynaptic specialization of muscles. The stimulation of myotubes with the glycoprotein agrin or laminin-111 induces the clustering of postsynaptic machinery that contains acetylcholine receptors (AChRs). When myotubes are grown on laminin-coated surfaces, AChR clusters undergo developmental remodeling to form topologically complex structures that resemble mature NMJs. Needing further exploration are the molecular processes that govern AChR cluster assembly and its developmental maturation. Here, we describe an improved protocol for culturing muscle cells to promote the formation of complex AChR clusters. We screened various laminin isoforms and showed that laminin-221 was the most potent for inducing AChR clusters, whereas laminin-121, laminin-211, and laminin-221 afforded the highest percentages of topologically complex assemblies. Human primary myotubes that were formed by myoblasts obtained from patient biopsies also assembled AChR clusters that underwent remodeling in vitro. Collectively, these results demonstrate an advancement of culturing myotubes that can facilitate high-throughput screening for potential therapeutic targets for neuromuscular disorders.
Collapse
Affiliation(s)
- Marcin Pęziński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Patrycja Daszczuk
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bhola Shankar Pradhan
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, Germany.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Tomasz J Prószyński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland. .,Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
15
|
Seetharaman S, Etienne-Manneville S. Microtubules at focal adhesions – a double-edged sword. J Cell Sci 2019; 132:132/19/jcs232843. [DOI: 10.1242/jcs.232843] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ABSTRACT
Cell adhesion to the extracellular matrix is essential for cellular processes, such as migration and invasion. In response to cues from the microenvironment, integrin-mediated adhesions alter cellular behaviour through cytoskeletal rearrangements. The tight association of the actin cytoskeleton with adhesive structures has been extensively studied, whereas the microtubule network in this context has gathered far less attention. In recent years, however, microtubules have emerged as key regulators of cell adhesion and migration through their participation in adhesion turnover and cellular signalling. In this Review, we focus on the interactions between microtubules and integrin-mediated adhesions, in particular, focal adhesions and podosomes. Starting with the association of microtubules with these adhesive structures, we describe the classical role of microtubules in vesicular trafficking, which is involved in the turnover of cell adhesions, before discussing how microtubules can also influence the actin–focal adhesion interplay through RhoGTPase signalling, thereby orchestrating a very crucial crosstalk between the cytoskeletal networks and adhesions.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France
- Université Paris Descartes, Center for Research and Interdisciplinarity, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France
| |
Collapse
|
16
|
Rojek KO, Krzemień J, Doleżyczek H, Boguszewski PM, Kaczmarek L, Konopka W, Rylski M, Jaworski J, Holmgren L, Prószyński TJ. Amot and Yap1 regulate neuronal dendritic tree complexity and locomotor coordination in mice. PLoS Biol 2019; 17:e3000253. [PMID: 31042703 PMCID: PMC6513106 DOI: 10.1371/journal.pbio.3000253] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 05/13/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022] Open
Abstract
The angiomotin (Amot)-Yes-associated protein 1 (Yap1) complex plays a major role in regulating the inhibition of cell contact, cellular polarity, and cell growth in many cell types. However, the function of Amot and the Hippo pathway transcription coactivator Yap1 in the central nervous system remains unclear. We found that Amot is a critical mediator of dendritic morphogenesis in cultured hippocampal cells and Purkinje cells in the brain. Amot function in developing neurons depends on interactions with Yap1, which is also indispensable for dendrite growth and arborization in vitro. The conditional deletion of Amot and Yap1 in neurons led to a decrease in the complexity of Purkinje cell dendritic trees, abnormal cerebellar morphology, and impairments in motor coordination. Our results indicate that the function of Amot and Yap1 in dendrite growth does not rely on interactions with TEA domain (TEAD) transcription factors or the expression of Hippo pathway-dependent genes. Instead, Amot and Yap1 regulate dendrite development by affecting the phosphorylation of S6 kinase and its target S6 ribosomal protein.
Collapse
Affiliation(s)
- Katarzyna O. Rojek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Krzemień
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Hubert Doleżyczek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Paweł M. Boguszewski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Leszek Kaczmarek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Witold Konopka
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marcin Rylski
- Centre of Postgraduate Medical Education, Warsaw, Poland
- Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | - Tomasz J. Prószyński
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- * E-mail:
| |
Collapse
|
17
|
Histone deacetylase 4 protects from denervation and skeletal muscle atrophy in a murine model of amyotrophic lateral sclerosis. EBioMedicine 2019; 40:717-732. [PMID: 30713114 PMCID: PMC6414308 DOI: 10.1016/j.ebiom.2019.01.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
Background Histone deacetylase 4 (HDAC4) has been proposed as a target for Amyotrophic Lateral Sclerosis (ALS) because it mediates nerve-skeletal muscle interaction and since its expression in skeletal muscle correlates with the severity of the disease. However, our recent studies on the skeletal muscle response upon long-term denervation highlighted the importance of HDAC4 in maintaining muscle integrity. Methods To fully identify the yet uncharacterized HDAC4 functions in ALS, we genetically deleted HDAC4 in skeletal muscles of a mouse model of ALS. Body weight, skeletal muscle, innervation and spinal cord were analyzed over time by morphological and molecular analyses. Transcriptome analysis was also performed to delineate the signaling modulated by HDAC4 in skeletal muscle of a mouse model of ALS. Findings HDAC4 deletion in skeletal muscle caused earlier ALS onset, characterized by body weight loss, muscle denervation and atrophy, and compromised muscle performance, although the main catabolic pathways were not activated. Transcriptome analysis identified the gene networks modulated by HDAC4 in ALS, revealing UCP1 as a top regulator that may be implicated in worsening ALS features. Interpretation HDAC4 plays an important role in preserving innervations and skeletal muscle in ALS, likely by modulating the UCP1 gene network. Our study highlights a possible risk in considering HDAC inhibitors for the treatment of ALS. Fund This work was supported by FIRB grant (RBFR12BUMH) from Ministry of Education, Universities and Research, by Fondazione Veronesi, by Sapienza research project 2017 (RM11715C78539BD8) and Polish National Science Center grant (UMO-2016/21/B/NZ3/03638).
Collapse
|
18
|
van Gurp L, Muraro MJ, Dielen T, Seneby L, Dharmadhikari G, Gradwohl G, van Oudenaarden A, de Koning EJP. A transcriptomic roadmap to alpha- and beta cell differentiation in the embryonic pancreas. Development 2019; 146:dev.173716. [DOI: 10.1242/dev.173716] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/24/2019] [Indexed: 12/13/2022]
Abstract
During pancreatic development, endocrine cells appear from the pancreatic epithelium when Neurog3 positive cells delaminate and differentiate into alpha, beta, gamma and delta cells. The mechanisms involved in this process are still incompletely understood. We characterized the temporal, lineage-specific developmental programs during pancreatic development by sequencing the transcriptome of thousands of individual pancreatic cells from embryonic day E12.5 to E18.5 in mice, and identified all known cell types that are present in the embryonic pancreas, but focused specifically on alpha and beta cell differentiation by enrichment of a MIP-GFP reporter. We characterized transcriptomic heterogeneity in the tip domain based on proliferation, and characterized two endocrine precursor clusters marked by expression of Neurog3 and Fev. Pseudotime analysis revealed specific branches for developing alpha- and beta cells, which allowed identification of specific gene regulation patterns. These include some known and many previously unreported genes that appear to define pancreatic cell fate transitions. This resource allows dynamic profiling of embryonic pancreas development at single cell resolution and reveals novel gene signatures during pancreatic differentiation into alpha and beta cells.
Collapse
Affiliation(s)
- Léon van Gurp
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
| | - Mauro J. Muraro
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
- Single Cell Discoveries, Utrecht, the Netherlands
| | - Tim Dielen
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
| | - Lina Seneby
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
| | - Gitanjali Dharmadhikari
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
| | - Gerard Gradwohl
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Alexander van Oudenaarden
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
- Single Cell Discoveries, Utrecht, the Netherlands
- Oncode Institute, the Netherlands
| | - Eelco J. P. de Koning
- Hubrecht Institute\KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
19
|
|
20
|
Huang T, Zhou Y, Zhang J, Cheng ASL, Yu J, To KF, Kang W. The physiological role of Motin family and its dysregulation in tumorigenesis. J Transl Med 2018; 16:98. [PMID: 29650031 PMCID: PMC5898069 DOI: 10.1186/s12967-018-1466-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/28/2018] [Indexed: 11/30/2022] Open
Abstract
Members in Motin family, or Angiomotins (AMOTs), are adaptor proteins that localize in the membranous, cytoplasmic or nuclear fraction in a cell context-dependent manner. They control the bioprocesses such as migration, tight junction formation, cell polarity, and angiogenesis. Emerging evidences have demonstrated that AMOTs participate in cancer initiation and progression. Many of the previous studies have focused on the involvement of AMOTs in Hippo-YAP1 pathway. However, it has been controversial for years that AMOTs serve as either positive or negative growth regulators in different cancer types because of the various cellular origins. The molecular mechanisms of these opposite roles of AMOTs remain elusive. This review comprehensively summarized how AMOTs function physiologically and how their dysregulation promotes or inhibits tumorigenesis. Better understanding the functional roles of AMOTs in cancers may lead to an improvement of clinical interventions as well as development of novel therapeutic strategies for cancer patients.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China
| | - Jinglin Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China
| | - Alfred S L Cheng
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| |
Collapse
|
21
|
Affiliation(s)
- Lei Li
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Wen-Cheng Xiong
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| | - Lin Mei
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
22
|
Sala K, Raimondi A, Tonoli D, Tacchetti C, de Curtis I. Identification of a membrane-less compartment regulating invadosome function and motility. Sci Rep 2018; 8:1164. [PMID: 29348417 PMCID: PMC5773524 DOI: 10.1038/s41598-018-19447-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023] Open
Abstract
Depletion of liprin-α1, ERC1 or LL5 scaffolds inhibits extracellular matrix degradation by invasive cells. These proteins co-accumulate near invadosomes in NIH-Src cells, identifying a novel invadosome–associated compartment distinct from the core and adhesion ring of invadosomes. Depletion of either protein perturbs the organization of invadosomes without influencing the recruitment of MT1-MMP metalloprotease. Liprin-α1 is not required for de novo formation of invadosomes after their disassembly by microtubules and Src inhibitors, while its depletion inhibits invadosome motility, thus affecting matrix degradation. Fluorescence recovery after photobleaching shows that the invadosome–associated compartment is dynamic, while correlative light immunoelectron microscopy identifies bona fide membrane–free invadosome–associated regions enriched in liprin-α1, which is virtually excluded from the invadosome core. The results indicate that liprin-α1, LL5 and ERC1 define a novel dynamic membrane-less compartment that regulates matrix degradation by affecting invadosome motility.
Collapse
Affiliation(s)
- Kristyna Sala
- Cell Adhesion Unit - Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132, Milano, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCSS San Raffaele Scientific Institute, 20132, Milano, Italy
| | - Diletta Tonoli
- Cell Adhesion Unit - Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132, Milano, Italy
| | - Carlo Tacchetti
- Experimental Imaging Center, IRCSS San Raffaele Scientific Institute, 20132, Milano, Italy.,San Raffaele Vita-Salute University, via Olgettina 58, 20132, Milano, Italy
| | - Ivan de Curtis
- Cell Adhesion Unit - Division of Neuroscience, IRCSS San Raffaele Scientific Institute, 20132, Milano, Italy. .,San Raffaele Vita-Salute University, via Olgettina 58, 20132, Milano, Italy.
| |
Collapse
|
23
|
Bernadzki KM, Gawor M, Pęziński M, Mazurek P, Niewiadomski P, Rędowicz MJ, Prószyński TJ. Liprin-α-1 is a novel component of the murine neuromuscular junction and is involved in the organization of the postsynaptic machinery. Sci Rep 2017; 7:9116. [PMID: 28831123 PMCID: PMC5567263 DOI: 10.1038/s41598-017-09590-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/25/2017] [Indexed: 01/26/2023] Open
Abstract
Neuromuscular junctions (NMJs) are specialized synapses that connect motor neurons to skeletal muscle fibers and orchestrate proper signal transmission from the nervous system to muscles. The efficient formation and maintenance of the postsynaptic machinery that contains acetylcholine receptors (AChR) are indispensable for proper NMJ function. Abnormalities in the organization of synaptic components often cause severe neuromuscular disorders, such as muscular dystrophy. The dystrophin-associated glycoprotein complex (DGC) was shown to play an important role in NMJ development. We recently identified liprin-α-1 as a novel binding partner for one of the cytoplasmic DGC components, α-dystrobrevin-1. In the present study, we performed a detailed analysis of localization and function of liprin-α-1 at the murine NMJ. We showed that liprin-α-1 localizes to both pre- and postsynaptic compartments at the NMJ, and its synaptic enrichment depends on the presence of the nerve. Using cultured muscle cells, we found that liprin-α-1 plays an important role in AChR clustering and the organization of cortical microtubules. Our studies provide novel insights into the function of liprin-α-1 at vertebrate neuromuscular synapses.
Collapse
Affiliation(s)
- Krzysztof M Bernadzki
- Laboratory of Synaptogenesis, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland
| | - Marta Gawor
- Laboratory of Synaptogenesis, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland
| | - Marcin Pęziński
- Laboratory of Synaptogenesis, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland
| | - Paula Mazurek
- Laboratory of Synaptogenesis, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland
| | - Paweł Niewiadomski
- Laboratory of Synaptogenesis, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland
| | - Maria J Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland
| | - Tomasz J Prószyński
- Laboratory of Synaptogenesis, Polish Academy of Sciences, 3 Pasteura Street, Warsaw, 02-093, Poland.
| |
Collapse
|
24
|
Lv M, Shen Y, Yang J, Li S, Wang B, Chen Z, Li P, Liu P, Yang J. Angiomotin Family Members: Oncogenes or Tumor Suppressors? Int J Biol Sci 2017; 13:772-781. [PMID: 28656002 PMCID: PMC5485632 DOI: 10.7150/ijbs.19603] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/09/2017] [Indexed: 12/17/2022] Open
Abstract
Angiomotin (Amot) family contains three members: Amot (p80 and p130 isoforms), Amot-like protein 1 (Amotl1), and Amot-like protein 2 (Amotl2). Amot proteins play an important role in tube formation and migration of endothelial cells and the regulation of tight junctions, polarity, and epithelial-mesenchymal transition in epithelial cells. Moreover, these proteins regulate the proliferation and migration of cancer cells. In most cancers, Amot family members promote the proliferation and invasion of cancer cells, including breast cancer, osteosarcoma, colon cancer, prostate cancer, head and neck squamous cell carcinoma, cervical cancer, liver cancer, and renal cell cancer. However, in glioblastoma, ovarian cancer, and lung cancer, Amot inhibits the growth of cancer cells. In addition, there are controversies on the regulation of Yes-associated protein (YAP) by Amot. Amot promotes either the internalization of YAP into the nucleus or the retention of YAP in the cytoplasm of different cell types. Moreover, Amot regulates the AMPK, mTOR, Wnt, and MAPK signaling pathways. However, it is unclear whether Amot is an oncogene or a tumor suppressor gene in different cellular processes. This review focuses on the multifunctional roles of Amot in cancers.
Collapse
Affiliation(s)
- Meng Lv
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Yanwei Shen
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Jiao Yang
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Shuting Li
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Biyuan Wang
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Zheling Chen
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Pan Li
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Peijun Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Yang
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| |
Collapse
|
25
|
Abstract
Exocytosis is a fundamental cellular process whereby secreted molecules are packaged into vesicles that move along cytoskeletal filaments and fuse with the plasma membrane. To function optimally, cells are strongly dependent on precisely controlled delivery of exocytotic cargo. In mammalian cells, microtubules serve as major tracks for vesicle transport by motor proteins, and thus microtubule organization is important for targeted delivery of secretory carriers. Over the years, multiple microtubule-associated and cortical proteins have been discovered that facilitate the interaction between the microtubule plus ends and the cell cortex. In this review, we focus on mammalian protein complexes that have been shown to participate in both cortical microtubule capture and exocytosis, thereby regulating the spatial organization of secretion. These complexes include microtubule plus-end tracking proteins, scaffolding factors, actin-binding proteins, and components of vesicle docking machinery, which together allow efficient coordination of cargo transport and release.
Collapse
Affiliation(s)
- Ivar Noordstra
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| |
Collapse
|
26
|
Bouchet BP, Gough RE, Ammon YC, van de Willige D, Post H, Jacquemet G, Altelaar AM, Heck AJ, Goult BT, Akhmanova A. Talin-KANK1 interaction controls the recruitment of cortical microtubule stabilizing complexes to focal adhesions. eLife 2016; 5. [PMID: 27410476 PMCID: PMC4995097 DOI: 10.7554/elife.18124] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/12/2016] [Indexed: 12/23/2022] Open
Abstract
The cross-talk between dynamic microtubules and integrin-based adhesions to the extracellular matrix plays a crucial role in cell polarity and migration. Microtubules regulate the turnover of adhesion sites, and, in turn, focal adhesions promote the cortical microtubule capture and stabilization in their vicinity, but the underlying mechanism is unknown. Here, we show that cortical microtubule stabilization sites containing CLASPs, KIF21A, LL5β and liprins are recruited to focal adhesions by the adaptor protein KANK1, which directly interacts with the major adhesion component, talin. Structural studies showed that the conserved KN domain in KANK1 binds to the talin rod domain R7. Perturbation of this interaction, including a single point mutation in talin, which disrupts KANK1 binding but not the talin function in adhesion, abrogates the association of microtubule-stabilizing complexes with focal adhesions. We propose that the talin-KANK1 interaction links the two macromolecular assemblies that control cortical attachment of actin fibers and microtubules. DOI:http://dx.doi.org/10.7554/eLife.18124.001 Animal cells are organized into tissues and organs. A scaffold-like framework outside of the cells called the extracellular matrix provides support to the cells and helps to hold them in place. Cells attach to the extracellular matrix via structures called focal adhesions on the cell surface; these structures contain a protein called talin. For a cell to be able to move, the existing focal adhesions must be broken down and new adhesions allowed to form. This process is regulated by the delivery and removal of different materials along fibers called microtubules. Microtubules can usually grow and shrink rapidly, but near focal adhesions they are captured at the surface of the cell and become more stable. However, it is not clear how focal adhesions promote microtubule capture and stability. Bouchet et al. found that a protein called KANK1 binds to the focal adhesion protein talin in human cells grown in a culture dish. This allows KANK1 to recruit microtubules to the cell surface around the focal adhesions by binding to particular proteins that are associated with microtubules. Disrupting the interaction between KANK1 and talin by making small alterations in these two proteins blocked the ability of focal adhesions to capture surrounding microtubules. The next step following on from this work will be to find out whether this process also takes place in the cells within an animal’s body, such as a fly or a mouse. DOI:http://dx.doi.org/10.7554/eLife.18124.002
Collapse
Affiliation(s)
- Benjamin P Bouchet
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Rosemarie E Gough
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - York-Christoph Ammon
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Dieudonnée van de Willige
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics, Utrecht University, Utrecht, The Netherlands.,Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,The Netherlands Proteomics Centre, Utrecht University, Utrecht, The Netherlands
| | | | - Af Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Utrecht University, Utrecht, The Netherlands.,Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,The Netherlands Proteomics Centre, Utrecht University, Utrecht, The Netherlands
| | - Albert Jr Heck
- Biomolecular Mass Spectrometry and Proteomics, Utrecht University, Utrecht, The Netherlands.,Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,The Netherlands Proteomics Centre, Utrecht University, Utrecht, The Netherlands
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
27
|
Editors T. Muscle Decline in Aging and Neuromuscular Disorders - Mechanisms and Countermeasures: Terme Euganee, Padova (Italy), April 13-16, 2016. Eur J Transl Myol 2016; 26:5904. [PMID: 27054021 PMCID: PMC4821223 DOI: 10.4081/ejtm.2016.5904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Not available.
Collapse
|
28
|
Gingras J, Gawor M, Bernadzki KM, Grady RM, Hallock P, Glass DJ, Sanes JR, Proszynski TJ. Α-Dystrobrevin-1 recruits Grb2 and α-catulin to organize neurotransmitter receptors at the neuromuscular junction. J Cell Sci 2016; 129:898-911. [PMID: 26769899 DOI: 10.1242/jcs.181180] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/11/2016] [Indexed: 12/17/2022] Open
Abstract
Neuromuscular junctions (NMJs), the synapses made by motor neurons on muscle fibers, form during embryonic development but undergo substantial remodeling postnatally. Several lines of evidence suggest that α-dystrobrevin, a component of the dystrophin-associated glycoprotein complex (DGC), is a crucial regulator of the remodeling process and that tyrosine phosphorylation of one isoform, α-dystrobrevin-1, is required for its function at synapses. We identified a functionally important phosphorylation site on α-dystrobrevin-1, generated phosphorylation-specific antibodies to it and used them to demonstrate dramatic increases in phosphorylation during the remodeling period, as well as in nerve-dependent regulation in adults. We then identified proteins that bind to this site in a phosphorylation-dependent manner and others that bind to α-dystrobrevin-1 in a phosphorylation-independent manner. They include multiple members of the DGC, as well as α-catulin, liprin-α1, Usp9x, PI3K, Arhgef5 and Grb2. Finally, we show that two interactors, α-catulin (phosphorylation independent) and Grb2 (phosphorylation dependent) are localized to NMJs in vivo, and that they are required for proper organization of neurotransmitter receptors on myotubes.
Collapse
Affiliation(s)
- Jacinthe Gingras
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Marta Gawor
- Laboratory of Synaptogenesis, Dept. of Cell Biology, Nencki Institute of Experimental Biology, Warsaw 02-093, Poland
| | - Krzysztof M Bernadzki
- Laboratory of Synaptogenesis, Dept. of Cell Biology, Nencki Institute of Experimental Biology, Warsaw 02-093, Poland
| | - R Mark Grady
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Peter Hallock
- Novartis Biomedical Institute, Cambridge, MA 02139, USA
| | - David J Glass
- Novartis Biomedical Institute, Cambridge, MA 02139, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Tomasz J Proszynski
- Laboratory of Synaptogenesis, Dept. of Cell Biology, Nencki Institute of Experimental Biology, Warsaw 02-093, Poland
| |
Collapse
|
29
|
Sorbs1 and -2 Interact with CrkL and Are Required for Acetylcholine Receptor Cluster Formation. Mol Cell Biol 2015; 36:262-70. [PMID: 26527617 PMCID: PMC4719301 DOI: 10.1128/mcb.00775-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/23/2015] [Indexed: 01/30/2023] Open
Abstract
Crk and CrkL are noncatalytic adaptor proteins necessary for the formation of neuromuscular synapses which function downstream of muscle-specific kinase (MuSK), a receptor tyrosine kinase expressed in skeletal muscle, and the MuSK binding protein Dok-7. How Crk/CrkL regulate neuromuscular endplate formation is not known. To better understand the roles of Crk/CrkL, we identified CrkL binding proteins using mass spectrometry and have identified Sorbs1 and Sorbs2 as two functionally redundant proteins that associate with the initiating MuSK/Dok-7/Crk/CrkL complex, regulate acetylcholine receptor (AChR) clustering in vitro, and are localized at synapses in vivo.
Collapse
|
30
|
Involvement of unconventional myosin VI in myoblast function and myotube formation. Histochem Cell Biol 2015; 144:21-38. [PMID: 25896210 PMCID: PMC4469105 DOI: 10.1007/s00418-015-1322-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2015] [Indexed: 01/01/2023]
Abstract
The important role of unconventional myosin VI (MVI) in skeletal and cardiac muscle has been recently postulated (Karolczak et al. in Histochem Cell Biol 139:873-885, 2013). Here, we addressed for the first time a role for this unique myosin motor in myogenic cells as well as during their differentiation into myotubes. During myoblast differentiation, the isoform expression pattern of MVI and its subcellular localization underwent changes. In undifferentiated myoblasts, MVI-stained puncti were seen throughout the cytoplasm and were in close proximity to actin filaments, Golgi apparatus, vinculin-, and talin-rich focal adhesion as well as endoplasmic reticulum. Colocalization of MVI with endoplasmic reticulum was enhanced during myotube formation, and differentiation-dependent association was also seen in sarcoplasmic reticulum of neonatal rat cardiomyocytes (NRCs). Moreover, we observed enrichment of MVI in myotube regions containing acetylcholine receptor-rich clusters, suggesting its involvement in the organization of the muscle postsynaptic machinery. Overexpression of the H246R MVI mutant (associated with hypertrophic cardiomyopathy) in myoblasts and NRCs caused the formation of abnormally large intracellular vesicles. MVI knockdown caused changes in myoblast morphology and inhibition of their migration. On the subcellular level, MVI-depleted myoblasts exhibited aberrations in the organization of actin cytoskeleton and adhesive structures as well as in integrity of Golgi apparatus and endoplasmic reticulum. Also, MVI depletion or overexpression of H246R mutant caused the formation of significantly wider or aberrant myotubes, respectively, indicative of involvement of MVI in myoblast differentiation. The presented results suggest an important role for MVI in myogenic cells and possibly in myoblast differentiation.
Collapse
|
31
|
Astro V, de Curtis I. Plasma membrane-associated platforms: Dynamic scaffolds that organize membrane-associated events. Sci Signal 2015; 8:re1. [DOI: 10.1126/scisignal.aaa3312] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Basu S, Sladecek S, Martinez de la Peña y Valenzuela I, Akaaboune M, Smal I, Martin K, Galjart N, Brenner HR. CLASP2-dependent microtubule capture at the neuromuscular junction membrane requires LL5β and actin for focal delivery of acetylcholine receptor vesicles. Mol Biol Cell 2015; 26:938-51. [PMID: 25589673 PMCID: PMC4342029 DOI: 10.1091/mbc.e14-06-1158] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A novel mechanism is described for the agrin-mediated focal delivery of acetylcholine receptors (AChRs) to the postsynaptic membrane of the neuromuscular junction. Microtubule capture mediated by CLASP2 and its interaction partner, LL5β, and an intact subsynaptic actin cytoskeleton are both required for focal AChR transport to the synaptic membrane. A hallmark of the neuromuscular junction (NMJ) is the high density of acetylcholine receptors (AChRs) in the postsynaptic muscle membrane. The postsynaptic apparatus of the NMJ is organized by agrin secreted from motor neurons. The mechanisms that underlie the focal delivery of AChRs to the adult NMJ are not yet understood in detail. We previously showed that microtubule (MT) capture by the plus end–tracking protein CLASP2 regulates AChR density at agrin-induced AChR clusters in cultured myotubes via PI3 kinase acting through GSK3β. Here we show that knockdown of the CLASP2-interaction partner LL5β by RNAi and forced expression of a CLASP2 fragment blocking the CLASP2/LL5β interaction inhibit microtubule capture. The same treatments impair focal vesicle delivery to the clusters. Consistent with these findings, knockdown of LL5β at the NMJ in vivo reduces the density and insertion of AChRs into the postsynaptic membrane. MT capture and focal vesicle delivery to agrin-induced AChR clusters are also inhibited by microtubule- and actin-depolymerizing drugs, invoking both cytoskeletal systems in MT capture and in the fusion of AChR vesicles with the cluster membrane. Combined our data identify a transport system, organized by agrin through PI3 kinase, GSK3β, CLASP2, and LL5β, for precise delivery of AChR vesicles from the subsynaptic nuclei to the overlying synaptic membrane.
Collapse
Affiliation(s)
- Sreya Basu
- Department of Biomedicine, University of Basel, CH-4056 Basel, Switzerland Department of Cell Biology, Erasmus Medical Center, 3015 GE Rotterdam, Netherlands
| | - Stefan Sladecek
- Department of Biomedicine, University of Basel, CH-4056 Basel, Switzerland
| | | | - Mohammed Akaaboune
- Department of Molecular, Cellular, and Developmental Biology and Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109
| | - Ihor Smal
- Biomedical Imaging Group, Erasmus Medical Center, 3015 GE Rotterdam, Netherlands
| | - Katrin Martin
- Department of Biomedicine, University of Basel, CH-4056 Basel, Switzerland
| | - Niels Galjart
- Department of Cell Biology, Erasmus Medical Center, 3015 GE Rotterdam, Netherlands
| | | |
Collapse
|
33
|
Bernadzki KM, Rojek KO, Prószyński TJ. Podosomes in muscle cells and their role in the remodeling of neuromuscular postsynaptic machinery. Eur J Cell Biol 2014; 93:478-85. [DOI: 10.1016/j.ejcb.2014.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/23/2014] [Accepted: 06/05/2014] [Indexed: 11/28/2022] Open
|
34
|
The expression and function of gelatinolytic activity at the rat neuromuscular junction upon physical exercise. Histochem Cell Biol 2014; 143:143-52. [PMID: 25212659 PMCID: PMC4298673 DOI: 10.1007/s00418-014-1273-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2014] [Indexed: 12/03/2022]
Abstract
The gelatinases MMP-9 and MMP-2 have been implicated in skeletal muscle adaptation to training; however, their specific role(s) in the different muscle types are only beginning to be unraveled. Recently, we found that treadmill running increased the activity and/or expression of these enzymes in myonuclei and in activated satellite cells of the soleus (Sol), but not extensor digitorum longus (EDL) muscles on the fifth day of training of adult rats. Here, we asked whether the gelatinases can be involved in physical exercise-induced adaptation of the neuromuscular compartment. To determine the subcellular localization of the gelatinolytic activity, we used high-resolution in situ zymography and immunofluorescence techniques. In both control and trained muscles, strong gelatinolytic activity was associated with myelin sheaths within intramuscular nerve twigs. In EDL, but not Sol, there was an increase in the gelatinolytic activity at the postsynaptic domain of the neuromuscular junction (NMJ). The increased activity was found within punctate structures situated in the vicinity of synaptic cleft of the NMJ, colocalizing with a marker of endoplasmic reticulum. Our results support the hypothesis that the gelatinolytic activity at the NMJ may be involved in NMJ plasticity.
Collapse
|
35
|
Dürnberger G, Camurdanoglu BZ, Tomschik M, Schutzbier M, Roitinger E, Hudecz O, Mechtler K, Herbst R. Global analysis of muscle-specific kinase signaling by quantitative phosphoproteomics. Mol Cell Proteomics 2014; 13:1993-2003. [PMID: 24899341 DOI: 10.1074/mcp.m113.036087] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The development of the neuromuscular synapse depends on signaling processes that involve protein phosphorylation as a crucial regulatory event. Muscle-specific kinase (MuSK) is the key signaling molecule at the neuromuscular synapse whose activity is required for the formation of a mature and functional synapse. However, the signaling cascade downstream of MuSK and the regulation of the different components are still poorly understood. In this study we used a quantitative phosphoproteomics approach to study the phosphorylation events and their temporal regulation downstream of MuSK. We identified a total of 10,183 phosphopeptides, of which 203 were significantly up- or down-regulated. Regulated phosphopeptides were classified into four different clusters according to their temporal profiles. Within these clusters we found an overrepresentation of specific protein classes associated with different cellular functions. In particular, we found an enrichment of regulated phosphoproteins involved in posttranscriptional mechanisms and in cytoskeletal organization. These findings provide novel insights into the complex signaling network downstream of MuSK and form the basis for future mechanistic studies.
Collapse
Affiliation(s)
- Gerhard Dürnberger
- From the ‡Gregor Mendel Institute of Molecular Plant Biology, Dr. Bohr-Gasse 3, 1030 Vienna, Austria; §Institute for Molecular Pathology (IMP), Dr. Bohr-Gasse 7, 1030 Vienna, Austria; ¶Institute of Molecular Biotechnology (IMBA), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Bahar Z Camurdanoglu
- ‖Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Matthias Tomschik
- ‖Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Michael Schutzbier
- From the ‡Gregor Mendel Institute of Molecular Plant Biology, Dr. Bohr-Gasse 3, 1030 Vienna, Austria; §Institute for Molecular Pathology (IMP), Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Elisabeth Roitinger
- §Institute for Molecular Pathology (IMP), Dr. Bohr-Gasse 7, 1030 Vienna, Austria; ¶Institute of Molecular Biotechnology (IMBA), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Otto Hudecz
- §Institute for Molecular Pathology (IMP), Dr. Bohr-Gasse 7, 1030 Vienna, Austria; ¶Institute of Molecular Biotechnology (IMBA), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Karl Mechtler
- §Institute for Molecular Pathology (IMP), Dr. Bohr-Gasse 7, 1030 Vienna, Austria; ¶Institute of Molecular Biotechnology (IMBA), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Ruth Herbst
- ‖Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria; ‡‡Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria
| |
Collapse
|