1
|
Luo J, Min Q, Sun X, Guo X, Song M, Zeng X, Guo J, Zhang H, Han Y, Li L. Comparative Whole-Genome Analysis of Production Traits and Genetic Structure in Baiyu and Chuanzhong Black Goats. Animals (Basel) 2024; 14:3616. [PMID: 39765520 PMCID: PMC11672699 DOI: 10.3390/ani14243616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Natural selection and artificial breeding are crucial methods for developing new animal groups. The Baiyu black goats and Chuanzhong black goats are indigenous goat breeds from distinct ecological regions in Sichuan Province, with dramatically different growth and reproductivity. This study aimed to systematically elucidate the differences in production performance and genetic traits between Baiyu black goats and Chuanzhong black goats. We quantified growth and reproductive attributes for both breeds. Furthermore, we conducted a comprehensive analysis of genetic diversity, population structure, and selection signatures using whole-genome resequencing data. This dataset included 30 individuals from the Baiyu black goat breed, 41 from the Chuanzhong black goat breed, and an additional 59 individuals representing Chengdu grey goats, Tibetan cashmere goats, and Jianchang black goats, totaling 130 individuals across five goat breeds. The comparative analysis of production performance revealed that the weight and body size of Chuanzhong black goats were significantly higher than those of Baiyu black goats (p < 0.01). At the same time, the average kidding rate and kid-weaning survival rate of Chuanzhong black goats were also notably superior to those of Baiyu black goats (p < 0.01). The Baiyu black goats exhibited a more abundant genetic diversity and distinct genetic differences compared to the Chuanzhong black goat, according to an analysis grounded on genomic variation. The Baiyu black goats are more closely related to Tibetan cashmere goats, whereas Chuanzhong black goats share a closer genetic relationship with Chengdu grey goats. Additionally, we employed the π, Fst, and XP-EHH methodologies to identify genes related to immunity (TRIM10, TRIM15, TRIM26, and TRIM5), neurodevelopment (FOXD4L1, PCDHB14, PCDHB4, PCDHB5, PCDHB6, and PCDHB7), reproduction (BTNL2 and GABBR1), body size (NCAPG, IBSP, and MKNK1), and meat quality traits (SUCLG2 and PGM5). These results provide a theoretical basis for further resource conservation and breeding improvement of the Baiyu black goat and Chuanzhong black goat.
Collapse
Affiliation(s)
- Jing Luo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Qi Min
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Xueliang Sun
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Xinyu Guo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Meijun Song
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Xuehui Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Jiazhong Guo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Hongping Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| | - Yanguo Han
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Li Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.L.); (Q.M.); (X.S.); (X.G.); (M.S.); (X.Z.); (J.G.); (H.Z.)
| |
Collapse
|
2
|
Kokot T, Zimmermann JP, Schwäble AN, Reimann L, Herr AL, Höfflin N, Köhn M, Warscheid B. Protein phosphatase-1 regulates the binding of filamin C to FILIP1 in cultured skeletal muscle cells under mechanical stress. Sci Rep 2024; 14:27348. [PMID: 39521905 PMCID: PMC11550807 DOI: 10.1038/s41598-024-78953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
The actin-binding protein filamin c (FLNc) is a key mediator in the response of skeletal muscle cells to mechanical stress. In addition to its function as a structural scaffold, FLNc acts as a signaling adaptor which is phosphorylated at S2234 in its mechanosensitive domain 20 (d20) through AKT. Here, we discovered a strong dephosphorylation of FLNc-pS2234 in cultured skeletal myotubes under acute mechanical stress, despite high AKT activity. We found that all three protein phosphatase 1 (PP1) isoforms are part of the FLNc d18-21 interactome. Enzymatic assays demonstrate that PP1 efficiently dephosphorylates FLNc-pS2234 and in vitro and in cells upon PP1 activation using specific modulators. FLNc-pS2234 dephosphorylation promotes the interaction with FILIP1, a mediator for filamin degradation. Altogether, we present a model in which dephosphorylation of FLNc d20 by the dominant action of PP1c prevails over AKT activity to promote the binding of the filamin degradation-inducing factor FILIP1 during acute mechanical stress.
Collapse
Affiliation(s)
- Thomas Kokot
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Johannes P Zimmermann
- Biochemistry II, Theodor-Boveri-Institut, Biozentrum, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany
| | - Anja N Schwäble
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Celonic AG, Basel, Switzerland
| | - Lena Reimann
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Celonic AG, Basel, Switzerland
| | - Anna L Herr
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Sartorius CellGenix GmbH, Freiburg, Germany
| | - Nico Höfflin
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Maja Köhn
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Bettina Warscheid
- Biochemistry II, Theodor-Boveri-Institut, Biozentrum, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany.
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Goliusova DV, Sharikova MY, Lavrenteva KA, Lebedeva OS, Muranova LK, Gusev NB, Bogomazova AN, Lagarkova MA. Role of Filamin C in Muscle Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1546-1557. [PMID: 39418514 DOI: 10.1134/s0006297924090025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/02/2024] [Accepted: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Filamin C (FLNC) is a member of a high-molecular weight protein family, which bind actin filaments in the cytoskeleton of various cells. In human genome FLNC is encoded by the FLNC gene located on chromosome 7 and is expressed predominantly in striated skeletal and cardiac muscle cells. Filamin C is involved in organization and stabilization of thin actin filaments three-dimensional network in sarcomeres, and is supposed to play a role of mechanosensor transferring mechanical signals to different protein targets. Under mechanical stress FLNC can undergo unfolding that increases the risk of its aggregation. FLNC molecules with an impaired native structure could be eliminated by the BAG3-mediated chaperone-assisted selective autophagy. Mutations in the FLNC gene could be accompanied by the changes in FLNC interaction with its protein partners and could lead to formation of aggregates, which overload the autophagy and proteasome protein degradation systems, thus facilitating development of various pathological processes. Molecular mechanisms of the FLNC-associated congenital disorders, called filaminopathies, remain poorly understood. This review is devoted to analysis of the structure and mechanisms of filamin C function in muscle and heart cells in normal state and in the FLNC-associated pathologies. The presented data summarize the results of research at the molecular, cellular, and tissue levels and allow us to outline promising ways for further investigation of pathogenetic mechanisms in filaminopathies.
Collapse
Affiliation(s)
- Daria V Goliusova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia.
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita Y Sharikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Kristina A Lavrenteva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Olga S Lebedeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Lidia K Muranova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nikolai B Gusev
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexandra N Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| |
Collapse
|
4
|
Hsu JE, Ruiz L, Hwang Y, Guzman S, Cho CS, Cheng W, Si Y, Macpherson P, Schrank M, Jun G, Kang HM, Kim M, Brooks S, Lee JH. High-Resolution Spatial Transcriptomic Atlas of Mouse Soleus Muscle: Unveiling Single Cell and Subcellular Heterogeneity in Health and Denervation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582103. [PMID: 38464282 PMCID: PMC10925160 DOI: 10.1101/2024.02.26.582103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Skeletal muscle is essential for both movement and metabolic processes, characterized by a complex and ordered structure. Despite its importance, a detailed spatial map of gene expression within muscle tissue has been challenging to achieve due to the limitations of existing technologies, which struggle to provide high-resolution views. In this study, we leverage the Seq-Scope technique, an innovative method that allows for the observation of the entire transcriptome at an unprecedented submicron spatial resolution. By applying this technique to the mouse soleus muscle, we analyze and compare the gene expression profiles in both healthy conditions and following denervation, a process that mimics aspects of muscle aging. Our approach reveals detailed characteristics of muscle fibers, other cell types present within the muscle, and specific subcellular structures such as the postsynaptic nuclei at neuromuscular junctions, hybrid muscle fibers, and areas of localized expression of genes responsive to muscle injury, along with their histological context. The findings of this research significantly enhance our understanding of the diversity within the muscle cell transcriptome and its variation in response to denervation, a key factor in the decline of muscle function with age. This breakthrough in spatial transcriptomics not only deepens our knowledge of muscle biology but also sets the stage for the development of new therapeutic strategies aimed at mitigating the effects of aging on muscle health, thereby offering a more comprehensive insight into the mechanisms of muscle maintenance and degeneration in the context of aging and disease.
Collapse
Affiliation(s)
- Jer-En Hsu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lloyd Ruiz
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Yongha Hwang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Space Planning and Analysis, University of Michigan, Ann Arbor, MI, USA
| | - Steve Guzman
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Weiqiu Cheng
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Yichen Si
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Peter Macpherson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Mitchell Schrank
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Goo Jun
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hyun-Min Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Susan Brooks
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Vasileva F, Font-Lladó R, Carreras-Badosa G, Roman-Viñas B, Cadellans-Arróniz A, López-Bermejo A, Prats-Puig A. Salivary cardiac-enriched FHL2-interacting protein is associated with higher diastolic-to-systolic-blood pressure ratio, sedentary time and center of pressure displacement in healthy 7-9 years old school-children. Front Endocrinol (Lausanne) 2024; 15:1292653. [PMID: 38304464 PMCID: PMC10830845 DOI: 10.3389/fendo.2024.1292653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
Introduction Cardiac-enriched FHL2-interacting protein (CEFIP) is a recently identified protein, first found in the z-disc of striated muscles, and related to cardiovascular diseases. Our objectives are: 1) to quantify CEFIP in saliva in healthy 7-9 years old school-children; and 2) to assess the associations of salivary CEFIP concentration and blood pressure, physical (in)activity and physical fitness in these children. Methods A total of 72 children (7.6 ± 0.3 years) were included in the study, recruited in primary schools in Girona (Spain). A sandwich enzyme-linked immunosorbent assay was used (abx506878; Abbexa, United Kingdom) to quantify CEFIP in saliva. Anthropometric evaluation was performed [body mass, height and body mass index (BMI)]. Systolic and diastolic blood pressure were measured by means of an electronic oscillometer and the diastolic-to-systolic blood pressure ratio (D/S BP ratio) was calculated. Physical (in)activity [sedentary time and time spent in physical activity (PA)] were assessed by means of a triaxial Actigraph GT3X accelerometer (Actigraph, Pensacola, FL, USA) that children were instructed to wear for 24h during 7 conssecutive days. Finally, physical fitness (speed and agility, explosive power of legs, handgrip strength, flexibility and balance) were assessed through validated and standardized testing batteries. Results CEFIP was easily detected and measured in all saliva samples (mean concentration: 0.6 ± 0.2 pg/ml). Salivary CEFIP was positively associated with D/S BP ratio (r=0.305, p=0.010) and sedentary time (r=0.317, p=0.012), but negatively associated with PA in 7-9 years old school-children (r=-0.350, p=0.002). Furthermore, salivary CEFIP was related to lower level of balance i.e., higher center of pressure (CoP) displacement in these children (r=0.411, p<0.001). The associations of salivary CEFIP with D/S BP ratio (Beta=0.349, p=0.004), sedentary time (Beta=0.354, p=0.009) and CoP displacement (Beta=0.401, p=0.001), were maintained significant after adjustment for potential confounding variables such as age, gender and BMI in linear regression analyses. Conclusion CEFIP can be easily assessed in saliva as a promising biomarker associated with cardiovascular health in 7-9 years old school-children. Interestingly, higher salivary CEFIP concentration was related to higher D/S BP ratio, more sedentary time and higher CoP displacement i.e., lower level of balance in these children.
Collapse
Affiliation(s)
- Fidanka Vasileva
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research, Girona, Spain
- University School of Health and Sport, University of Girona, Girona, Spain
| | - Raquel Font-Lladó
- University School of Health and Sport, University of Girona, Girona, Spain
- Research Group of Culture and Education, Institute of Educational Research, University of Girona, Girona, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research, Girona, Spain
| | - Blanca Roman-Viñas
- University School of Health and Sport, University of Girona, Girona, Spain
- Department of Physical Activity and Sport Sciences, Blanquerna-Universitat Ramon Llull, Barcelona, Spain
| | - Aïda Cadellans-Arróniz
- Department of Physiotherapy, Faculty of Medicine and Health Sciences, International University of Catalonia, Barcelona, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research, Girona, Spain
- Department of Medical Sciences, University of Girona, Girona, Spain
- Pediatric Endocrinology, Dr. Josep Trueta Hospital, Girona, Spain
| | - Anna Prats-Puig
- University School of Health and Sport, University of Girona, Girona, Spain
- Research Group of Clinical Anatomy, Embryology and Neuroscience, Department of Medical Sciences, University of Girona, Girona, Spain
| |
Collapse
|
6
|
Quintana‐Torres D, Valle‐Cao A, Bousquets‐Muñoz P, Freitas‐Rodríguez S, Rodríguez F, Lucia A, López‐Otín C, López‐Soto A, Folgueras AR. The secretome atlas of two mouse models of progeria. Aging Cell 2023; 22:e13952. [PMID: 37565451 PMCID: PMC10577534 DOI: 10.1111/acel.13952] [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: 06/06/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease caused by nuclear envelope alterations that lead to accelerated aging and premature death. Several studies have linked health and longevity to cell-extrinsic mechanisms, highlighting the relevance of circulating factors in the aging process as well as in age-related diseases. We performed a global plasma proteomic analysis in two preclinical progeroid models (LmnaG609G/G609G and Zmpste24-/- mice) using aptamer-based proteomic technology. Pathways related to the extracellular matrix, growth factor response and calcium ion binding were among the most enriched in the proteomic signature of progeroid samples compared to controls. Despite the global downregulation trend found in the plasma proteome of progeroid mice, several proteins associated with cardiovascular disease, the main cause of death in HGPS, were upregulated. We also developed a chronological age predictor using plasma proteome data from a cohort of healthy mice (aged 1-30 months), that reported an age acceleration when applied to progeroid mice, indicating that these mice exhibit an "old" plasma proteomic signature. Furthermore, when compared to naturally-aged mice, a great proportion of differentially expressed circulating proteins in progeroid mice were specific to premature aging, highlighting secretome-associated differences between physiological and accelerated aging. This is the first large-scale profiling of the plasma proteome in progeroid mice, which provides an extensive list of candidate circulating plasma proteins as potential biomarkers and/or therapeutic targets for further exploration and hypothesis generation in the context of both physiological and premature aging.
Collapse
Affiliation(s)
- Diego Quintana‐Torres
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Alejandra Valle‐Cao
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Pablo Bousquets‐Muñoz
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Sandra Freitas‐Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Francisco Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Alejandro Lucia
- CIBER of Frailty and Healthy Aging (CIBERFES) and Instituto de Investigación 12 de Octubre (i+12)MadridSpain
- Faculty of Sport SciencesUniversidad EuropeaMadridSpain
| | - Carlos López‐Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
| | - Alejandro López‐Soto
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Alicia R. Folgueras
- Departamento de Bioquímica y Biología Molecular, Facultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de OviedoOviedoSpain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| |
Collapse
|
7
|
Deng Y, Yan J. Force-Dependent Structural Changes of Filamin C Rod Domains Regulated by Filamin C Dimer. J Am Chem Soc 2023; 145:14670-14678. [PMID: 37369984 PMCID: PMC10348313 DOI: 10.1021/jacs.3c02303] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Indexed: 06/29/2023]
Abstract
Filamin C (FLNC), a large dimeric actin-binding protein in muscle cells, plays a critical role in transmitting force in the cytoskeleton and that between membrane receptors and the cytoskeleton. It performs crucial mechanosensing and downstream mechanotransduction functions via force-dependent interactions with signaling proteins. Mutations in FLNC have been linked to muscle and heart diseases. The mechanical responses of the force-bearing elements in FLNC have not been determined. This study investigated the mechanical responses of FLNC domains and their dimerization interface using magnetic tweezers. Results showed high stability of the N-terminal domains in the rod-1 segment but significant changes in the rod-2 domains in response to forces of a few piconewtons (pN). The dimerization interface, formed by the R24 domain, has a lifetime of seconds to tens of seconds at pN forces, and it dissociates within 1 s at forces greater than 14 pN. The findings suggest the FLNC dimerization interface provides sufficient mechanical stability that enables force-dependent structural changes in rod-2 domains for signaling protein binding and maintains structural integrity of the rod-1 domains.
Collapse
Affiliation(s)
- Yunxin Deng
- Mechanobiology
Institute, National University of Singapore, Singapore 117411, Singapore
| | - Jie Yan
- Mechanobiology
Institute, National University of Singapore, Singapore 117411, Singapore
- Department
of Physics, National University of Singapore, Singapore 117542, Singapore
- Joint
School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
| |
Collapse
|
8
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Krause K, Eggers B, Uszkoreit J, Eulitz S, Rehmann R, Güttsches AK, Schreiner A, van der Ven PFM, Fürst DO, Marcus K, Vorgerd M, Kley RA. Target formation in muscle fibres indicates reinnervation - A proteomic study in muscle samples from peripheral neuropathies. Neuropathol Appl Neurobiol 2023; 49:e12853. [PMID: 36180966 DOI: 10.1111/nan.12853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/20/2022] [Accepted: 07/23/2022] [Indexed: 11/30/2022]
Abstract
AIMS Target skeletal muscle fibres - defined by different concentric areas in oxidative enzyme staining - can occur in patients with neurogenic muscular atrophy. Here, we used our established hypothesis-free proteomic approach with the aim of deciphering the protein composition of targets. We also searched for potential novel interactions between target proteins. METHODS Targets and control areas were laser microdissected from skeletal muscle sections of 20 patients with neurogenic muscular atrophy. Samples were analysed by a highly sensitive mass spectrometry approach, enabling relative protein quantification. The results were validated by immunofluorescence studies. Protein interactions were investigated by yeast two-hybrid assays, coimmunoprecipitation experiments and bimolecular fluorescence complementation. RESULTS More than 1000 proteins were identified. Among these, 55 proteins were significantly over-represented and 40 proteins were significantly under-represented in targets compared to intraindividual control samples. The majority of over-represented proteins were associated with the myofibrillar Z-disc and actin dynamics, followed by myosin and myosin-associated proteins, proteins involved in protein biosynthesis and chaperones. Under-represented proteins were mainly mitochondrial proteins. Functional studies revealed that the LIM domain of the over-represented protein LIMCH1 interacts with isoform A of Xin actin-binding repeat-containing protein 1 (XinA). CONCLUSIONS In particular, proteins involved in myofibrillogenesis are over-represented in target structures, which indicate an ongoing process of sarcomere assembly and/or remodelling within this specific area of the muscle fibres. We speculate that target structures are the result of reinnervation processes in which filamin C-associated myofibrillogenesis is tightly regulated by the BAG3-associated protein quality system.
Collapse
Affiliation(s)
- Karsten Krause
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Britta Eggers
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Julian Uszkoreit
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Stefan Eulitz
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Robert Rehmann
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Anne K Güttsches
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Anja Schreiner
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | | | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Rudolf A Kley
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany.,Department of Neurology and Clinical Neurophysiology, St. Marien-Hospital Borken, Borken, Germany
| |
Collapse
|
10
|
Desmin Knock-Out Cardiomyopathy: A Heart on the Verge of Metabolic Crisis. Int J Mol Sci 2022; 23:ijms231912020. [PMID: 36233322 PMCID: PMC9570457 DOI: 10.3390/ijms231912020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 12/05/2022] Open
Abstract
Desmin mutations cause familial and sporadic cardiomyopathies. In addition to perturbing the contractile apparatus, both desmin deficiency and mutated desmin negatively impact mitochondria. Impaired myocardial metabolism secondary to mitochondrial defects could conceivably exacerbate cardiac contractile dysfunction. We performed metabolic myocardial phenotyping in left ventricular cardiac muscle tissue in desmin knock-out mice. Our analyses revealed decreased mitochondrial number, ultrastructural mitochondrial defects, and impaired mitochondria-related metabolic pathways including fatty acid transport, activation, and catabolism. Glucose transporter 1 and hexokinase-1 expression and hexokinase activity were increased. While mitochondrial creatine kinase expression was reduced, fetal creatine kinase expression was increased. Proteomic analysis revealed reduced expression of proteins involved in electron transport mainly of complexes I and II, oxidative phosphorylation, citrate cycle, beta-oxidation including auxiliary pathways, amino acid catabolism, and redox reactions and oxidative stress. Thus, desmin deficiency elicits a secondary cardiac mitochondriopathy with severely impaired oxidative phosphorylation and fatty and amino acid metabolism. Increased glucose utilization and fetal creatine kinase upregulation likely portray attempts to maintain myocardial energy supply. It may be prudent to avoid medications worsening mitochondrial function and other metabolic stressors. Therapeutic interventions for mitochondriopathies might also improve the metabolic condition in desmin deficient hearts.
Collapse
|
11
|
Cardiovascular Involvement in Pediatric FLNC Variants: A Case Series of Fourteen Patients. J Cardiovasc Dev Dis 2022; 9:jcdd9100332. [PMID: 36286284 PMCID: PMC9604120 DOI: 10.3390/jcdd9100332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Filamin C is a protein specifically expressed in myocytes and cardiomyocytes and is involved in several biological functions, including sarcomere contractile activity, signaling, cellular adhesion, and repair. FLNC variants are associated with different disorders ranging from striated muscle (myofibrillar distal or proximal) myopathy to cardiomyopathies (CMPs) (restrictive, hypertrophic, and dilated), or both. The outcome depends on functional consequences of the detected variants, which result either in FLNC haploinsufficiency or in an aberrant protein, the latter affecting sarcomere structure leading to protein aggregates. Cardiac manifestations of filaminopathies are most often described as adult onset CMPs and limited reports are available in children or on other cardiac spectrums (congenital heart defects-CHDs, or arrhythmias). Here we report on 13 variants in 14 children (2.8%) out of 500 pediatric patients with early-onset different cardiac features ranging from CMP to arrhythmias and CHDs. In one patient, we identified a deletion encompassing FLNC detected by microarray, which was overlooked by next generation sequencing. We established a potential genotype-phenotype correlation of the p.Ala1186Val variant in severe and early-onset restrictive cardiomyopathy (RCM) associated with a limb-girdle defect (two new patients in addition to the five reported in the literature). Moreover, in three patients (21%), we identified a relatively frequent finding of long QT syndrome (LQTS) associated with RCM (n = 2) and a hypertrabeculated left ventricle (n = 1). RCM and LQTS in children might represent a specific red flag for FLNC variants. Further studies are warranted in pediatric cohorts to delineate potential expanding phenotypes related to FLNC.
Collapse
|
12
|
Sun J, Wang F, Zhou H, Zhao C, Li K, Fan C, Wang J. Downregulation of PGM5 expression correlates with tumor progression and poor prognosis in human prostate cancer. Discov Oncol 2022; 13:63. [PMID: 35819729 PMCID: PMC9276915 DOI: 10.1007/s12672-022-00525-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/15/2022] [Indexed: 11/27/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy in men in developed countries. Prostate-specific antigen (PSA) remains the most widely used serum marker for prostate cancer. Here, we reported that the expression of phosphoglucomutase-like protein 5 (PGM5) is significantly lower in prostate cancer tissue. The low expression of PGM5 and its related gene signature were found to be linked to poor clinical outcome and high Gleason score. In vitro assays showed that overexpression of PGM5 significantly repressed proliferation and migration of prostate cancer cells. GO and pathway analyses showed the enrichment of genes in regulation of cell growth and migration, and pathways related in cancer. Our additional results showed that the downregulation of PGM5 is closely related to DNA methylation. Taken together, our findings provide the first evidence that PGM5 expression is associated with prostate cancer progression. These results also highlight a preclinical rationale that PGM5 represents a prognostic marker and a promising target for new therapeutic strategies in prostate cancer.
Collapse
Affiliation(s)
- Jian Sun
- Department of Urology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 26 Daoqian Rd, Suzhou, Jiangsu, 215000, PR China
| | - Fei Wang
- Department of Urology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 26 Daoqian Rd, Suzhou, Jiangsu, 215000, PR China
| | - Huihui Zhou
- Department of Pathology, Affiliated Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Chunchun Zhao
- Department of Urology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 26 Daoqian Rd, Suzhou, Jiangsu, 215000, PR China
| | - Kai Li
- Department of Urology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 26 Daoqian Rd, Suzhou, Jiangsu, 215000, PR China
| | - Caibin Fan
- Department of Urology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 26 Daoqian Rd, Suzhou, Jiangsu, 215000, PR China.
| | - Jianqing Wang
- Department of Urology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 26 Daoqian Rd, Suzhou, Jiangsu, 215000, PR China.
| |
Collapse
|
13
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
14
|
Murgia M, Nogara L, Baraldo M, Reggiani C, Mann M, Schiaffino S. Protein profile of fiber types in human skeletal muscle: a single-fiber proteomics study. Skelet Muscle 2021; 11:24. [PMID: 34727990 PMCID: PMC8561870 DOI: 10.1186/s13395-021-00279-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/19/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Human skeletal muscle is composed of three major fiber types, referred to as type 1, 2A, and 2X fibers. This heterogeneous cellular composition complicates the interpretation of studies based on whole skeletal muscle lysate. A single-fiber proteomics approach is required to obtain a fiber-type resolved quantitative information on skeletal muscle pathophysiology. METHODS Single fibers were dissected from vastus lateralis muscle biopsies of young adult males and processed for mass spectrometry-based single-fiber proteomics. We provide and analyze a resource dataset based on relatively pure fibers, containing at least 80% of either MYH7 (marker of slow type 1 fibers), MYH2 (marker of fast 2A fibers), or MYH1 (marker of fast 2X fibers). RESULTS In a dataset of more than 3800 proteins detected by single-fiber proteomics, we selected 404 proteins showing a statistically significant difference among fiber types. We identified numerous type 1 or 2X fiber type-specific protein markers, defined as proteins present at 3-fold or higher levels in these compared to other fiber types. In contrast, we could detect only two 2A-specific protein markers in addition to MYH2. We observed three other major patterns: proteins showing a differential distribution according to the sequence 1 > 2A > 2X or 2X > 2A > 1 and type 2-specific proteins expressed in 2A and 2X fibers at levels 3 times greater than in type 1 fibers. In addition to precisely quantifying known fiber type-specific protein patterns, our study revealed several novel features of fiber type specificity, including the selective enrichment of components of the dystrophin and integrin complexes, as well as microtubular proteins, in type 2X fibers. The fiber type-specific distribution of some selected proteins revealed by proteomics was validated by immunofluorescence analyses with specific antibodies. CONCLUSION We here show that numerous muscle proteins, including proteins whose function is unknown, are selectively enriched in specific fiber types, pointing to potential implications in muscle pathophysiology. This reinforces the notion that single-fiber proteomics, together with recently developed approaches to single-cell proteomics, will be instrumental to explore and quantify muscle cell heterogeneity.
Collapse
Affiliation(s)
- Marta Murgia
- Department of Biomedical Science, University of Padova, 35121, Padova, Italy.
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany.
| | - Leonardo Nogara
- Department of Biomedical Science, University of Padova, 35121, Padova, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35121, Padova, Italy
| | - Martina Baraldo
- Department of Biomedical Science, University of Padova, 35121, Padova, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35121, Padova, Italy
| | - Carlo Reggiani
- Department of Biomedical Science, University of Padova, 35121, Padova, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, 6000, Koper, Slovenia
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
15
|
Abstract
Cardiomyopathy affects approximately 1 in 500 adults and is the leading cause of death. Familial cases are common, and mutations in many genes are involved in cardiomyopathy, especially those in genes encoding cytoskeletal, sarcomere, and nuclear envelope proteins. Filamin C is an actin-binding protein encoded by filamin C (FLNC) gene and participates in sarcomere stability maintenance. FLNC was first demonstrated to be a causal gene of myofibrillar myopathy; recently, it has been found that FLNC mutation plays a critical role in the pathogenesis of cardiomyopathy. In this review, we summarized the physiological roles of filamin C in cardiomyocytes and the genetic evidence for links between FLNC mutations and cardiomyopathies. Truncated FLNC is enriched in dilated cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy. Non-truncated FLNC is enriched in hypertrophic cardiomyopathy and restrictive cardiomyopathy. Two major pathomechanisms in FLNC-related cardiomyopathy have been described: protein aggregation resulting from non-truncating mutations and haploinsufficiency triggered by filamin C truncation. Therefore, it is important to understand the cellular biology and molecular regulation of FLNC to design new therapies to treat patients with FLNC-related cardiomyopathy.
Collapse
|
16
|
Li B, Guo Y, Zhan Y, Zhou X, Li Y, Zhao C, Sun N, Xu C, Liang Q. Cardiac Overexpression of XIN Prevents Dilated Cardiomyopathy Caused by TNNT2 ΔK210 Mutation. Front Cell Dev Biol 2021; 9:691749. [PMID: 34222259 PMCID: PMC8247596 DOI: 10.3389/fcell.2021.691749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
TNNT2 mutation is associated with a range of cardiac diseases, including dilated cardiomyopathy (DCM). However, the mechanisms underlying the development of DCM and heart failure remain incompletely understood. In the present study, we found the expression of cardiac XIN protein was reduced in TNNT2-ΔK210 hESCs-derived cardiomyocytes and mouse heart tissues. We further investigated whether XIN protects against TNNT2 mutation-induced DCM. Overexpression of the repeat-containing isoform XINB decreased the percentage of myofilaments disorganization and increased cell contractility of TNNT2-ΔK210 cardiomyocytes. Moreover, overexpression of XINB by heart-specific delivery via AAV9 ameliorates DCM remodeling caused by TNNT2-ΔK210 mutation in mice, revealed by partially reversed cardiac dilation, systolic dysfunction and heart fibrosis. These results suggest that deficiency of XIN may play a critical role in the development of DCM. Consequently, our findings may provide a new mechanistic insight and represent a therapeutic target for the treatment of idiopathic DCM.
Collapse
Affiliation(s)
- Bin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongkun Zhan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xinyan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongbo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chao Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qianqian Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Kley RA, Leber Y, Schrank B, Zhuge H, Orfanos Z, Kostan J, Onipe A, Sellung D, Güttsches AK, Eggers B, Jacobsen F, Kress W, Marcus K, Djinovic-Carugo K, van der Ven PFM, Fürst DO, Vorgerd M. FLNC-Associated Myofibrillar Myopathy: New Clinical, Functional, and Proteomic Data. NEUROLOGY-GENETICS 2021; 7:e590. [PMID: 34235269 PMCID: PMC8237399 DOI: 10.1212/nxg.0000000000000590] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/28/2020] [Indexed: 11/15/2022]
Abstract
Objective To determine whether a new indel mutation in the dimerization domain of filamin C (FLNc) causes a hereditary myopathy with protein aggregation in muscle fibers, we clinically and molecularly studied a German family with autosomal dominant myofibrillar myopathy (MFM). Methods We performed mutational analysis in 3 generations, muscle histopathology, and proteomic studies of IM protein aggregates. Functional consequences of the FLNC mutation were investigated with interaction and transfection studies and biophysics molecular analysis. Results Eight patients revealed clinical features of slowly progressive proximal weakness associated with a heterozygous c.8025_8030delCAAGACinsA (p.K2676Pfs*3) mutation in FLNC. Two patients exhibited a mild cardiomyopathy. MRI of skeletal muscle revealed lipomatous changes typical for MFM with FLNC mutations. Muscle biopsies showed characteristic MFM findings with protein aggregation and lesion formation. The proteomic profile of aggregates was specific for MFM-filaminopathy and indicated activation of the ubiquitin-proteasome system (UPS) and autophagic pathways. Functional studies revealed that mutant FLNc is misfolded, unstable, and incapable of forming homodimers and heterodimers with wild-type FLNc. Conclusions This new MFM-filaminopathy family confirms that expression of mutant FLNC leads to an adult-onset muscle phenotype with intracellular protein accumulation. Mutant FLNc protein is biochemically compromised and leads to dysregulation of protein quality control mechanisms. Proteomic analysis of MFM protein aggregates is a potent method to identify disease-relevant proteins, differentiate MFM subtypes, evaluate the relevance of gene variants, and identify novel MFM candidate genes.
Collapse
Affiliation(s)
- Rudolf Andre Kley
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Yvonne Leber
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Bertold Schrank
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Heidi Zhuge
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Zacharias Orfanos
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Julius Kostan
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Adekunle Onipe
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Dominik Sellung
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Anne Katrin Güttsches
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Britta Eggers
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Frank Jacobsen
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Wolfram Kress
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Katrin Marcus
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Kristina Djinovic-Carugo
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Peter F M van der Ven
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Dieter O Fürst
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Matthias Vorgerd
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
18
|
Accurate contact-based modelling of repeat proteins predicts the structure of new repeats protein families. PLoS Comput Biol 2021; 17:e1008798. [PMID: 33857128 PMCID: PMC8078820 DOI: 10.1371/journal.pcbi.1008798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 04/27/2021] [Accepted: 02/15/2021] [Indexed: 12/18/2022] Open
Abstract
Repeat proteins are abundant in eukaryotic proteomes. They are involved in many eukaryotic specific functions, including signalling. For many of these proteins, the structure is not known, as they are difficult to crystallise. Today, using direct coupling analysis and deep learning it is often possible to predict a protein’s structure. However, the unique sequence features present in repeat proteins have been a challenge to use direct coupling analysis for predicting contacts. Here, we show that deep learning-based methods (trRosetta, DeepMetaPsicov (DMP) and PconsC4) overcomes this problem and can predict intra- and inter-unit contacts in repeat proteins. In a benchmark dataset of 815 repeat proteins, about 90% can be correctly modelled. Further, among 48 PFAM families lacking a protein structure, we produce models of forty-one families with estimated high accuracy. Repeat proteins are widespread among organisms and particularly abundant in eukaryotic proteomes. Their primary sequence presents repetition in the amino acid sequences that origin structures with repeated folds/domains. Although the repeated units often can be recognised from the sequence alone, often structural information is missing. Here, we used contact prediction for predicting the structure of repeats protein directly from their primary sequences. We benchmark the methods on a dataset comprehensive of all the known repeated structures. We evaluate the contact predictions and the obtained models for different classes of repeat proteins. Further, we develop and benchmark a quality assessment (QA) method specific for repeat proteins. Finally, we used the prediction pipeline for all PFAM repeat families without resolved structures and found that forty-one of them could be modelled with high accuracy.
Collapse
|
19
|
The Role of Z-disc Proteins in Myopathy and Cardiomyopathy. Int J Mol Sci 2021; 22:ijms22063058. [PMID: 33802723 PMCID: PMC8002584 DOI: 10.3390/ijms22063058] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The Z-disc acts as a protein-rich structure to tether thin filament in the contractile units, the sarcomeres, of striated muscle cells. Proteins found in the Z-disc are integral for maintaining the architecture of the sarcomere. They also enable it to function as a (bio-mechanical) signalling hub. Numerous proteins interact in the Z-disc to facilitate force transduction and intracellular signalling in both cardiac and skeletal muscle. This review will focus on six key Z-disc proteins: α-actinin 2, filamin C, myopalladin, myotilin, telethonin and Z-disc alternatively spliced PDZ-motif (ZASP), which have all been linked to myopathies and cardiomyopathies. We will summarise pathogenic variants identified in the six genes coding for these proteins and look at their involvement in myopathy and cardiomyopathy. Listing the Minor Allele Frequency (MAF) of these variants in the Genome Aggregation Database (GnomAD) version 3.1 will help to critically re-evaluate pathogenicity based on variant frequency in normal population cohorts.
Collapse
|
20
|
Kim M, Franke V, Brandt B, Lowenstein ED, Schöwel V, Spuler S, Akalin A, Birchmeier C. Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells. Nat Commun 2020; 11:6375. [PMID: 33311457 PMCID: PMC7732842 DOI: 10.1038/s41467-020-20064-9] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [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: 11/25/2022] Open
Abstract
Syncytial skeletal muscle cells contain hundreds of nuclei in a shared cytoplasm. We investigated nuclear heterogeneity and transcriptional dynamics in the uninjured and regenerating muscle using single-nucleus RNA-sequencing (snRNAseq) of isolated nuclei from muscle fibers. This revealed distinct nuclear subtypes unrelated to fiber type diversity, previously unknown subtypes as well as the expected ones at the neuromuscular and myotendinous junctions. In fibers of the Mdx dystrophy mouse model, distinct subtypes emerged, among them nuclei expressing a repair signature that were also abundant in the muscle of dystrophy patients, and a nuclear population associated with necrotic fibers. Finally, modifications of our approach revealed the compartmentalization in the rare and specialized muscle spindle. Our data identifies nuclear compartments of the myofiber and defines a molecular roadmap for their functional analyses; the data can be freely explored on the MyoExplorer server ( https://shiny.mdc-berlin.de/MyoExplorer/ ).
Collapse
Affiliation(s)
- Minchul Kim
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Vedran Franke
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Bettina Brandt
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Elijah D Lowenstein
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Verena Schöwel
- Muscle Research Unit, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Altuna Akalin
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
21
|
Gustafsson R, Eckhard U, Ye W, Enbody ED, Pettersson M, Jemth P, Andersson L, Selmer M. Structure and Characterization of Phosphoglucomutase 5 from Atlantic and Baltic Herring-An Inactive Enzyme with Intact Substrate Binding. Biomolecules 2020; 10:E1631. [PMID: 33287293 PMCID: PMC7761743 DOI: 10.3390/biom10121631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/31/2022] Open
Abstract
Phosphoglucomutase 5 (PGM5) in humans is known as a structural muscle protein without enzymatic activity, but detailed understanding of its function is lacking. PGM5 belongs to the alpha-D-phosphohexomutase family and is closely related to the enzymatically active metabolic enzyme PGM1. In the Atlantic herring, Clupea harengus, PGM5 is one of the genes strongly associated with ecological adaptation to the brackish Baltic Sea. We here present the first crystal structures of PGM5, from the Atlantic and Baltic herring, differing by a single substitution Ala330Val. The structure of PGM5 is overall highly similar to structures of PGM1. The structure of the Baltic herring PGM5 in complex with the substrate glucose-1-phosphate shows conserved substrate binding and active site compared to human PGM1, but both PGM5 variants lack phosphoglucomutase activity under the tested conditions. Structure comparison and sequence analysis of PGM5 and PGM1 from fish and mammals suggest that the lacking enzymatic activity of PGM5 is related to differences in active-site loops that are important for flipping of the reaction intermediate. The Ala330Val substitution does not alter structure or biophysical properties of PGM5 but, due to its surface-exposed location, could affect interactions with protein-binding partners.
Collapse
Affiliation(s)
- Robert Gustafsson
- Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden; (R.G.); (U.E.)
| | - Ulrich Eckhard
- Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden; (R.G.); (U.E.)
| | - Weihua Ye
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE-751 23 Uppsala, Sweden; (W.Y.); (E.D.E.); (M.P.); (P.J.); (L.A.)
| | - Erik D. Enbody
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE-751 23 Uppsala, Sweden; (W.Y.); (E.D.E.); (M.P.); (P.J.); (L.A.)
| | - Mats Pettersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE-751 23 Uppsala, Sweden; (W.Y.); (E.D.E.); (M.P.); (P.J.); (L.A.)
| | - Per Jemth
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE-751 23 Uppsala, Sweden; (W.Y.); (E.D.E.); (M.P.); (P.J.); (L.A.)
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE-751 23 Uppsala, Sweden; (W.Y.); (E.D.E.); (M.P.); (P.J.); (L.A.)
- Department of Veterinary Integrative Biosciences, Texas A & M University, College Station, TX 77843, USA
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden
| | - Maria Selmer
- Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden; (R.G.); (U.E.)
| |
Collapse
|
22
|
Wang J, Fan Y, Dube S, Agassy NW, Dube DK, Sanger JM, Sanger JW. Myofibril assembly and the roles of the ubiquitin proteasome system. Cytoskeleton (Hoboken) 2020; 77:456-479. [PMID: 33124174 DOI: 10.1002/cm.21641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/07/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
De novo assembly of myofibrils in vertebrate cross-striated muscles progresses in three distinct steps, first from a minisarcomeric alignment of several nonmuscle and muscle proteins in premyofibrils, followed by insertions of additional proteins and increased organization in nascent myofibrils, ending with mature contractile myofibrils. In a search for controls of the process of myofibril assembly, we discovered that the transition from nascent to mature myofibrils could be halted by inhibitors of three distinct functions of the ubiquitin proteasome system (UPS). First, inhibition of pathway to E3 Cullin ligases that ubiquitinate proteins led to an arrest of myofibrillogenesis at the nascent myofibril stage. Second, inhibition of p97 protein extractions of ubiquitinated proteins led to a similar arrest of myofibrillogenesis at the nascent myofibril stage. Third, inhibitors of proteolytic action by proteasomes also blocked nascent myofibrils from transitioning to mature myofibrils. In contrast, inhibitors of autophagy or lysosomes did not affect myofibrillogenesis. To probe for differences in the effects of UPS inhibitors during myofibrillogenesis, we analyzed by fluorescence recovery after photobleaching the exchange rates of two selected sarcomeric proteins (muscle myosin II heavy chains and light chains). In the presence of p97 and proteasomal inhibitors, the dynamics of each of these two myosin proteins decreased in the nascent myofibril stage, but were unaffected in the mature myofibril stage. The increased stability of myofibrils occurring in the transition from nascent to mature myofibril assembly indicates the importance of dynamics and selective destruction in the muscle myosin II proteins for the remodeling of nascent to mature myofibrils.
Collapse
Affiliation(s)
- Jushuo Wang
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Yingli Fan
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Syamalima Dube
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Nicodeme Wanko Agassy
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Dipak K Dube
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jean M Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Joseph W Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
23
|
Diofano F, Weinmann K, Schneider I, Thiessen KD, Rottbauer W, Just S. Genetic compensation prevents myopathy and heart failure in an in vivo model of Bag3 deficiency. PLoS Genet 2020; 16:e1009088. [PMID: 33137814 PMCID: PMC7605898 DOI: 10.1371/journal.pgen.1009088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022] Open
Abstract
Mutations in the molecular co-chaperone Bcl2-associated athanogene 3 (BAG3) are found to cause dilated cardiomyopathy (DCM), resulting in systolic dysfunction and heart failure, as well as myofibrillar myopathy (MFM), which is characterized by protein aggregation and myofibrillar disintegration in skeletal muscle cells. Here, we generated a CRISPR/Cas9-induced Bag3 knockout zebrafish line and found the complete preservation of heart and skeletal muscle structure and function during embryonic development, in contrast to morpholino-mediated knockdown of Bag3. Intriguingly, genetic compensation, a process of transcriptional adaptation which acts independent of protein feedback loops, was found to prevent heart and skeletal muscle damage in our Bag3 knockout model. Proteomic profiling and quantitative real-time PCR analyses identified Bag2, another member of the Bag protein family, significantly upregulated on a transcript and protein level in bag3-/- mutants. This implied that the decay of bag3 mutant mRNA in homozygous bag3-/- embryos caused the transcriptional upregulation of bag2 expression. We further demonstrated that morpholino-mediated knockdown of Bag2 in bag3-/- embryos evoked severe functional and structural heart and skeletal muscle defects, which are similar to Bag3 morphants. However, Bag2 knockdown in bag3+/+ or bag3+/- embryos did not result in (cardio-)myopathy. Finally, we found that inhibition of the nonsense-mediated mRNA decay (NMD) machinery by knockdown of upf1, an essential NMD factor, caused severe heart and skeletal muscle defects in bag3-/- mutants due to the blockade of transcriptional adaptation of bag2 expression. Our findings provide evidence that genetic compensation might vitally influence the penetrance of disease-causing bag3 mutations in vivo. One form of genetic compensation is described as transcriptional adaptation of gene expression triggered by deleterious gene mutations. Although the precise molecular mechanism that induces genetic compensation needs to be defined, it represents a powerful biological phenomenon that warrants genetic robustness. We find that antisense-mediated knockdown of Bag3 in zebrafish embryos causes heart failure and myopathy. By contrast, CRISPR/Cas9-induced depletion of Bag3 does not result in the abrogation of heart and skeletal muscle function in zebrafish embryos. We find here that transcriptional activation of the Bag family member bag2 is capable of restoring heart and skeletal muscle function in bag3 mutant embryos, whereas this compensatory mechanism is not present in the bag3 morphants. Furthermore, we show that nonsense-mediated decay of bag3 mRNA is the molecular trigger for the compensatory upregulation of bag2. Our study provides evidence that genetic compensation via transcriptional adaptation is a vital modulator of disease peculiarity and penetrance in bag3 mutant zebrafish and that this biological phenomenon might also be active in certain human BAG3 mutation carriers.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis/genetics
- Apoptosis Regulatory Proteins/deficiency
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Cardiomyopathies/genetics
- Cardiomyopathies/metabolism
- Cardiomyopathies/pathology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Disease Models, Animal
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Molecular Chaperones/genetics
- Molecular Chaperones/metabolism
- Muscle Fibers, Skeletal/metabolism
- Muscular Diseases/genetics
- Muscular Diseases/metabolism
- Muscular Diseases/pathology
- Mutation
- Myocardium/metabolism
- Myopathies, Structural, Congenital/metabolism
- Phenotype
- Proteomics
- Zebrafish
- Zebrafish Proteins/deficiency
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Federica Diofano
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Karolina Weinmann
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
- Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Isabelle Schneider
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Kevin D. Thiessen
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
24
|
Schuld J, Orfanos Z, Chevessier F, Eggers B, Heil L, Uszkoreit J, Unger A, Kirfel G, van der Ven PFM, Marcus K, Linke WA, Clemen CS, Schröder R, Fürst DO. Homozygous expression of the myofibrillar myopathy-associated p.W2710X filamin C variant reveals major pathomechanisms of sarcomeric lesion formation. Acta Neuropathol Commun 2020; 8:154. [PMID: 32887649 PMCID: PMC7650280 DOI: 10.1186/s40478-020-01001-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/22/2020] [Indexed: 01/06/2023] Open
Abstract
Filamin C (FLNc) is mainly expressed in striated muscle cells where it localizes to Z-discs, myotendinous junctions and intercalated discs. Recent studies have revealed numerous mutations in the FLNC gene causing familial and sporadic myopathies and cardiomyopathies with marked clinical variability. The most frequent myopathic mutation, p.W2710X, which is associated with myofibrillar myopathy, deletes the carboxy-terminal 16 amino acids from FLNc and abolishes the dimerization property of Ig-like domain 24. We previously characterized "knock-in" mice heterozygous for this mutation (p.W2711X), and have now investigated homozygous mice using protein and mRNA expression analyses, mass spectrometry, and extensive immunolocalization and ultrastructural studies. Although the latter mice display a relatively mild myopathy under normal conditions, our analyses identified major mechanisms causing the pathophysiology of this disease: in comparison to wildtype animals (i) the expression level of FLNc protein is drastically reduced; (ii) mutant FLNc is relocalized from Z-discs to particularly mechanically strained parts of muscle cells, i.e. myotendinous junctions and myofibrillar lesions; (iii) the number of lesions is greatly increased and these lesions lack Bcl2-associated athanogene 3 (BAG3) protein; (iv) the expression of heat shock protein beta-7 (HSPB7) is almost completely abolished. These findings indicate grave disturbances of BAG3-dependent and -independent autophagy pathways that are required for efficient lesion repair. In addition, our studies reveal general mechanisms of lesion formation and demonstrate that defective FLNc dimerization via its carboxy-terminal domain does not disturb assembly and basic function of myofibrils. An alternative, more amino-terminally located dimerization site might compensate for that loss. Since filamins function as stress sensors, our data further substantiate that FLNc is important for mechanosensing in the context of Z-disc stabilization and maintenance.
Collapse
|
25
|
Kölbel H, Roos A, van der Ven PFM, Evangelista T, Nolte K, Johnson K, Töpf A, Wilson M, Kress W, Sickmann A, Straub V, Kollipara L, Weis J, Fürst DO, Schara U. First clinical and myopathological description of a myofibrillar myopathy with congenital onset and homozygous mutation in FLNC. Hum Mutat 2020; 41:1600-1614. [PMID: 32516863 DOI: 10.1002/humu.24062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 05/17/2020] [Accepted: 06/07/2020] [Indexed: 02/06/2023]
Abstract
Filamin C (encoded by the FLNC gene) is a large actin-cross-linking protein involved in shaping the actin cytoskeleton in response to signaling events both at the sarcolemma and at myofibrillar Z-discs of cross-striated muscle cells. Multiple mutations in FLNC are associated with myofibrillar myopathies of autosomal-dominant inheritance. Here, we describe for the first time a boy with congenital onset of generalized muscular hypotonia and muscular weakness, delayed motor development but no cardiac involvement associated with a homozygous FLNC mutation c.1325C>G (p.Pro442Arg). We performed ultramorphological, proteomic, and functional investigations as well as immunological studies of known marker proteins for dominant filaminopathies. We show that the mutant protein is expressed in similar quantities as the wild-type variant in control skeletal muscle fibers. The proteomic signature of quadriceps muscle is altered and ultrastructural perturbations are evident. Moreover, filaminopathy marker proteins are comparable both in our homozygous and a dominant control case (c.5161delG). Biochemical investigations demonstrate that the recombinant mutant protein is less stable and more prone to degradation by proteolytic enzymes than the wild-type variant. The unusual congenital presentation of the disease clearly demonstrates that homozygosity for mutations in FLNC severely aggravates the phenotype.
Collapse
Affiliation(s)
- Heike Kölbel
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children's Hospital University of Essen, Essen, Germany
| | - Andreas Roos
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children's Hospital University of Essen, Essen, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Teresinha Evangelista
- Neuromuscular Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, Paris, France
| | - Kay Nolte
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Katherine Johnson
- The John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| | - Ana Töpf
- The John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| | - Michael Wilson
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts.,Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Wolfram Kress
- Department of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Albert Sickmann
- Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, UK.,Medizinische Proteom-Center (MPC), Medizinische Fakultät, Ruhr-Universität Bochum, Bochum, Germany
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK
| | - Laxmikanth Kollipara
- Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Ulrike Schara
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children's Hospital University of Essen, Essen, Germany
| |
Collapse
|
26
|
Structure and Function of Filamin C in the Muscle Z-Disc. Int J Mol Sci 2020; 21:ijms21082696. [PMID: 32295012 PMCID: PMC7216277 DOI: 10.3390/ijms21082696] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
Filamin C (FLNC) is one of three filamin proteins (Filamin A (FLNA), Filamin B (FLNB), and FLNC) that cross-link actin filaments and interact with numerous binding partners. FLNC consists of a N-terminal actin-binding domain followed by 24 immunoglobulin-like repeats with two intervening calpain-sensitive hinges separating R15 and R16 (hinge 1) and R23 and R24 (hinge-2). The FLNC subunit is dimerized through R24 and calpain cleaves off the dimerization domain to regulate mobility of the FLNC subunit. FLNC is localized in the Z-disc due to the unique insertion of 82 amino acid residues in repeat 20 and necessary for normal Z-disc formation that connect sarcomeres. Since phosphorylation of FLNC by PKC diminishes the calpain sensitivity, assembly, and disassembly of the Z-disc may be regulated by phosphorylation of FLNC. Mutations of FLNC result in cardiomyopathy and muscle weakness. Although this review will focus on the current understanding of FLNC structure and functions in muscle, we will also discuss other filamins because they share high sequence similarity and are better characterized. We will also discuss a possible role of FLNC as a mechanosensor during muscle contraction.
Collapse
|
27
|
Verdonschot JAJ, Vanhoutte EK, Claes GRF, Helderman-van den Enden ATJM, Hoeijmakers JGJ, Hellebrekers DMEI, de Haan A, Christiaans I, Lekanne Deprez RH, Boen HM, van Craenenbroeck EM, Loeys BL, Hoedemaekers YM, Marcelis C, Kempers M, Brusse E, van Waning JI, Baas AF, Dooijes D, Asselbergs FW, Barge-Schaapveld DQCM, Koopman P, van den Wijngaard A, Heymans SRB, Krapels IPC, Brunner HG. A mutation update for the FLNC gene in myopathies and cardiomyopathies. Hum Mutat 2020; 41:1091-1111. [PMID: 32112656 PMCID: PMC7318287 DOI: 10.1002/humu.24004] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/12/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022]
Abstract
Filamin C (FLNC) variants are associated with cardiac and muscular phenotypes. Originally, FLNC variants were described in myofibrillar myopathy (MFM) patients. Later, high‐throughput screening in cardiomyopathy cohorts determined a prominent role for FLNC in isolated hypertrophic and dilated cardiomyopathies (HCM and DCM). FLNC variants are now among the more prevalent causes of genetic DCM. FLNC‐associated DCM is associated with a malignant clinical course and a high risk of sudden cardiac death. The clinical spectrum of FLNC suggests different pathomechanisms related to variant types and their location in the gene. The appropriate functioning of FLNC is crucial for structural integrity and cell signaling of the sarcomere. The secondary protein structure of FLNC is critical to ensure this function. Truncating variants with subsequent haploinsufficiency are associated with DCM and cardiac arrhythmias. Interference with the dimerization and folding of the protein leads to aggregate formation detrimental for muscle function, as found in HCM and MFM. Variants associated with HCM are predominantly missense variants, which cluster in the ROD2 domain. This domain is important for binding to the sarcomere and to ensure appropriate cell signaling. We here review FLNC genotype–phenotype correlations based on available evidence.
Collapse
Affiliation(s)
- Job A J Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Els K Vanhoutte
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Godelieve R F Claes
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | - Debby M E I Hellebrekers
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Amber de Haan
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Imke Christiaans
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Ronald H Lekanne Deprez
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Hanne M Boen
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | | | - Bart L Loeys
- Department of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | - Yvonne M Hoedemaekers
- Department of Clinical Genetics, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Carlo Marcelis
- Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marlies Kempers
- Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Esther Brusse
- Department of Neurology, Erasmus MC University Medical Centre, Rotterdam, The Netherlands
| | - Jaap I van Waning
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Annette F Baas
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis Dooijes
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Stephane R B Heymans
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium.,The Netherlands Heart Institute, Utrecht, The Netherlands
| | - Ingrid P C Krapels
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Han G Brunner
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Genetics and Cell Biology, GROW Institute for Developmental Biology and Cancer, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
28
|
Sun Y, Long H, Sun L, Sun X, Pang L, Chen J, Yi Q, Liang T, Shen Y. PGM5 is a promising biomarker and may predict the prognosis of colorectal cancer patients. Cancer Cell Int 2019; 19:253. [PMID: 31582909 PMCID: PMC6771116 DOI: 10.1186/s12935-019-0967-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022] Open
Abstract
Background Phosphoglucomutase (PGM), a key enzyme in the metabolism of glucose-1-phosphate and glucose-6-phosphate, has been found to be associated with proliferation, invasion, and metastasis of cancer. However, the expression and function of PGM5 in colorectal cancer (CRC) remains unknown. Methods We tested PGM5 mRNA and protein expression levels in 79 CRC tissue and their matched adjacent tissue samples by qRT-PCR and immunohistochemistry, respectively. Overall survival (OS) was estimated with the Kaplan-Meier method and compared between groups with the log-rank test. We performed multivariable Cox regression analyses to identify factors associated with CRC risk. The cell proliferation, migration and invasion abilities of CRC cells were detected by using CCK-8, Transwell migration and invasion assays, respectively. Results The PGM5 protein levels expression in CRC tissues were significantly lower than those in the adjacent tissues (t = 5.035, P < 0.001), and Kaplan-Meier analysis indicated that low PGM5 expression were significantly associated with poor overall survival (P = 0.0069). Univariate and multivariate analyses demonstrated that PGM5 was an independent risk factor for overall survival (hazard ratio = 0.3951, P = 0.014). PGM5 overexpression significantly inhibited the proliferation, invasion and migration abilities of CRC cells. On the contrary, knockdown of PGM5 promotes the invasion and migration of CRC cells. Conclusions PMG5 regulates proliferation, invasion, and migration in the CRC and decreased PGM5 is associated with poor prognosis. Therefore, PGM5 is a promising biomarker in CRC and decreased PGM5 may predict poor overall survival in patients with CRC.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Clinical Laboratory, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Haihua Long
- Department of Endoscopy, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Lin Sun
- Department of Clinical Laboratory, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Xiujuan Sun
- Department of Pathology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Liping Pang
- Department of Endoscopy, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Jianlin Chen
- Department of Clinical Laboratory, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Qingqun Yi
- Department of Gastroenterology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Tianwei Liang
- Department of Gastroenterology, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi China
| | - Yongqi Shen
- Department of Oncology, Liuzhou Municipal Liutie Central Hospital, No.14 Fei-e Road, Liuzhou, 545007 Guangxi China
| |
Collapse
|
29
|
Jiao Y, Li Y, Jiang P, Han W, Liu Y. PGM5: a novel diagnostic and prognostic biomarker for liver cancer. PeerJ 2019; 7:e7070. [PMID: 31218127 PMCID: PMC6568247 DOI: 10.7717/peerj.7070] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Liver cancer is a common malignancy and a significant public health problem worldwide, but diagnosis and prognostic evaluation remain challenging for clinicians. Metabolic reprogramming is a hallmark of cancer, and we therefore examined the diagnostic and prognostic value of a metabolic enzyme, phosphoglucomutase-like protein 5 (PGM5), in liver cancer. METHODS All data were from The Cancer Genome Atlas database. R and related statistical packages were used for data analysis. Hepatic PGM5 expression was determined in different groups, and the chi-squared test and Fisher's exact test were used to determine the significance of differences. The pROC package was used to determine receiver operating characteristic (ROC) curves, the survival package was used to for survival analysis and development of a Cox multivariable model, and the ggplot2 package was used for data visualization. RESULTS PGM5 expression was significantly lower in cancerous than adjacent normal liver tissues, and had modest diagnostic value based on ROC analysis and calculations of area under the curve (AUC). Hepatic PGM5 expression had positive associations with male sex and survival, but negative associations with advanced histologic type, advanced histologic grade, advanced stage, and advanced T classification. Patents with low PGM5 levels had poorer overall survival and relapse-free survival. PGM5 was independently associated with patient prognosis. CONCLUSION PGM5 has potential use as a diagnostic and prognostic biomarker for liver cancer.
Collapse
Affiliation(s)
- Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yanqing Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peiqiang Jiang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Han
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Blondelle J, Marrocco V, Clark M, Desmond P, Myers S, Nguyen J, Wright M, Bremner S, Pierantozzi E, Ward S, Estève E, Sorrentino V, Ghassemian M, Lange S. Murine obscurin and Obsl1 have functionally redundant roles in sarcolemmal integrity, sarcoplasmic reticulum organization, and muscle metabolism. Commun Biol 2019; 2:178. [PMID: 31098411 PMCID: PMC6509138 DOI: 10.1038/s42003-019-0405-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/28/2019] [Indexed: 12/19/2022] Open
Abstract
Biological roles of obscurin and its close homolog Obsl1 (obscurin-like 1) have been enigmatic. While obscurin is highly expressed in striated muscles, Obsl1 is found ubiquitously. Accordingly, obscurin mutations have been linked to myopathies, whereas mutations in Obsl1 result in 3M-growth syndrome. To further study unique and redundant functions of these closely related proteins, we generated and characterized Obsl1 knockouts. Global Obsl1 knockouts are embryonically lethal. In contrast, skeletal muscle-specific Obsl1 knockouts show a benign phenotype similar to obscurin knockouts. Only deletion of both proteins and removal of their functional redundancy revealed their roles for sarcolemmal stability and sarcoplasmic reticulum organization. To gain unbiased insights into changes to the muscle proteome, we analyzed tibialis anterior and soleus muscles by mass spectrometry, uncovering additional changes to the muscle metabolism. Our analyses suggest that all obscurin protein family members play functions for muscle membrane systems.
Collapse
Affiliation(s)
- Jordan Blondelle
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Valeria Marrocco
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Madison Clark
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Patrick Desmond
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Stephanie Myers
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Jim Nguyen
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Matthew Wright
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Shannon Bremner
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Samuel Ward
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Eric Estève
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Université Grenoble Alpes, HP2, Grenoble, 38706 France
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, University of California, San Diego, 92093 CA USA
| | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, 413 45 Sweden
| |
Collapse
|
31
|
Segert J, Schneider I, Berger IM, Rottbauer W, Just S. Mediator complex subunit Med12 regulates cardiac jelly development and AV valve formation in zebrafish. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:20-31. [PMID: 30036562 DOI: 10.1016/j.pbiomolbio.2018.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/30/2018] [Accepted: 07/17/2018] [Indexed: 11/25/2022]
Abstract
The molecular mechanism essential for the formation of heart valves involves complex interactions of signaling molecules and transcription factors. The Mediator Complex (MC) functions as multi-subunit machinery to orchestrate gene transcription, especially for tissue-specific fine-tuning of transcriptional processes during development, also in the heart. Here, we analyzed the role of the MC subunit Med12 during atrioventricular canal (AVC) development and endocardial cushion formation, using the Med12-deficient zebrafish mutant trapped (tpd). Whereas primary heart formation was only slightly affected in tpd, we identified defects in AVC development and cardiac jelly formation. We found that although misexpression of bmp4 and versican in tpd hearts can be restored by overexpression of a modified version of the Sox9b transcription factor (harboring VP16 transactivation domain) that functions independent of its co-activator Med12, endocardial cushion development in tpd was not reconstituted. Interestingly, expression of tbx2b and its target hyaluronan synthase 2 (has2) - the synthase of hyaluronan (HA) in the heart - was absent in both uninjected and Sox9b-VP16 overexpressing tpd hearts. HA is a major ECM component of the cardiac jelly and required for endocardial cushion formation. Furthermore, we found secreted phosphoprotein 1 (spp1), an endocardial marker of activated AV endocardial cells, completely absent in tpd hearts, suggesting that crucial steps of the transformation of AV endocardial cells into endocardial cushions is blocked. We demonstrate that Med12 controls cardiac jelly formation Sox9-independently by regulating tbx2b and has2 expression and therefore the production of the glycosaminoglycan HA at the AVC to guarantee proper endocardial cushion development.
Collapse
Affiliation(s)
- Julia Segert
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Isabelle Schneider
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Ina M Berger
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany.
| |
Collapse
|
32
|
Ehsan M, Jiang H, L Thomson K, Gehmlich K. When signalling goes wrong: pathogenic variants in structural and signalling proteins causing cardiomyopathies. J Muscle Res Cell Motil 2017; 38:303-316. [PMID: 29119312 PMCID: PMC5742121 DOI: 10.1007/s10974-017-9487-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/28/2017] [Indexed: 12/20/2022]
Abstract
Cardiomyopathies are a diverse group of cardiac disorders with distinct phenotypes, depending on the proteins and pathways affected. A substantial proportion of cardiomyopathies are inherited and those will be the focus of this review article. With the wide application of high-throughput sequencing in the practice of clinical genetics, the roles of novel genes in cardiomyopathies are recognised. Here, we focus on a subgroup of cardiomyopathy genes [TTN, FHL1, CSRP3, FLNC and PLN, coding for Titin, Four and a Half LIM domain 1, Muscle LIM Protein, Filamin C and Phospholamban, respectively], which, despite their diverse biological functions, all have important signalling functions in the heart, suggesting that disturbances in signalling networks can contribute to cardiomyopathies.
Collapse
Affiliation(s)
- Mehroz Ehsan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - He Jiang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Kate L Thomson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Muenks AG, Stiers KM, Beamer LJ. Sequence-structure relationships, expression profiles, and disease-associated mutations in the paralogs of phosphoglucomutase 1. PLoS One 2017; 12:e0183563. [PMID: 28837627 PMCID: PMC5570346 DOI: 10.1371/journal.pone.0183563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022] Open
Abstract
The key metabolic enzyme phosphoglucomutase 1 (PGM1) controls glucose homeostasis in most human cells. Four proteins related to PGM1, known as PGM2, PGM2L1, PGM3 and PGM5, and referred to herein as paralogs, are encoded in the human genome. Although all members of the same enzyme superfamily, these proteins have distinct substrate preferences and different functional roles. The recent association of PGM1 and PGM3 with inherited enzyme deficiencies prompts us to revisit sequence-structure and other relationships among the PGM1 paralogs, which are understudied despite their importance in human biology. Using currently available sequence, structure, and expression data, we investigated evolutionary relationships, tissue-specific expression profiles, and the amino acid preferences of key active site motifs. Phylogenetic analyses indicate both ancient and more recent divergence between the different enzyme sub-groups comprising the human paralogs. Tissue-specific protein and RNA expression profiles show widely varying patterns for each paralog, providing insight into function and disease pathology. Multiple sequence alignments confirm high conservation of key active site regions, but also reveal differences related to substrate specificity. In addition, we find that sequence variants of PGM2, PGM2L1, and PGM5 verified in the human population affect residues associated with disease-related mutants in PGM1 or PGM3. This suggests that inherited diseases related to dysfunction of these paralogs will likely occur in humans.
Collapse
Affiliation(s)
- Andrew G Muenks
- Biochemistry Department, University of Missouri, Columbia, Missouri, United States of America
| | - Kyle M Stiers
- Biochemistry Department, University of Missouri, Columbia, Missouri, United States of America
| | - Lesa J Beamer
- Biochemistry Department, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
34
|
González-Morales N, Holenka TK, Schöck F. Filamin actin-binding and titin-binding fulfill distinct functions in Z-disc cohesion. PLoS Genet 2017; 13:e1006880. [PMID: 28732005 PMCID: PMC5521747 DOI: 10.1371/journal.pgen.1006880] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/21/2017] [Indexed: 01/03/2023] Open
Abstract
Many proteins contribute to the contractile properties of muscles, most notably myosin thick filaments, which are anchored at the M-line, and actin thin filaments, which are anchored at the Z-discs that border each sarcomere. In humans, mutations in the actin-binding protein Filamin-C result in myopathies, but the underlying molecular function is not well understood. Here we show using Drosophila indirect flight muscle that the filamin ortholog Cheerio in conjunction with the giant elastic protein titin plays a crucial role in keeping thin filaments stably anchored at the Z-disc. We identify the filamin domains required for interaction with the titin ortholog Sallimus, and we demonstrate a genetic interaction of filamin with titin and actin. Filamin mutants disrupting the actin- or the titin-binding domain display distinct phenotypes, with Z-discs breaking up in parallel or perpendicularly to the myofibril, respectively. Thus, Z-discs require filamin to withstand the strong contractile forces acting on them. The Z-disc is a macromolecular complex required to attach and stabilize actin thin filaments in the sarcomere, the smallest contractile unit of striated muscles. Mutations in Z-disc-associated proteins typically result in muscle disorders. Dimeric filamin organizes actin filaments, localizes at the Z-disc in vertebrates and causes muscle disorders in humans when mutated. Despite its clinical relevance, the molecular function of filamin in the sarcomere is not well understood. Here we use Drosophila muscles and an array of filamin mutations to address the molecular and cell biological function of filamin in the sarcomere. We show that filamin mainly serves as a Z-disc cohesive element, binding both thin filaments and titin. This configuration enables filamin to act as a bridge between thin filaments and the elastic scaffold protein titin from the adjacent sarcomere, maintaining sarcomere stability during muscle contraction.
Collapse
Affiliation(s)
| | | | - Frieder Schöck
- Department of Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
35
|
Dierck F, Kuhn C, Rohr C, Hille S, Braune J, Sossalla S, Molt S, van der Ven PFM, Fürst DO, Frey N. The novel cardiac z-disc protein CEFIP regulates cardiomyocyte hypertrophy by modulating calcineurin signaling. J Biol Chem 2017; 292:15180-15191. [PMID: 28717008 DOI: 10.1074/jbc.m117.786764] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Indexed: 11/06/2022] Open
Abstract
The z-disc is a structural component at the lateral borders of the sarcomere and is important for mechanical stability and contractility of both cardiac and skeletal muscles. Of note, the sarcomeric z-disc also represents a nodal point in cardiomyocyte function and signaling. Mutations of numerous z-disc proteins are associated with cardiomyopathies and muscle diseases. To identify additional z-disc proteins that might contribute to cardiac disease, we employed an in silico screen for cardiac-enriched cDNAs. This screen yielded a previously uncharacterized protein named cardiac-enriched FHL2-interacting protein (CEFIP), which exhibited a heart- and skeletal muscle-specific expression profile. Importantly, CEFIP was located at the z-disc and was up-regulated in several models of cardiomyopathy. We also found that CEFIP overexpression induced the fetal gene program and cardiomyocyte hypertrophy. Yeast two-hybrid screens revealed that CEFIP interacts with the calcineurin-binding protein four and a half LIM domains 2 (FHL2). Because FHL2 binds calcineurin, a phosphatase controlling hypertrophic signaling, we examined the effects of CEFIP on the calcineurin/nuclear factor of activated T-cell (NFAT) pathway. These experiments revealed that CEFIP overexpression further enhances calcineurin-dependent hypertrophic signal transduction, and its knockdown repressed hypertrophy and calcineurin/NFAT activity. In summary, we report on a previously uncharacterized protein CEFIP that modulates calcineurin/NFAT signaling in cardiomyocytes, a finding with possible implications for the pathogenesis of cardiomyopathy.
Collapse
Affiliation(s)
- Franziska Dierck
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Christian Kuhn
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Claudia Rohr
- the Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, and
| | - Susanne Hille
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Julia Braune
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Samuel Sossalla
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel.,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| | - Sibylle Molt
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Peter F M van der Ven
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Dieter O Fürst
- the Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Norbert Frey
- From the Department of Internal Medicine III, University Medical Center of Schleswig-Holstein, 24105 Kiel, .,the DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105 Kiel
| |
Collapse
|
36
|
Nikolić N, Görgens SW, Thoresen GH, Aas V, Eckel J, Eckardt K. Electrical pulse stimulation of cultured skeletal muscle cells as a model for in vitro exercise - possibilities and limitations. Acta Physiol (Oxf) 2017; 220:310-331. [PMID: 27863008 DOI: 10.1111/apha.12830] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/28/2016] [Accepted: 11/06/2016] [Indexed: 12/19/2022]
Abstract
The beneficial health-related effects of exercise are well recognized, and numerous studies have investigated underlying mechanism using various in vivo and in vitro models. Although electrical pulse stimulation (EPS) for the induction of muscle contraction has been used for quite some time, its application on cultured skeletal muscle cells of animal or human origin as a model of in vitro exercise is a more recent development. In this review, we compare in vivo exercise and in vitro EPS with regard to effects on signalling, expression level and metabolism. We provide a comprehensive overview of different EPS protocols and their applications, discuss technical aspects of this model including critical controls and the importance of a proper maintenance procedure and finally discuss the limitations of the EPS model.
Collapse
Affiliation(s)
- N. Nikolić
- Department of Pharmaceutical Biosciences; School of Pharmacy; University of Oslo; Oslo Norway
| | - S. W. Görgens
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - G. H. Thoresen
- Department of Pharmaceutical Biosciences; School of Pharmacy; University of Oslo; Oslo Norway
- Department of Pharmacology; Institute of Clinical Medicine; Faculty of Medicine; University of Oslo; Oslo Norway
| | - V. Aas
- Department of Life Sciences and Health; Oslo and Akershus University College of Applied Sciences; Oslo Norway
| | - J. Eckel
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
- German Center for Diabetes Research (DZD e.V.); Düsseldorf Germany
| | - K. Eckardt
- Department of Nutrition; Institute for Basic Medical Sciences; Faculty of Medicine; University of Oslo; Oslo Norway
| |
Collapse
|
37
|
Xin X, Wang T, Liu X, Sui G, Jin C, Yue Y, Yang S, Guo H. A yeast two-hybrid assay reveals CMYA1 interacting proteins. C R Biol 2017; 340:314-323. [DOI: 10.1016/j.crvi.2017.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 05/19/2017] [Accepted: 06/13/2017] [Indexed: 10/19/2022]
|
38
|
Stiers KM, Muenks AG, Beamer LJ. Biology, Mechanism, and Structure of Enzymes in the α-d-Phosphohexomutase Superfamily. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 109:265-304. [PMID: 28683921 PMCID: PMC5802415 DOI: 10.1016/bs.apcsb.2017.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Enzymes in the α-d-phosphohexomutases superfamily catalyze the reversible conversion of phosphosugars, such as glucose 1-phosphate and glucose 6-phosphate. These reactions are fundamental to primary metabolism across the kingdoms of life and are required for a myriad of cellular processes, ranging from exopolysaccharide production to protein glycosylation. The subject of extensive mechanistic characterization during the latter half of the 20th century, these enzymes have recently benefitted from biophysical characterization, including X-ray crystallography, NMR, and hydrogen-deuterium exchange studies. This work has provided new insights into the unique catalytic mechanism of the superfamily, shed light on the molecular determinants of ligand recognition, and revealed the evolutionary conservation of conformational flexibility. Novel associations with inherited metabolic disease and the pathogenesis of bacterial infections have emerged, spurring renewed interest in the long-appreciated functional roles of these enzymes.
Collapse
Affiliation(s)
| | | | - Lesa J Beamer
- University of Missouri, Columbia, MO, United States.
| |
Collapse
|
39
|
Reimann L, Wiese H, Leber Y, Schwäble AN, Fricke AL, Rohland A, Knapp B, Peikert CD, Drepper F, van der Ven PFM, Radziwill G, Fürst DO, Warscheid B. Myofibrillar Z-discs Are a Protein Phosphorylation Hot Spot with Protein Kinase C (PKCα) Modulating Protein Dynamics. Mol Cell Proteomics 2016; 16:346-367. [PMID: 28028127 DOI: 10.1074/mcp.m116.065425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Indexed: 11/06/2022] Open
Abstract
The Z-disc is a protein-rich structure critically important for the development and integrity of myofibrils, which are the contractile organelles of cross-striated muscle cells. We here used mouse C2C12 myoblast, which were differentiated into myotubes, followed by electrical pulse stimulation (EPS) to generate contracting myotubes comprising mature Z-discs. Using a quantitative proteomics approach, we found significant changes in the relative abundance of 387 proteins in myoblasts versus differentiated myotubes, reflecting the drastic phenotypic conversion of these cells during myogenesis. Interestingly, EPS of differentiated myotubes to induce Z-disc assembly and maturation resulted in increased levels of proteins involved in ATP synthesis, presumably to fulfill the higher energy demand of contracting myotubes. Because an important role of the Z-disc for signal integration and transduction was recently suggested, its precise phosphorylation landscape further warranted in-depth analysis. We therefore established, by global phosphoproteomics of EPS-treated contracting myotubes, a comprehensive site-resolved protein phosphorylation map of the Z-disc and found that it is a phosphorylation hotspot in skeletal myocytes, underscoring its functions in signaling and disease-related processes. In an illustrative fashion, we analyzed the actin-binding multiadaptor protein filamin C (FLNc), which is essential for Z-disc assembly and maintenance, and found that PKCα phosphorylation at distinct serine residues in its hinge 2 region prevents its cleavage at an adjacent tyrosine residue by calpain 1. Fluorescence recovery after photobleaching experiments indicated that this phosphorylation modulates FLNc dynamics. Moreover, FLNc lacking the cleaved Ig-like domain 24 exhibited remarkably fast kinetics and exceedingly high mobility. Our data set provides research community resource for further identification of kinase-mediated changes in myofibrillar protein interactions, kinetics, and mobility that will greatly advance our understanding of Z-disc dynamics and signaling.
Collapse
Affiliation(s)
- Lena Reimann
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Heike Wiese
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Yvonne Leber
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Anja N Schwäble
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anna L Fricke
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne Rohland
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Bettina Knapp
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christian D Peikert
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Friedel Drepper
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Peter F M van der Ven
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Gerald Radziwill
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,§BIOSS Centre for Biological Signalling Studies, University of Freiburg
| | - Dieter O Fürst
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Bettina Warscheid
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; .,§BIOSS Centre for Biological Signalling Studies, University of Freiburg
| |
Collapse
|
40
|
Ferraz EP, Oliveira FS, de Oliveira PT, Crovace MC, Peitl-Filho O, Beloti MM, Rosa AL. Bioactive glass-based surfaces induce differential gene expression profiling of osteoblasts. J Biomed Mater Res A 2016; 105:419-423. [PMID: 27682446 DOI: 10.1002/jbm.a.35915] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 01/08/2023]
Abstract
The ability of Biosilicate® with two crystalline phases (BioS-2P) to drive osteoblast differentiation encourages the investigation of the cellular mechanisms involved in this process. Then, the aim of our study was to analyze the large-scale gene expression of osteoblasts grown on BioS-2P compared with Bioglass® 45S5 (45S5). Osteoblasts differentiated from rat bone marrow mesenchymal stem cells were cultured under osteogenic conditions on BioS-2P, 45S5 and polystyrene (control). After 10 days, the expression of 23,794 genes was analyzed using mRNA Sequencing and the data were validated by real-time PCR. The BioS-2P exhibited 5 genes upregulated and 3 downregulated compared with 45S5. Compared with control, BioS-2P upregulated 15 and downregulated 11 genes, while 45S5 upregulated 25 and downregulated 21 genes. Eight genes were commonly upregulated and 4 downregulated by both bioactive glasses. In conclusion, our results demonstrated that bioactive glasses affect the gene expression profiling of osteoblasts. Most of the regulated genes by both BioS-2P and 45S5 are associated with the process of mineralization highlighting their osteostimulation property that is, at least in part, derived from the ability to modulate the intracellular machinery to promote osteoblast genotype expression. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 419-423, 2017.
Collapse
Affiliation(s)
- Emanuela P Ferraz
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Fabiola S Oliveira
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Paulo T de Oliveira
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Murilo C Crovace
- Vitreous Materials Laboratory (LaMaV), Federal University of São Carlos, Rod Washington Luis, Km 235, São Carlos, SP, 13565-905, Brazil
| | - Oscar Peitl-Filho
- Vitreous Materials Laboratory (LaMaV), Federal University of São Carlos, Rod Washington Luis, Km 235, São Carlos, SP, 13565-905, Brazil
| | - Marcio M Beloti
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| | - Adalberto L Rosa
- Cell Culture Laboratory, School of Dentistry of Ribeirão Preto, University of São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-904, Brazil
| |
Collapse
|
41
|
Fröhlich T, Kemter E, Flenkenthaler F, Klymiuk N, Otte KA, Blutke A, Krause S, Walter MC, Wanke R, Wolf E, Arnold GJ. Progressive muscle proteome changes in a clinically relevant pig model of Duchenne muscular dystrophy. Sci Rep 2016; 6:33362. [PMID: 27634466 PMCID: PMC5025886 DOI: 10.1038/srep33362] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 08/24/2016] [Indexed: 01/16/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by genetic deficiency of dystrophin and characterized by massive structural and functional changes of skeletal muscle tissue, leading to terminal muscle failure. We recently generated a novel genetically engineered pig model reflecting pathological hallmarks of human DMD better than the widely used mdx mouse. To get insight into the hierarchy of molecular derangements during DMD progression, we performed a proteome analysis of biceps femoris muscle samples from 2-day-old and 3-month-old DMD and wild-type (WT) pigs. The extent of proteome changes in DMD vs. WT muscle increased markedly with age, reflecting progression of the pathological changes. In 3-month-old DMD muscle, proteins related to muscle repair such as vimentin, nestin, desmin and tenascin C were found to be increased, whereas a large number of respiratory chain proteins were decreased in abundance in DMD muscle, indicating serious disturbances in aerobic energy production and a reduction of functional muscle tissue. The combination of proteome data for fiber type specific myosin heavy chain proteins and immunohistochemistry showed preferential degeneration of fast-twitch fiber types in DMD muscle. The stage-specific proteome changes detected in this large animal model of clinically severe muscular dystrophy provide novel molecular readouts for future treatment trials.
Collapse
Affiliation(s)
- Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Kathrin A Otte
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology, Centre for Clinical Veterinary Medicine, LMU Munich, Veterinärstr. 13, D-80539 Munich, Germany
| | - Sabine Krause
- Friedrich-Baur-Institute, Department of Neurology, LMU Munich, Ziemssenstr. 1, D-80336 Munich, Germany
| | - Maggie C Walter
- Friedrich-Baur-Institute, Department of Neurology, LMU Munich, Ziemssenstr. 1, D-80336 Munich, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Centre for Clinical Veterinary Medicine, LMU Munich, Veterinärstr. 13, D-80539 Munich, Germany
| | - Eckhard Wolf
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany.,Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| |
Collapse
|
42
|
Kebir S, Orfanos Z, Schuld J, Linhart M, Lamberz C, van der Ven PFM, Schrickel J, Kirfel G, Fürst DO, Meyer R. Sarcomeric lesions and remodeling proximal to intercalated disks in overload-induced cardiac hypertrophy. Exp Cell Res 2016; 348:95-105. [PMID: 27639425 DOI: 10.1016/j.yexcr.2016.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 10/21/2022]
Abstract
Pressure overload induces cardiac remodeling involving both the contractile machinery and intercalated disks (IDs). Filamin C (FlnC) and Xin actin-binding repeat-containing proteins (XIRPs) are multi-adapters localizing in IDs of higher vertebrates. Knockout of the gene encoding Xin (Xirp1) in mice leads to a mild cardiac phenotype with ID mislocalization. In order to amplify this phenotype, we performed transverse aortic constriction (TAC) on control and Xirp1-deficient mice. TAC induced similar left ventricular hypertrophy in both genotypes, suggesting that the lack of Xin does not lead to higher susceptibility to cardiac overload. However, in both genotypes, FlnC appeared in "streaming" localizations across multiple sarcomeres proximal to the IDs, suggesting a remodeling response. Furthermore, FlnC-positive areas of remodeling, reminiscent of sarcomeric lesions previously described for skeletal muscles (but so far unreported in the heart), were also observed. These adaptations reflect a similarly strong effect of the pressure induced by TAC in both genotypes. However, 2 weeks post-operation TAC-treated knockout hearts had reduced levels of connexin43 and slightly increased incidents of ventricular tachycardia compared to their wild-type (WT) counterparts. Our findings highlight the FlnC-positive sarcomeric lesions and ID-proximal streaming as general remodeling responses in cardiac overload-induced hypertrophy.
Collapse
Affiliation(s)
- Sied Kebir
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115 Bonn, Germany.
| | - Zacharias Orfanos
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Julia Schuld
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Markus Linhart
- Department of Medicine-Cardiology, University of Bonn Medical Center, Sigmund-Freud-Straße 25, 53127 Bonn, Germany.
| | - Christian Lamberz
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Peter F M van der Ven
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Jan Schrickel
- Department of Medicine-Cardiology, University of Bonn Medical Center, Sigmund-Freud-Straße 25, 53127 Bonn, Germany.
| | - Gregor Kirfel
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Dieter O Fürst
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Rainer Meyer
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115 Bonn, Germany.
| |
Collapse
|
43
|
Leber Y, Ruparelia AA, Kirfel G, van der Ven PFM, Hoffmann B, Merkel R, Bryson-Richardson RJ, Fürst DO. Filamin C is a highly dynamic protein associated with fast repair of myofibrillar microdamage. Hum Mol Genet 2016; 25:2776-2788. [PMID: 27206985 DOI: 10.1093/hmg/ddw135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 11/12/2022] Open
Abstract
Filamin c (FLNc) is a large dimeric actin-binding protein located at premyofibrils, myofibrillar Z-discs and myofibrillar attachment sites of striated muscle cells, where it is involved in mechanical stabilization, mechanosensation and intracellular signaling. Mutations in the gene encoding FLNc give rise to skeletal muscle diseases and cardiomyopathies. Here, we demonstrate by fluorescence recovery after photobleaching that a large fraction of FLNc is highly mobile in cultured neonatal mouse cardiomyocytes and in cardiac and skeletal muscles of live transgenic zebrafish embryos. Analysis of cardiomyocytes from Xirp1 and Xirp2 deficient animals indicates that both Xin actin-binding repeat-containing proteins stabilize FLNc selectively in premyofibrils. Using a novel assay to analyze myofibrillar microdamage and subsequent repair in cultured contracting cardiomyocytes by live cell imaging, we demonstrate that repair of damaged myofibrils is achieved within only 4 h, even in the absence of de novo protein synthesis. FLNc is immediately recruited to these sarcomeric lesions together with its binding partner aciculin and precedes detectable assembly of filamentous actin and recruitment of other myofibrillar proteins. These data disclose an unprecedented degree of flexibility of the almost crystalline contractile machinery and imply FLNc as a dynamic signaling hub, rather than a primarily structural protein. Our myofibrillar damage/repair model illustrates how (cardio)myocytes are kept functional in their mechanically and metabolically strained environment. Our results help to better understand the pathomechanisms and pathophysiology of early stages of FLNc-related myofibrillar myopathy and skeletal and cardiac diseases preceding pathological protein aggregation.
Collapse
Affiliation(s)
- Yvonne Leber
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Avnika A Ruparelia
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia
| | - Gregor Kirfel
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Bernd Hoffmann
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and
| | - Rudolf Merkel
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and.,Department of Biomechanics, Institute for Physical and Theoretical Chemistry, University of Bonn, D53115 Bonn, Germany
| | | | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| |
Collapse
|
44
|
Orfanos Z, Gödderz MPO, Soroka E, Gödderz T, Rumyantseva A, van der Ven PFM, Hawke TJ, Fürst DO. Breaking sarcomeres by in vitro exercise. Sci Rep 2016; 6:19614. [PMID: 26804343 PMCID: PMC4726327 DOI: 10.1038/srep19614] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/16/2015] [Indexed: 11/30/2022] Open
Abstract
Eccentric exercise leads to focal disruptions in the myofibrils, referred to as “lesions”. These structures are thought to contribute to the post-exercise muscle weakness, and to represent areas of mechanical damage and/or remodelling. Lesions have been investigated in human biopsies and animal samples after exercise. However, this approach does not examine the mechanisms behind lesion formation, or their behaviour during contraction. To circumvent this, we used electrical pulse stimulation (EPS) to simulate exercise in C2C12 myotubes, combined with live microscopy. EPS application led to the formation of sarcomeric lesions in the myotubes, resembling those seen in exercised mice, increasing in number with the time of application or stimulation intensity. Furthermore, transfection with an EGFP-tagged version of the lesion and Z-disc marker filamin-C allowed us to observe the formation of lesions using live cell imaging. Finally, using the same technique we studied the behaviour of these structures during contraction, and observed them to be passively stretching. This passive behaviour supports the hypothesis that lesions contribute to the post-exercise muscle weakness, protecting against further damage. We conclude that EPS can be reliably used as a model for the induction and study of sarcomeric lesions in myotubes in vitro.
Collapse
Affiliation(s)
- Zacharias Orfanos
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Markus P O Gödderz
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Ekaterina Soroka
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Tobias Gödderz
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Anastasia Rumyantseva
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Peter F M van der Ven
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8N 3Z5, Canada
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| |
Collapse
|
45
|
Chevessier F, Schuld J, Orfanos Z, Plank AC, Wolf L, Maerkens A, Unger A, Schlötzer-Schrehardt U, Kley RA, Von Hörsten S, Marcus K, Linke WA, Vorgerd M, van der Ven PFM, Fürst DO, Schröder R. Myofibrillar instability exacerbated by acute exercise in filaminopathy. Hum Mol Genet 2015; 24:7207-20. [PMID: 26472074 DOI: 10.1093/hmg/ddv421] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/02/2015] [Indexed: 12/12/2022] Open
Abstract
Filamin C (FLNC) mutations in humans cause myofibrillar myopathy (MFM) and cardiomyopathy, characterized by protein aggregation and myofibrillar degeneration. We generated the first patient-mimicking knock-in mouse harbouring the most common disease-causing filamin C mutation (p.W2710X). These heterozygous mice developed muscle weakness and myofibrillar instability, with formation of filamin C- and Xin-positive lesions streaming between Z-discs. These lesions, which are distinct from the classical MFM protein aggregates by their morphology and filamentous appearance, were greatly increased in number upon acute physical exercise in the mice. This pathology suggests that mutant filamin influences the mechanical stability of myofibrillar Z-discs, explaining the muscle weakness in mice and humans. Re-evaluation of biopsies from MFM-filaminopathy patients with different FLNC mutations revealed a similar, previously unreported lesion pathology, in addition to the classical protein aggregates, and suggested that structures previously interpreted as aggregates may be in part sarcomeric lesions. We postulate that these lesions define preclinical disease stages, preceding the formation of protein aggregates.
Collapse
Affiliation(s)
| | - Julia Schuld
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Zacharias Orfanos
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Anne-C Plank
- Department for Experimental Therapy, Preclinical Experimental Animal Center and
| | | | - Alexandra Maerkens
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil, Department of Functional Proteomics, Medizinisches Proteom-Center and
| | - Andreas Unger
- Department of Cardiovascular Physiology, Ruhr-University Bochum, Bochum, Germany
| | | | - Rudolf A Kley
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil
| | - Stephan Von Hörsten
- Department for Experimental Therapy, Preclinical Experimental Animal Center and
| | - Katrin Marcus
- Department of Functional Proteomics, Medizinisches Proteom-Center and
| | - Wolfgang A Linke
- Department of Cardiovascular Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany,
| | | |
Collapse
|
46
|
Cross-tissue and cross-species analysis of gene expression in skeletal muscle and electric organ of African weakly-electric fish (Teleostei; Mormyridae). BMC Genomics 2015; 16:668. [PMID: 26335922 PMCID: PMC4558960 DOI: 10.1186/s12864-015-1858-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/18/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND African weakly-electric fishes of the family Mormyridae are able to produce and perceive weak electric signals (typically less than one volt in amplitude) owing to the presence of a specialized, muscle-derived electric organ (EO) in their tail region. Such electric signals, also known as Electric Organ Discharges (EODs), are used for objects/prey localization, for the identification of conspecifics, and in social and reproductive behaviour. This feature might have promoted the adaptive radiation of this family by acting as an effective pre-zygotic isolation mechanism. Despite the physiological and evolutionary importance of this trait, the investigation of the genetic basis of its function and modification has so far remained limited. In this study, we aim at: i) identifying constitutive differences in terms of gene expression between electric organ and skeletal muscle (SM) in two mormyrid species of the genus Campylomormyrus: C. compressirostris and C. tshokwe, and ii) exploring cross-specific patterns of gene expression within the two tissues among C. compressirostris, C. tshokwe, and the outgroup species Gnathonemus petersii. RESULTS Twelve paired-end (100 bp) strand-specific RNA-seq Illumina libraries were sequenced, producing circa 330 M quality-filtered short read pairs. The obtained reads were assembled de novo into four reference transcriptomes. In silico cross-tissue DE-analysis allowed us to identify 271 shared differentially expressed genes between EO and SM in C. compressirostris and C.tshokwe. Many of these genes correspond to myogenic factors, ion channels and pumps, and genes involved in several metabolic pathways. Cross-species analysis has revealed that the electric organ transcriptome is more variable in terms of gene expression levels across species than the skeletal muscle transcriptome. CONCLUSIONS The data obtained indicate that: i) the loss of contractile activity and the decoupling of the excitation-contraction processes are reflected by the down-regulation of the corresponding genes in the electric organ's transcriptome; ii) the metabolic activity of the EO might be specialized towards the production and turn-over of membrane structures; iii) several ion channels are highly expressed in the EO in order to increase excitability; iv) several myogenic factors might be down-regulated by transcription repressors in the EO.
Collapse
|
47
|
Al-Sajee D, Nissar AA, Coleman SK, Rebalka IA, Chiang A, Wathra R, van der Ven PFM, Orfanos Z, Hawke TJ. Xin-deficient mice display myopathy, impaired contractility, attenuated muscle repair and altered satellite cell functionality. Acta Physiol (Oxf) 2015; 214:248-60. [PMID: 25582411 DOI: 10.1111/apha.12455] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/20/2014] [Accepted: 01/07/2015] [Indexed: 12/26/2022]
Abstract
AIM Xin is an F-actin-binding protein expressed during development of cardiac and skeletal muscle. We used Xin-/- mice to determine the impact of Xin deficiency on different aspects of skeletal muscle health, including functionality and regeneration. METHODS Xin-/- skeletal muscles and their satellite cell (SC) population were investigated for the presence of myopathic changes by a series of histological and immunofluorescent stains on resting uninjured muscles. To further understand the effect of Xin loss on muscle health and its SCs, we studied SCs responses following cardiotoxin-induced muscle injury. Functional data were determined using in situ muscle stimulation protocol. RESULTS Compared to age-matched wild-type (WT), Xin-/- muscles exhibited generalized myopathy and increased fatigability with a significantly decreased force recovery post-fatiguing contractions. Muscle regeneration was attenuated in Xin-/- mice. This impaired regeneration prompted an investigation into SC content and functionality. Although SC content was not different, significantly more activated SCs were present in Xin-/- vs. WT muscles. Primary Xin-/- myoblasts displayed significant reductions (approx. 50%) in proliferative capacity vs. WT; a finding corroborated by significantly decreased MyoD-positive nuclei in 3 days post-injury Xin-/- muscle vs. WT. As more activated SCs did not translate to more proliferating myoblasts, we investigated whether Xin-/- SCs displayed an exaggerated loss by apoptosis. More apoptotic SCs (TUNEL+/Pax7+) were present in Xin-/- muscle vs. WT. Furthermore, more Xin-/- myoblasts were expressing nuclear caspase-3 compared to WT at 3 days post-injury. CONCLUSION Xin deficiency leads to a myopathic condition characterized by increased muscle fatigability, impaired regeneration and SC dysfunction.
Collapse
Affiliation(s)
- D. Al-Sajee
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| | - A. A. Nissar
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| | - S. K. Coleman
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| | - I. A. Rebalka
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| | - A. Chiang
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| | - R. Wathra
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| | | | - Z. Orfanos
- Institute for Cell Biology; University of Bonn; Bonn Germany
| | - T. J. Hawke
- Pathology and Molecular Medicine; McMaster University; Hamilton ON Canada
| |
Collapse
|
48
|
Bührdel JB, Hirth S, Kessler M, Westphal S, Forster M, Manta L, Wiche G, Schoser B, Schessl J, Schröder R, Clemen CS, Eichinger L, Fürst DO, van der Ven PFM, Rottbauer W, Just S. In vivo characterization of human myofibrillar myopathy genes in zebrafish. Biochem Biophys Res Commun 2015; 461:217-23. [PMID: 25866181 DOI: 10.1016/j.bbrc.2015.03.149] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/26/2015] [Indexed: 01/31/2023]
Abstract
Myofibrillar myopathies (MFM) are progressive diseases of human heart and skeletal muscle with a severe impact on life quality and expectancy of affected patients. Although recently several disease genes for myofibrillar myopathies could be identified, today most genetic causes and particularly the associated mechanisms and signaling events that lead from the mutation to the disease phenotype are still mostly unknown. To assess whether the zebrafish is a suitable model system to validate MFM candidate genes using targeted antisense-mediated knock-down strategies, we here specifically inactivated known human MFM disease genes and evaluated the resulting muscular and cardiac phenotypes functionally and structurally. Consistently, targeted ablation of MFM genes in zebrafish led to compromised skeletal muscle function mostly due to myofibrillar degeneration as well as severe heart failure. Similar to what was shown in MFM patients, MFM gene-deficient zebrafish showed pronounced gene-specific phenotypic and structural differences. In summary, our results indicate that the zebrafish is a suitable model to functionally and structurally evaluate novel MFM disease genes in vivo.
Collapse
Affiliation(s)
- John B Bührdel
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | - Sofia Hirth
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | - Mirjam Kessler
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | - Sören Westphal
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | - Monika Forster
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | - Linda Manta
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | - Gerhard Wiche
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, 1030 Vienna, Austria
| | - Benedikt Schoser
- Department of Neurology, Friedrich-Baur-Institut, Ludwig-Maximilians-University, Munich, Germany
| | - Joachim Schessl
- Department of Neurology, Friedrich-Baur-Institut, Ludwig-Maximilians-University, Munich, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Christoph S Clemen
- Institute for Biochemistry I, University of Cologne, 50931 Köln, Germany
| | - Ludwig Eichinger
- Institute for Biochemistry I, University of Cologne, 50931 Köln, Germany
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | | | - Wolfgang Rottbauer
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany.
| | - Steffen Just
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany.
| |
Collapse
|