1
|
Lopes LSF, Fonseca PAS, Makanjuola BO, Miglior F, Tulpan D, Baes CF, Schenkel FS. A genome-wide association study on rumination time in first-lactation dairy cattle. J Dairy Sci 2025:S0022-0302(25)00274-7. [PMID: 40306420 DOI: 10.3168/jds.2024-26054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025]
Abstract
Rumination time (RT) in dairy cattle is a crucial indicator of health, production, reproduction, and greenhouse gas emissions. With moderate heritability estimates for RT, there is potential for further analyses regarding the genetic architecture of the trait. To identify genomic regions associated with RT, we conducted a GWAS on SNPs in a cohort of 452 mid-first-lactation Canadian Holstein cows, followed by the annotation of genes and enrichment analyses of quantitative trait loci (QTL). Animals were genotyped using a medium-density SNP panel (50 K). Quality control measures were used to remove markers residing on nonautosomal chromosomes or with minor allele frequencies <5%, and SNP or animals with call rates lower than 90%. The SNP effects were estimated using single-step genomic BLUP. Significant markers were identified using a chromosome-wise modified Bonferroni correction, based on the expected number of independent chromosome segments. We identified 35 SNPs significantly associated with RT, mapping 34 genes within a 50-kbp interval up and downstream from these SNPs. Additionally, 19 QTL were found enriched in these genomic regions. Notably, genes such as ATP2B4, LDB3,WARS2, and PTPRO were identified, suggesting potential links to muscle fiber activity and milk solids percentage. The enriched QTL were associated with traits related to fat and protein synthesis and deposition in both milk and muscle tissues. Gene Ontology analysis highlighted terms related to muscle contraction and neuronal communication, consistent with the physiological processes underlying RT. Our findings offer new insights into the genetic architecture of RT, advancing the understanding of the physiological mechanisms governing this complex trait.
Collapse
Affiliation(s)
- L S F Lopes
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - P A S Fonseca
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Spain
| | - B O Makanjuola
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - F Miglior
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; Lactanet Canada, Guelph, ON N1K 1E5, Canada
| | - D Tulpan
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - C F Baes
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - F S Schenkel
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
2
|
Bahout M, Severa G, Kamoun E, Bouhour F, Pegat A, Toutain A, Lagrange E, Duval F, Tard C, De la Cruz E, Féasson L, Jacquin-Piques A, Richard P, Métay C, Cavalli M, Romero NB, Evangelista T, Sole G, Carlier RY, Laforêt P, Acket B, Behin A, Fernández-Eulate G, Léonard-Louis S, Quijano-Roy S, Pereon Y, Salort-Campana E, Nadaj-Pakleza A, Masingue M, Malfatti E, Stojkovic T, Villar-Quiles RN. MYH7-related myopathies: clinical, myopathological and genotypic spectrum in a multicentre French cohort. J Neurol Neurosurg Psychiatry 2025; 96:453-461. [PMID: 39448255 PMCID: PMC12015026 DOI: 10.1136/jnnp-2024-334263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/27/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Myosin heavy chain 7 (MYH7)-related myopathies (MYH7-RMs) are a group of muscle disorders linked to pathogenic variants in the MYH7 gene, encoding the slow/beta-cardiac myosin heavy chain, which is highly expressed in skeletal muscle and heart. The phenotype is heterogeneous including distal, predominantly axial or scapuloperoneal myopathies with variable cardiac involvement. METHODS We retrospectively analysed the clinical, muscle MRI, genetic and myopathological features of 57 MYH7 patients. Patients received a thorough neurological (n=57, 100%), cardiac (n=51, 89%) and respiratory (n=45, 79%) assessment. Muscle imaging findings and muscle biopsies were reappraised in 19 (33%) and 27 (47%) patients, respectively. RESULTS We identified three phenotypes with varying degrees of overlap: distal myopathy (70%), scapuloperoneal (23%) and axial with peculiar cervical spine rigidity called the 'sphinx' phenotype (7%). 14% of patients had either dilated cardiomyopathy, hypertrophic cardiomyopathy or left ventricular non-compaction cardiomyopathy. 31% of patients had prominent respiratory involvement, including all patients with the 'sphinx' phenotype. Muscle MRI showed involvement of tibialis anterior, followed by quadriceps, and erector spinae in patients with axial phenotype. Cores represented the most common myopathological lesion. We report 26 pathogenic variants of MYH7 gene, 9 of which are novel. CONCLUSIONS MYH7-RMs have a large phenotypic spectrum, including distal, scapuloperoneal or axial weakness, and variable cardiac and respiratory involvement. Tibialis anterior is constantly and precociously affected both clinically and on muscle imaging. Cores represent the most common myopathological lesion. Our detailed description of MYH7-RMs should improve their recognition and management.
Collapse
Affiliation(s)
- Marie Bahout
- Assistance Publique Hôpitaux de Paris, Département de Neurologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Gianmarco Severa
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, Créteil, France
- APHP, Neuromsucular Reference Center, Hôpitaux Universitaires Henri Mondor, Creteil, France
| | - Emna Kamoun
- Service de neurologie, Hôpital Paris-Saclay, Orsay, France
| | - Françoise Bouhour
- Service ENMG et de pathologies neuromusculaires, centre de référence des maladies neuromusculaires PACA-Réunion-Rhône Alpes, Hôpital Neurologique P. Wertheimer, Hospices Civils, Lyon, France
| | - Antoine Pegat
- Service ENMG et de pathologies neuromusculaires, centre de référence des maladies neuromusculaires PACA-Réunion-Rhône Alpes, Hôpital Neurologique P. Wertheimer, Hospices Civils, Lyon, France
| | - Annick Toutain
- CHRU Tours Pôle de Gynécologie Obstétrique Médecine fœtale et Reproduction, Tours, France
| | - Emmeline Lagrange
- Département de Neurologie, Centre de Référence des Maladies Neuromusculaires, CHU de Grenoble, Grenoble, France
| | - Fanny Duval
- Service de Neurologie, CHU Bordeaux, Pessac, France
| | - Celine Tard
- U1172, service de neurologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, CHU de Lille, Lille, France
- Filière nationale, FILNEMUS, France
| | - Elisa De la Cruz
- Filière nationale, FILNEMUS, France
- Department of Neurology, Montpellier University Hospital Center, Gui de Chauliac Hospital, Montpellier, France
| | - Léonard Féasson
- Filière nationale, FILNEMUS, France
- UJM-Saint-Etienne, Inter-university Laboratory of Human Movement Biology, EA 7424, Unit of Myology, Neuromuscular Reference Center Euro-NmD, University Hospital, Saint-Etienne, France
| | - Agnès Jacquin-Piques
- Service de Neurophysiologie adulte, University Hospital Centre Dijon, Dijon, France
| | - Pascale Richard
- Unité Fonctionnelle de Cardiogénétique et Myogénétique moléculaire et cellulaire, Centre de Génétique Moléculaire et Chromosomique, Hôpital Pitié-Salpêtrière, INSERM UMRS1166, Sorbonne Université, Paris, France
| | - Corinne Métay
- Filière nationale, FILNEMUS, France
- AP-HP, Pitie-Salpetriere hospital, Molecular and Chromosomic Genetics Center, Cardiogenetic and myogenetic Functional Unit, and INSERM UMRS 974, Sorbonne University, Institute of Myology, Paris, France
| | - Michele Cavalli
- Filière nationale, FILNEMUS, France
- Peripheral Nervous System and Muscle Department, CHU Nice, Hôpital Pasteur 2, Nice, France
| | - Norma Beatriz Romero
- Unité de morphologie Neuromusculaire, Institut de Myologie, GHU La Pitié-Salpêtrière; Université Pierre et Marie Curie-Paris6; INSERM UMR974, Paris, France
| | - Teresinha Evangelista
- Filière nationale, FILNEMUS, France
- Institut de Myologie, Paris, France
- European Reference Network for Rare Neuromuscular Diseases, (EURO-NMD), France
| | - Guilhem Sole
- Centre de référence des maladies neuromusculaires, Service de neurologie et des maladies neuromusculaires, CHU de Bordeaux (Hôpital Pellegrin), FILNEMUS, EURO-NMD, Bordeaux, France
| | - Robert Yves Carlier
- AP-HP, GHU Paris Saclay, Hôpital Raymond Poincaré, DMU Smart Imaging, UMR1179 INSERM, Garches, France
| | - Pascal Laforêt
- Filière nationale, FILNEMUS, France
- Neurology Department, Raymond Poincaré University Hospital, Assistance Publique des Hopitaux de Paris, Garches, France. Nord-Est-Ile-de-France Neuromuscular Reference Center, Fédération Hospitalo Universitaire PHENIX, Garches, France, INSERM U 1179, Paris-Saclay University, Versailles, France
| | - Blandine Acket
- Department of Neurology, Toulouse University Hospital, Toulouse, France
| | - Anthony Behin
- Filière nationale, FILNEMUS, France
- APHP, service de neuromyologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, GH Pitié-Salpêtrière, Paris, France
| | - Gorka Fernández-Eulate
- Filière nationale, FILNEMUS, France
- APHP, service de neuromyologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, GH Pitié-Salpêtrière, Paris, France
| | - Sarah Léonard-Louis
- Filière nationale, FILNEMUS, France
- APHP, service de neuromyologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, GH Pitié-Salpêtrière, Paris, France
| | - Susana Quijano-Roy
- Filière nationale, FILNEMUS, France
- European Reference Network for Rare Neuromuscular Diseases, (EURO-NMD), France
- APHP, service de Neurologie Pédiatrique et Réanimation, Centre de Référence Neuromusculaire Nord/Est/Ile-de-France (FILNEMUS), Hôpital Raymond Poincaré (UVSQ). GH Université Paris-Saclay, Garches, France
| | - Yann Pereon
- Filière nationale, FILNEMUS, France
- CHU Nantes, Centre de Référence des Maladies Neuromusculaires AOC, Filnemus, Euro-NMD, Hôtel-Dieu, Nantes, France
| | - Emmanuelle Salort-Campana
- Filière nationale, FILNEMUS, France
- Centre de référence neuromusculaire PACA réunion Rhône-Alpes, service du Pr Attarian, AP HM, Marseille, France
| | - Aleksandra Nadaj-Pakleza
- Filière nationale, FILNEMUS, France
- Centre de Reference des Maladies Neuromusculaires Nord-Est-Ile de France, Department of Neurology, University Hospital Centre, Strasbourg, France
| | - Marion Masingue
- Filière nationale, FILNEMUS, France
- APHP, service de neuromyologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, GH Pitié-Salpêtrière, Paris, France
| | - Edoardo Malfatti
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, Créteil, France
- APHP, Neuromsucular Reference Center, Hôpitaux Universitaires Henri Mondor, Creteil, France
- Filière nationale, FILNEMUS, France
- European Reference Network for Rare Neuromuscular Diseases, (EURO-NMD), France
| | - Tanya Stojkovic
- Filière nationale, FILNEMUS, France
- APHP, service de neuromyologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, GH Pitié-Salpêtrière, Paris, France
- Sorbonne University, Myology research center, UMRS974, Paris, France
| | - Rocío Nur Villar-Quiles
- Filière nationale, FILNEMUS, France
- APHP, service de neuromyologie, centre de référence de pathologie neuromusculaire Nord/Est/Ile-de-France, GH Pitié-Salpêtrière, Paris, France
- Sorbonne University, Myology research center, UMRS974, Paris, France
| |
Collapse
|
3
|
Sun S, Lu YN, Li XD. Structure of the Inhibited Smooth Muscle Myosin and Its Implications on the Regulation of Insect Striated Muscle Myosin. Life (Basel) 2025; 15:379. [PMID: 40141724 PMCID: PMC11944230 DOI: 10.3390/life15030379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/07/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Class II myosin (myosin-2) is an actin-based motor protein found in nearly all eukaryotes. One critical question is how the motor function of myosin-2 is regulated. Vertebrate myosin-2 comprises non-muscle myosin, smooth muscle myosin and striated muscle myosin. Recent studies have shown that smooth muscle myosin, in its inhibited state, adopts a folded conformation in which the two heads interact with each other asymmetrically, and the tail is folded into three segments that wrap around the two heads. It has been proposed that the asymmetric head-to-head interaction is a conserved, fundamental structure essential for the regulation of all types of myosin-2. Nearly all insects have only a single striated muscle myosin heavy chain (MHC) gene, which produces all MHC isoforms through alternative splicing of mutually exclusive exons. Most of the alternative exon-encoded regions in insect MHC are located in the motor domain and are critical for generating isoform-specific contraction velocity and force production. However, it remains unclear whether these alternative exon-encoded regions participate in the regulation of insect striated muscle myosin. Here, we review the recently resolved structure of the inhibited state of smooth muscle myosin and discuss its implications on the regulation of insect striated muscle myosin. We propose that the alternative exon-encoded regions in insect MHC not only affect motor properties but also contribute to stabilizing the folded conformation and play a crucial role in regulating insect striated muscle myosin.
Collapse
Affiliation(s)
- Shaopeng Sun
- Group of Cell Motility and Muscle Contraction, State Key Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.S.); (Y.-N.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Ning Lu
- Group of Cell Motility and Muscle Contraction, State Key Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.S.); (Y.-N.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang-dong Li
- Group of Cell Motility and Muscle Contraction, State Key Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.S.); (Y.-N.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Ni K, Liu Y, DI P, Wang L, Huang H, Holsinger RMD, Kiang KMY, Jiao J. Chromobox protein homolog 7 suppresses the stem-like phenotype of glioblastoma cells by regulating the myosin heavy chain 9-NF-κB signaling pathway. Cell Death Discov 2025; 11:74. [PMID: 39988672 PMCID: PMC11847914 DOI: 10.1038/s41420-025-02362-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Cancer stem cells (CSCs) are significant factors in the treatment resistance and recurrence of glioblastoma. Chromobox protein homolog 7 (CBX7) can inhibit the progression of various tumors, but its impact on the stem cell-like properties of glioblastoma cells remains unclear. Clinically, low levels of CBX7 are associated with poor prognosis and increased distant metastasis in glioblastoma patients, and this low expression is caused by methylation of the CBX7 promoter. Our current research indicates that CBX7 plays a key role in suppressing the stem-like phenotype of glioblastoma. In this study, through bioinformatics analysis, we found that CBX7 is the most significantly downregulated member of the CBX family in glioblastoma and is closely associated with the stem-like phenotype of glioblastoma cells. We show that CBX7 promotes the degradation of myosin heavy chain 9 (MYH9) protein through the ubiquitin-proteasome pathway via the polycomb repressive complex 1 (PRC1) and suppresses the stem-like phenotype of glioblastoma cells by inhibiting the nuclear factor kappa-B (NF-κB) signaling pathway. Furthermore, overexpression of MYH9 in glioblastoma cells reverses the inhibitory effects of CBX7 on migration, proliferation, invasion, and stemness of glioblastoma cells. In summary, CBX7 acts as a tumor suppressor by inhibiting the stem cell-like characteristics of glioblastoma. The CBX7-MYH9-NF-κB signaling axis may serve as a potential therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Kaixiang Ni
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - Yuankun Liu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - Pinggang DI
- Department of Emergency, Wulian County People's Hospital, 262300, Rizhao, Shandong, China
| | - Lu Wang
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - Hui Huang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China
| | - R M Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2050, Australia.
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Karrie Mei-Yee Kiang
- Division of Neurosurgery, Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Jiantong Jiao
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China.
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 214023, Wuxi, Jiangsu, China.
| |
Collapse
|
5
|
Otterpohl KL, Busselman BW, Zimmerman JL, Mukherjee M, Evans C, Graber K, Thakkar VP, Johnston JG, Ilyas A, Gumz ML, Eaton DC, Sands JM, Surendran K, Chandrasekar I. Thick Ascending Limb Specific Inactivation of Myh9 and Myh10 Myosin Motors Results in Progressive Kidney Disease and Drives Sex-specific Cellular Adaptation in the Distal Nephron and Collecting Duct. FUNCTION 2025; 6:zqae048. [PMID: 39500539 PMCID: PMC11815580 DOI: 10.1093/function/zqae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
Our previous work established a role for myosin motor proteins MYH9 and MYH10 in trafficking of thick ascending limb (TAL) cargoes uromodulin and Na+-K+-2Cl- cotransporter NKCC2. We have generated a TAL-specific Myh9&10 conditional knockout (Myh9&10 TAL-cKO) mouse model to determine the cell autonomous roles for MYH9&10 in TAL cargo trafficking and to understand the consequence of TAL dysfunction in adult kidney. Myh9&10 TAL-cKO mice develop progressive kidney disease with pathological tubular injury confirmed by histological changes, tubular injury markers, upregulated endoplasmic reticulum (ER) stress/unfolded protein response, and higher blood urea nitrogen and serum creatinine. However, male mice survive twice as long as female mice. We have determined this sexual dimorphism in morbidity is due to adaptation of the distal nephron and collecting duct in response to TAL dysfunction and lower NKCC2 expression. We demonstrate that this triggers a compensatory mechanism involving sex-specific cellular adaptation within the distal nephron and collecting duct to boost sodium reabsorption. While both sexes overcompensate by activating epithelial sodium channel (ENaC) expression in medullary collecting ducts resulting in hypernatremia, this is initially subdued in male Myh9&10 TAL-cKO mice through higher sodium chloride cotransporter (NCC) expression within the distal nephron. Our results indicate that compromised TAL function ultimately results in maladaptation of medullary collecting duct cells which acquire cortical-like properties including ENaC expression. This work further confirms a cell autonomous role for MYH9&10 in maintenance of NKCC2 expression in the TAL and uncover distal nephron and collecting duct adaptive mechanisms which respond to TAL dysfunction.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, SD 57069, USA
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Malini Mukherjee
- Functional Genomics and Bioinformatics Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Claire Evans
- Histology and Imaging Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Kelly Graber
- Histology and Imaging Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Jermaine G Johnston
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, SD 57069, USA
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Douglas C Eaton
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA
| | - Jeff M Sands
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, SD 57103, USA
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, SD 57103, USA
| |
Collapse
|
6
|
Heissler SM, Chinthalapudi K. Structural and functional mechanisms of actin isoforms. FEBS J 2025; 292:468-482. [PMID: 38779987 PMCID: PMC11796330 DOI: 10.1111/febs.17153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/01/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Actin is a highly conserved and fundamental protein in eukaryotes and participates in a broad spectrum of cellular functions. Cells maintain a conserved ratio of actin isoforms, with muscle and non-muscle actins representing the main actin isoforms in muscle and non-muscle cells, respectively. Actin isoforms have specific and redundant functional roles and display different biochemistries, cellular localization, and interactions with myosins and actin-binding proteins. Understanding the specific roles of actin isoforms from the structural and functional perspective is crucial for elucidating the intricacies of cytoskeletal dynamics and regulation and their implications in health and disease. Here, we review how the structure contributes to the functional mechanisms of actin isoforms with a special emphasis on the questions of how post-translational modifications and disease-linked mutations affect actin isoforms biochemistry, function, and interaction with actin-binding proteins and myosin motors.
Collapse
Affiliation(s)
- Sarah M. Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research InstituteThe Ohio State UniversityColumbusOHUSA
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research InstituteThe Ohio State UniversityColumbusOHUSA
| |
Collapse
|
7
|
Zheng S, Hong Z, Tan Y, Wang Y, Li J, Zhang Z, Feng T, Hong Z, Lin G, Ye D. MYO6 contributes to tumor progression and enzalutamide resistance in castration-resistant prostate cancer by activating the focal adhesion signaling pathway. Cell Commun Signal 2024; 22:517. [PMID: 39449086 PMCID: PMC11515482 DOI: 10.1186/s12964-024-01897-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Enzalutamide (Enz) resistance is a poor prognostic factor for patients with castration-resistant prostate cancer (CRPC), which often involves aberrant expression of the androgen receptor (AR). Myosin VI (MYO6), one member of the myosin family, plays an important role in regulating cell survival and is highly expressed in prostate cancer (PCa). However, whether MYO6 is involved in Enz resistance in CRPC and its mechanism remain unclear. METHODS Multiple open-access databases were utilized to examine the relationship between MYO6 expression and PCa progression, and to screen differentially expressed genes (DEGs) and potential signaling pathways associated with the MYO6-regulated Enz resistance. Both in vitro and in vivo tumorigenesis assays were employed to examine the impact of MYO6 on the growth and Enz resistance of PCa cells. Human PCa tissues and related clinical biochemical data were utilized to identify the role of MYO6 in promoting PCa progression and Enz resistance. The molecular mechanisms underlying the regulation of gene expression, PCa progression, and Enz resistance in CRPC by MYO6 were investigated. RESULTS MYO6 expression increases in patients with PCa and is positively correlated with AR expression in PCa cell lines and tissues. Overexpression of AR increases MYO6 expression to promote PCa cell proliferation, migration and invasion, and to inhibit PCa cell apoptosis; whereas knockdown of MYO6 expression reverses these outcomes and enhances Enz function in suppressing the proliferation of the Enz- sensitive and resistant PCa cells both in vitro and in vivo. Mechanistically, AR binds directly to the promoter region (residues - 503 to - 283 base pairs) of MYO6 gene and promotes its transcription. Furthermore, MYO6 activates focal adhesion kinase (FAK) phosphorylation at tyrosine-397 through integrin beta 8 (ITGB8) modulation to promote PCa progression and Enz resistance. Notably, inhibition of FAK activity by Y15, an inhibitor of FAK, can resensitize CRPC cells to Enz treatment in cell lines and mouse xenograft models. CONCLUSIONS MYO6 has pro-tumor and Enz-resistant effects in CRPC, suggesting that targeting MYO6 may be beneficial for ENZ-resistant CRPC therapy through the AR/MYO6/FAK signaling pathway.
Collapse
MESH Headings
- Humans
- Male
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Benzamides/pharmacology
- Phenylthiohydantoin/pharmacology
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Signal Transduction/drug effects
- Animals
- Nitriles/pharmacology
- Cell Line, Tumor
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Disease Progression
- Focal Adhesions/drug effects
- Focal Adhesions/metabolism
- Mice
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Proliferation/drug effects
- Mice, Nude
- Cell Movement/drug effects
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
Collapse
Affiliation(s)
- Shengfeng Zheng
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Qingdao Institute, School of Life Medicine, Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Qingdao, China
| | - Zhe Hong
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Yao Tan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Nursing Administration, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Junhong Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Zihao Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Tao Feng
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Zongyuan Hong
- Department of Pharmacology and Laboratory of Quantitative Pharmacology, Wannan Medical College, Wuhu, Anhui, 241000, China.
| | - Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
8
|
Hart RG, Kota D, Li F, Zhang M, Ramallo D, Price AJ, Otterpohl KL, Smith SJ, Dunn AR, Huising MO, Liu J, Chandrasekar I. Myosin II tension sensors visualize force generation within the actin cytoskeleton in living cells. J Cell Sci 2024; 137:jcs262281. [PMID: 39369303 PMCID: PMC11698044 DOI: 10.1242/jcs.262281] [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: 05/20/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Nonmuscle myosin II (NMII) generates cytoskeletal forces that drive cell division, embryogenesis, muscle contraction and many other cellular functions. However, at present there is no method that can directly measure the forces generated by myosins in living cells. Here, we describe a Förster resonance energy transfer (FRET)-based tension sensor that can detect myosin-associated force along the filamentous actin network. Fluorescence lifetime imaging microscopy (FLIM)-FRET measurements indicate that the forces generated by NMII isoform B (NMIIB) exhibit significant spatial and temporal heterogeneity as a function of donor lifetime and fluorophore energy exchange. These measurements provide a proxy for inferred forces that vary widely along the actin cytoskeleton. This initial report highlights the potential utility of myosin-based tension sensors in elucidating the roles of cytoskeletal contractility in a wide variety of contexts.
Collapse
Affiliation(s)
- Ryan G. Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Divya Kota
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Fangjia Li
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Mengdi Zhang
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Diego Ramallo
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Andrew J. Price
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Karla L. Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Steve J. Smith
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Alexander R. Dunn
- Department of Chemical Biology, Stanford University, Stanford, CA 94305, USA
| | - Mark O. Huising
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jing Liu
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN 46907
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
9
|
Childers MC, Regnier M. Dynamics of the Pre-Powerstroke Myosin Lever Arm and the Effects of Omecamtiv Mecarbil. Int J Mol Sci 2024; 25:10425. [PMID: 39408754 PMCID: PMC11477208 DOI: 10.3390/ijms251910425] [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: 09/06/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
The binding of small molecules to sarcomeric myosin can elicit powerful effects on the chemomechanical cycle, making them effective therapeutics in the clinic and research tools at the benchtop. However, these myotropes can have complex effects that act on different phases of the crossbridge cycle and which depend on structural, dynamic, and environmental variables. While small molecule binding sites have been identified crystallographically and their effects on contraction studied extensively, small molecule-induced dynamic changes that link structure-function are less studied. Here, we use molecular dynamics simulations to explore how omecamtiv mecarbil (OM), a cardiac myosin-specific myotrope, alters the coordinated dynamics of the lever arm and the motor domain in the pre-powerstroke state. We show that the lever arm adopts a range of orientations and find that different lever arm orientations are accompanied by changes in the hydrogen bonding patterns near the converter. We find that the binding of OM to myosin reduces the conformational heterogeneity of the lever arm orientation and also adjusts the average lever arm orientation. Finally, we map out the distinct conformations and ligand-protein interactions adopted by OM. These results uncover some structural factors that govern the motor domain-tail orientations and the mechanisms by which OM primes the pre-powerstroke myosin heads.
Collapse
Affiliation(s)
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
10
|
Lv HB, Wu QY, Zhang YJ, Quan SW, Ma N, Dai YQ, Sun Y. Study on the expression and prognostic relationship of MYL6B in liver cancer based on bioinformatics. World J Clin Oncol 2024; 15:1188-1197. [PMID: 39351463 PMCID: PMC11438851 DOI: 10.5306/wjco.v15.i9.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/21/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Primary liver cancer is a prevalent and deadly cancer type. Despite treatment advances, prognosis remains poor, with high recurrence rates. Early detection is crucial but challenging due to the disease's insidious nature. Myosin proteins play significant roles in cancer development, influencing cell migration, invasion, and tumor suppression. MYL6B, a myosin light chain, is involved in various cellular processes and has been associated with poor prognosis in colorectal adenocarcinoma and potential as a biomarker in breast cancer. AIM To investigate the expression of MYL6B in liver hepatocellular carcinoma (LIHC) and its impact on prognosis and potential mechanisms of action using bioinformatics methods. METHODS The expression of MYL6B in pan-cancer and normal tissues was analyzed using the gene expression profiling interactive analysis 2 and tumor immune estimation resource databases. The expression level of MYL6B in LIHC tissues and its relationship with prognosis were analyzed, immunohistochemical analysis of MYL6B and its effect on immune cell infiltration, and the protein network were further studied. RESULTS MYL6B was highly expressed in diffuse large b-cell lymphoma, LIHC, pancreatic adenocarcinoma, skin cutaneous melanoma, thymoma, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and lowly expressed in kidney chromophobe, acute myeloid leukemia, testicular germ cell tumors. The expression level of MYL6B was significantly different between cancer and normal tissues. It had a significant impact on both overall survival and disease-free survival. MYL6B is highly expressed in hepatocellular carcinoma and its expression level increases with cancer progression. High MYL6B expression is associated with poor prognosis in terms of overall survival and recurrence-free survival. The immunohistochemical level of MYL6B is high in hepatocellular carcinoma tissues, and MYL6B has a high level of immune infiltration inflammation. In protein network analysis, MYL6B is correlated with MYL2, MYL6, MYL9, MYLK4, MYLK2, MYL12A, MYL12B, MYH11, MYH9 and MYH10. CONCLUSION The expression level of MYL6B in LIHC was significantly higher than in normal liver tissues, and it was correlated with the degree of differentiation survival rate, and immune infiltration. MYL6B is a potential target for LIHC treatment.
Collapse
Affiliation(s)
- Hai-Bing Lv
- Department of General Surgery, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Qing-Yun Wu
- Department of General Surgery, Xianning Central Hospital, Xianning 437000, Hubei Province, China
| | - Yu-Jiao Zhang
- Department of Medical oncology, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Sheng-Wei Quan
- Department of General Surgery, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Ning Ma
- Department of General Surgery, Daqing Oilfield General Hospital, Daqing 163000, Heilongjiang Province, China
| | - Yu-Qing Dai
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Yan Sun
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| |
Collapse
|
11
|
Yamazaki R, Ohno N. Myosin superfamily members during myelin formation and regeneration. J Neurochem 2024; 168:2264-2274. [PMID: 39136255 DOI: 10.1111/jnc.16202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/10/2024] [Accepted: 07/31/2024] [Indexed: 10/04/2024]
Abstract
Myelin is an insulator that forms around axons that enhance the conduction velocity of nerve fibers. Oligodendrocytes dramatically change cell morphology to produce myelin throughout the central nervous system (CNS). Cytoskeletal alterations are critical for the morphogenesis of oligodendrocytes, and actin is involved in cell differentiation and myelin wrapping via polymerization and depolymerization, respectively. Various protein members of the myosin superfamily are known to be major binding partners of actin filaments and have been intensively researched because of their involvement in various cellular functions, including differentiation, cell movement, membrane trafficking, organelle transport, signal transduction, and morphogenesis. Some members of the myosin superfamily have been found to play important roles in the differentiation of oligodendrocytes and in CNS myelination. Interestingly, each member of the myosin superfamily expressed in oligodendrocyte lineage cells also shows specific spatial and temporal expression patterns and different distributions. In this review, we summarize previous findings related to the myosin superfamily and discuss how these molecules contribute to myelin formation and regeneration by oligodendrocytes.
Collapse
Affiliation(s)
- Reiji Yamazaki
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
12
|
Feroz W, Park BS, Siripurapu M, Ntim N, Kilroy MK, Sheikh AMA, Mishra R, Garrett JT. Non-Muscle Myosin II A: Friend or Foe in Cancer? Int J Mol Sci 2024; 25:9435. [PMID: 39273383 PMCID: PMC11395477 DOI: 10.3390/ijms25179435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Non-muscle myosin IIA (NM IIA) is a motor protein that belongs to the myosin II family. The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of NM IIA. NM IIA is a hexamer and contains three pairs of peptides, which include the dimer of heavy chains, essential light chains, and regulatory light chains. NM IIA is a part of the actomyosin complex that generates mechanical force and tension to carry out essential cellular functions, including adhesion, cytokinesis, migration, and the maintenance of cell shape and polarity. These functions are regulated via light and heavy chain phosphorylation at different amino acid residues. Apart from physiological functions, NM IIA is also linked to the development of cancer and genetic and neurological disorders. MYH9 gene mutations result in the development of several autosomal dominant disorders, such as May-Hegglin anomaly (MHA) and Epstein syndrome (EPS). Multiple studies have reported NM IIA as a tumor suppressor in melanoma and head and neck squamous cell carcinoma; however, studies also indicate that NM IIA is a critical player in promoting tumorigenesis, chemoradiotherapy resistance, and stemness. The ROCK-NM IIA pathway regulates cellular movement and shape via the control of cytoskeletal dynamics. In addition, the ROCK-NM IIA pathway is dysregulated in various solid tumors and leukemia. Currently, there are very few compounds targeting NM IIA, and most of these compounds are still being studied in preclinical models. This review provides comprehensive evidence highlighting the dual role of NM IIA in multiple cancer types and summarizes the signaling networks involved in tumorigenesis. Furthermore, we also discuss the role of NM IIA as a potential therapeutic target with a focus on the ROCK-NM IIA pathway.
Collapse
Affiliation(s)
- Wasim Feroz
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Briley SoYoung Park
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
- Cancer Research Scholars Program, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meghna Siripurapu
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Nicole Ntim
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Mary Kate Kilroy
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | | | - Rosalin Mishra
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Joan T. Garrett
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| |
Collapse
|
13
|
Carl AG, Reynolds MJ, Gurel PS, Phua DY, Sun X, Mei L, Hamilton K, Takagi Y, Noble AJ, Sellers JR, Alushin GM. Myosin forces elicit an F-actin structural landscape that mediates mechanosensitive protein recognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608188. [PMID: 39185238 PMCID: PMC11343212 DOI: 10.1101/2024.08.15.608188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Cells mechanically interface with their surroundings through cytoskeleton-linked adhesions, allowing them to sense physical cues that instruct development and drive diseases such as cancer. Contractile forces generated by myosin motor proteins mediate these mechanical signal transduction processes through unclear protein structural mechanisms. Here, we show that myosin forces elicit structural changes in actin filaments (F-actin) that modulate binding by the mechanosensitive adhesion protein α-catenin. Using correlative cryo-fluorescence microscopy and cryo-electron tomography, we identify F-actin featuring domains of nanoscale oscillating curvature at cytoskeleton-adhesion interfaces enriched in zyxin, a marker of actin-myosin generated traction forces. We next introduce a reconstitution system for visualizing F-actin in the presence of myosin forces with cryo-electron microscopy, which reveals morphologically similar superhelical F-actin spirals. In simulations, transient forces mimicking tugging and release of filaments by motors produce spirals, supporting a mechanistic link to myosin's ATPase mechanochemical cycle. Three-dimensional reconstruction of spirals uncovers extensive asymmetric remodeling of F-actin's helical lattice. This is recognized by α-catenin, which cooperatively binds along individual strands, preferentially engaging interfaces featuring extended inter-subunit distances while simultaneously suppressing rotational deviations to regularize the lattice. Collectively, we find that myosin forces can deform F-actin, generating a conformational landscape that is detected and reciprocally modulated by a mechanosensitive protein, providing a direct structural glimpse at active force transduction through the cytoskeleton.
Collapse
Affiliation(s)
- Ayala G. Carl
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
- Tri-Institutional Program in Chemical Biology, The Rockefeller University, New York, NY, USA
| | - Matthew J. Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Pinar S. Gurel
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Donovan Y.Z. Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Lin Mei
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
- Tri-Institutional Program in Chemical Biology, The Rockefeller University, New York, NY, USA
| | - Keith Hamilton
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Yasuharu Takagi
- Laboratory of Molecular Physiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Alex J. Noble
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, USA
| | - James R. Sellers
- Laboratory of Molecular Physiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
14
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. PNAS NEXUS 2024; 3:pgae279. [PMID: 39108304 PMCID: PMC11302452 DOI: 10.1093/pnasnexus/pgae279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (threefold) the maximum steady-state actin-activated ATPase activity (k cat) and decreases (eightfold) the actin concentration at which ATPase is one-half maximal (K ATPase). We also found a twofold to fourfold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio threefold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M L Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - Wen Ma
- Department of Physics, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - David V Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University School of Medicine, 64 Medical Center Dr, Morgantown, WV 26506, USA
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - J Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| |
Collapse
|
15
|
Taft MH, Redowicz MJ. Editorial: Unconventional myosins in motile and contractile functions: fifty years on the stage. Front Physiol 2024; 15:1439746. [PMID: 38933364 PMCID: PMC11199865 DOI: 10.3389/fphys.2024.1439746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Affiliation(s)
- Manuel H. Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Maria Jolanta Redowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
16
|
Horsthemke M, Arnaud CA, Hanley PJ. Are the class 18 myosins Myo18A and Myo18B specialist sarcomeric proteins? Front Physiol 2024; 15:1401717. [PMID: 38784114 PMCID: PMC11112018 DOI: 10.3389/fphys.2024.1401717] [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: 03/15/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Initially, the two members of class 18 myosins, Myo18A and Myo18B, appeared to exhibit highly divergent functions, complicating the assignment of class-specific functions. However, the identification of a striated muscle-specific isoform of Myo18A, Myo18Aγ, suggests that class 18 myosins may have evolved to complement the functions of conventional class 2 myosins in sarcomeres. Indeed, both genes, Myo18a and Myo18b, are predominantly expressed in the heart and somites, precursors of skeletal muscle, of developing mouse embryos. Genetic deletion of either gene in mice is embryonic lethal and is associated with the disorganization of cardiac sarcomeres. Moreover, Myo18Aγ and Myo18B localize to sarcomeric A-bands, albeit the motor (head) domains of these unconventional myosins have been both deduced and biochemically demonstrated to exhibit negligible ATPase activity, a hallmark of motor proteins. Instead, Myo18Aγ and Myo18B presumably coassemble with thick filaments and provide structural integrity and/or internal resistance through interactions with F-actin and/or other proteins. In addition, Myo18Aγ and Myo18B may play distinct roles in the assembly of myofibrils, which may arise from actin stress fibers containing the α-isoform of Myo18A, Myo18Aα. The β-isoform of Myo18A, Myo18Aβ, is similar to Myo18Aα, except that it lacks the N-terminal extension, and may serve as a negative regulator through heterodimerization with either Myo18Aα or Myo18Aγ. In this review, we contend that Myo18Aγ and Myo18B are essential for myofibril structure and function in striated muscle cells, while α- and β-isoforms of Myo18A play diverse roles in nonmuscle cells.
Collapse
Affiliation(s)
- Markus Horsthemke
- IMM Institute for Molecular Medicine, HMU Health and Medical University Potsdam, Potsdam, Germany
| | - Charles-Adrien Arnaud
- IMM Institute for Molecular Medicine, HMU Health and Medical University Potsdam, Potsdam, Germany
- Department of Medicine, Science Faculty, MSB Medical School Berlin, Berlin, Germany
| | - Peter J. Hanley
- IMM Institute for Molecular Medicine, HMU Health and Medical University Potsdam, Potsdam, Germany
| |
Collapse
|
17
|
Behbehani R, Johnson C, Holmes AJ, Gratian MJ, Mulvihill DP, Buss F. The two C. elegans class VI myosins, SPE-15/HUM-3 and HUM-8, share similar motor properties, but have distinct developmental and tissue expression patterns. Front Physiol 2024; 15:1368054. [PMID: 38660538 PMCID: PMC11040104 DOI: 10.3389/fphys.2024.1368054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Myosins of class VI move toward the minus-end of actin filaments and play vital roles in cellular processes such as endocytosis, autophagy, protein secretion, and the regulation of actin filament dynamics. In contrast to the majority of metazoan organisms examined to date which contain a single MYO6 gene, C. elegans, possesses two MYO6 homologues, SPE-15/HUM-3 and HUM-8. Through a combination of in vitro biochemical/biophysical analysis and cellular assays, we confirmed that both SPE-15/HUM-3 and HUM-8 exhibit reverse directionality, velocities, and ATPase activity similar to human MYO6. Our characterization also revealed that unlike SPE-15/HUM-3, HUM-8 is expressed as two distinct splice isoforms, one with an additional unique 14 amino acid insert in the cargo-binding domain. While lipid and adaptor binding sites are conserved in SPE-15/HUM-3 and HUM-8, this conservation does not enable recruitment to endosomes in mammalian cells. Finally, we performed super-resolution confocal imaging on transgenic worms expressing either mNeonGreen SPE-15/HUM-3 or wrmScarlet HUM-8. Our results show a clear distinction in tissue distribution between SPE-15/HUM-3 and HUM-8. While SPE-15/HUM-3 exhibited specific expression in the gonads and neuronal tissue in the head, HUM-8 was exclusively localized in the intestinal epithelium. Overall, these findings align with the established tissue distributions and localizations of human MYO6.
Collapse
Affiliation(s)
- Ranya Behbehani
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Chloe Johnson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Alexander J. Holmes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Matthew J. Gratian
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Miyoshi T, Belyantseva IA, Sajeevadathan M, Friedman TB. Pathophysiology of human hearing loss associated with variants in myosins. Front Physiol 2024; 15:1374901. [PMID: 38562617 PMCID: PMC10982375 DOI: 10.3389/fphys.2024.1374901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 04/04/2024] Open
Abstract
Deleterious variants of more than one hundred genes are associated with hearing loss including MYO3A, MYO6, MYO7A and MYO15A and two conventional myosins MYH9 and MYH14. Variants of MYO7A also manifest as Usher syndrome associated with dysfunction of the retina and vestibule as well as hearing loss. While the functions of MYH9 and MYH14 in the inner ear are debated, MYO3A, MYO6, MYO7A and MYO15A are expressed in inner ear hair cells along with class-I myosin MYO1C and are essential for developing and maintaining functional stereocilia on the apical surface of hair cells. Stereocilia are large, cylindrical, actin-rich protrusions functioning as biological mechanosensors to detect sound, acceleration and posture. The rigidity of stereocilia is sustained by highly crosslinked unidirectionally-oriented F-actin, which also provides a scaffold for various proteins including unconventional myosins and their cargo. Typical myosin molecules consist of an ATPase head motor domain to transmit forces to F-actin, a neck containing IQ-motifs that bind regulatory light chains and a tail region with motifs recognizing partners. Instead of long coiled-coil domains characterizing conventional myosins, the tails of unconventional myosins have various motifs to anchor or transport proteins and phospholipids along the F-actin core of a stereocilium. For these myosins, decades of studies have elucidated their biochemical properties, interacting partners in hair cells and variants associated with hearing loss. However, less is known about how myosins traffic in a stereocilium using their motor function, and how each variant correlates with a clinical condition including the severity and onset of hearing loss, mode of inheritance and presence of symptoms other than hearing loss. Here, we cover the domain structures and functions of myosins associated with hearing loss together with advances, open questions about trafficking of myosins in stereocilia and correlations between hundreds of variants in myosins annotated in ClinVar and the corresponding deafness phenotypes.
Collapse
Affiliation(s)
- Takushi Miyoshi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
- Division of Molecular and Integrative Physiology, Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Inna A. Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Mrudhula Sajeevadathan
- Division of Molecular and Integrative Physiology, Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Thomas B. Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
19
|
Greve JN, Marquardt A, Heiringhoff R, Reindl T, Thiel C, Di Donato N, Taft MH, Manstein DJ. The non-muscle actinopathy-associated mutation E334Q in cytoskeletal γ-actin perturbs interaction of actin filaments with myosin and ADF/cofilin family proteins. eLife 2024; 12:RP93013. [PMID: 38446501 PMCID: PMC10942649 DOI: 10.7554/elife.93013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Various heterozygous cytoskeletal γ-actin mutations have been shown to cause Baraitser-Winter cerebrofrontofacial syndrome, non-syndromic hearing loss, or isolated eye coloboma. Here, we report the biochemical characterization of human cytoskeletal γ-actin carrying mutation E334Q, a mutation that leads to a hitherto unspecified non-muscle actinopathy. Following expression, purification, and removal of linker and thymosin β4 tag sequences, the p.E334Q monomers show normal integration into linear and branched actin filaments. The mutation does not affect thermal stability, actin filament nucleation, elongation, and turnover. Model building and normal mode analysis predict significant differences in the interaction of p.E334Q filaments with myosin motors and members of the ADF/cofilin family of actin-binding proteins. Assays probing the interactions of p.E334Q filaments with human class 2 and class 5 myosin motor constructs show significant reductions in sliding velocity and actin affinity. E334Q differentially affects cofilin-mediated actin dynamics by increasing the rate of cofilin-mediated de novo nucleation of actin filaments and decreasing the efficiency of cofilin-mediated filament severing. Thus, it is likely that p.E334Q-mediated changes in myosin motor activity, as well as filament turnover, contribute to the observed disease phenotype.
Collapse
Affiliation(s)
- Johannes N Greve
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Anja Marquardt
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Robin Heiringhoff
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Theresia Reindl
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Claudia Thiel
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | | | - Manuel H Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Fritz Hartmann Centre for MedicalHannoverGermany
- Division for Structural Biochemistry, Hannover Medical SchoolHannoverGermany
- RESiST, Cluster of Excellence 2155, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
20
|
He Q, Wu C, Sun D, Yuan J, Hu H, Yang K, Chen W, Yan Y, Yin G, Zhang J, Li Y. Functional assessment of a novel biallelic MYH3 variation causing CPSKF1B (contractures, pterygia, and spondylocarpotarsal fusion syndrome1B). Mol Genet Genomic Med 2024; 12:e2401. [PMID: 38444278 PMCID: PMC10915484 DOI: 10.1002/mgg3.2401] [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: 08/07/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND The MYH3-associated myosinopathies comprise a spectrum of rare neuromuscular disorders mainly characterized by distal arthrogryposis with or without other features like pterygia and vertebrae fusion. CPSKF1B (contractures, pterygia, and spondylocarpotarsal fusion syndrome1B) is the only known autosomal recessiveMYH3-associated myosinopathy so far, with no more than two dozen cases being reported. MATERIALS AND METHODS A boy with CPSKF1B was recruited and subjected to a comprehensive clinical and imaging evaluation. Genetic detection with whole-exome sequencing (WES) was performed on the patient and extended family members to identify the causative variation. A series of in silico and in vitro investigations were carried out to verify the pathogenicity of the two variants of the identified compound heterozygous variation. RESULTS The patient exhibited moderate CPSKF1B symptoms including multiarticular contractures, webbed neck, and spondylocarpotarsal fusion. WES detected a compound heterozygous MYH3 variation consisting of two variants, namely NM_002470.4: c.3377A>G; p. (E1126G) and NM_002470.4: c.5161-2A>C. It was indicated that the NM_002470.4: c.3377A>G; p. (E1126G) variant mainly impaired the local hydrogen bond formation and impacted the TGF-B pathway, while the NM_002470.4: c.5161-2A>C variant could affect the normal splicing of pre-mRNA, resulting in the appearance of multiple abnormal transcripts. CONCLUSIONS The findings of this study expanded the mutation spectrum of CPSKF1B, provided an important basis for the counseling of the affected family, and also laid a foundation for the functional study of MYH3 mutations.
Collapse
Affiliation(s)
- Qing‐bing He
- Department of Pediatric OrthopaedicsThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Cai‐hong Wu
- Department of Clinical LaboratoryHebei Petrochina Central HospitalLangfangChina
| | - Dong‐lan Sun
- Prenatal Diagnosis CenterShijiazhuang Obstetrics and Gynecology HospitalShijiazhuangChina
- Hebei Key Laboratory of Maternal and Fetal Medicine; Shijiazhuang Key Laboratory of Reproductive HealthShijiazhuangChina
| | - Jia‐yu Yuan
- Department of Pediatric OrthopaedicsThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Hua‐ying Hu
- Birth Defects Prevention and Control Technology Research CenterMedical Innovation Research Division of Chinese PLA General HospitalBeijingChina
| | - Kai Yang
- Prenatal Diagnosis Center, Beijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijingChina
- Beijing Maternal and Child Health Care HospitalCapital Medical UniversityBeijingChina
| | - Wen‐qi Chen
- Prenatal Diagnosis CenterShijiazhuang Obstetrics and Gynecology HospitalShijiazhuangChina
- Hebei Key Laboratory of Maternal and Fetal Medicine; Shijiazhuang Key Laboratory of Reproductive HealthShijiazhuangChina
| | - You‐sheng Yan
- Prenatal Diagnosis Center, Beijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijingChina
- Beijing Maternal and Child Health Care HospitalCapital Medical UniversityBeijingChina
| | - Guang‐yue Yin
- Department of Clinical LaboratoryHebei Petrochina Central HospitalLangfangChina
| | - Jing Zhang
- Prenatal Diagnosis CenterShijiazhuang Obstetrics and Gynecology HospitalShijiazhuangChina
- Hebei Key Laboratory of Maternal and Fetal Medicine; Shijiazhuang Key Laboratory of Reproductive HealthShijiazhuangChina
| | - Ya‐zhou Li
- Department of Pediatric OrthopaedicsThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
21
|
Bedada FB, Thompson BR, Mikkila JL, Chan SSK, Choi SH, Toso EA, Kyba M, Metzger JM. Inducing positive inotropy in human iPSC-derived cardiac muscle by gene editing-based activation of the cardiac α-myosin heavy chain. Sci Rep 2024; 14:3915. [PMID: 38365813 PMCID: PMC10873390 DOI: 10.1038/s41598-024-53395-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/31/2024] [Indexed: 02/18/2024] Open
Abstract
Human induced pluripotent stem cells and their differentiation into cardiac myocytes (hiPSC-CMs) provides a unique and valuable platform for studies of cardiac muscle structure-function. This includes studies centered on disease etiology, drug development, and for potential clinical applications in heart regeneration/repair. Ultimately, for these applications to achieve success, a thorough assessment and physiological advancement of the structure and function of hiPSC-CMs is required. HiPSC-CMs are well noted for their immature and sub-physiological cardiac muscle state, and this represents a major hurdle for the field. To address this roadblock, we have developed a hiPSC-CMs (β-MHC dominant) experimental platform focused on directed physiological enhancement of the sarcomere, the functional unit of cardiac muscle. We focus here on the myosin heavy chain (MyHC) protein isoform profile, the molecular motor of the heart, which is essential to cardiac physiological performance. We hypothesized that inducing increased expression of α-MyHC in β-MyHC dominant hiPSC-CMs would enhance contractile performance of hiPSC-CMs. To test this hypothesis, we used gene editing with an inducible α-MyHC expression cassette into isogeneic hiPSC-CMs, and separately by gene transfer, and then investigated the direct effects of increased α-MyHC expression on hiPSC-CMs contractility and relaxation function. Data show improved cardiac functional parameters in hiPSC-CMs induced with α-MyHC. Positive inotropy and relaxation was evident in comparison to β-MyHC dominant isogenic controls both at baseline and during pacing induced stress. This approach should facilitate studies of hiPSC-CMs disease modeling and drug screening, as well as advancing fundamental aspects of cardiac function parameters for the optimization of future cardiac regeneration, repair and re-muscularization applications.
Collapse
Affiliation(s)
- Fikru B Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
- Present Address: Department of Clinical Laboratory Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, USA
| | - Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Jennifer L Mikkila
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Sunny S-K Chan
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Si Ho Choi
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Erik A Toso
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
22
|
Niu F, Li L, Wang L, Xiao J, Xu S, Liu Y, Lin L, Yu C, Wei Z. Autoinhibition and activation of myosin VI revealed by its cryo-EM structure. Nat Commun 2024; 15:1187. [PMID: 38331992 PMCID: PMC10853514 DOI: 10.1038/s41467-024-45424-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Myosin VI is the only molecular motor that moves towards the minus end along actin filaments. Numerous cellular processes require myosin VI and tight regulations of the motor's activity. Defects in myosin VI activity are known to cause genetic diseases such as deafness and cardiomyopathy. However, the molecular mechanisms underlying the activity regulation of myosin VI remain elusive. Here, we determined the high-resolution cryo-electron microscopic structure of myosin VI in its autoinhibited state. Our structure reveals that autoinhibited myosin VI adopts a compact, monomeric conformation via extensive interactions between the head and tail domains, orchestrated by an elongated single-α-helix region resembling a "spine". This autoinhibited structure effectively blocks cargo binding sites and represses the motor's ATPase activity. Certain cargo adaptors such as GIPC can release multiple inhibitory interactions and promote motor activity, pointing to a cargo-mediated activation of the processive motor. Moreover, our structural findings allow rationalization of disease-associated mutations in myosin VI. Beyond the activity regulation mechanisms of myosin VI, our study also sheds lights on how activities of other myosin motors such as myosin VII and X might be regulated.
Collapse
Affiliation(s)
- Fengfeng Niu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
| | - Lingxuan Li
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lei Wang
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinman Xiao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Shun Xu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yong Liu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Leishu Lin
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Zhiyi Wei
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
23
|
Lee E, May H, Kazmierczak K, Liang J, Nguyen N, Hill JA, Gillette TG, Szczesna-Cordary D, Chang AN. The MYPT2-regulated striated muscle-specific myosin light chain phosphatase limits cardiac myosin phosphorylation in vivo. J Biol Chem 2024; 300:105652. [PMID: 38224947 PMCID: PMC10851227 DOI: 10.1016/j.jbc.2024.105652] [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: 09/10/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
The physiological importance of cardiac myosin regulatory light chain (RLC) phosphorylation by its dedicated cardiac myosin light chain kinase has been established in both humans and mice. Constitutive RLC-phosphorylation, regulated by the balanced activities of cardiac myosin light chain kinase and myosin light chain phosphatase (MLCP), is fundamental to the biochemical and physiological properties of myofilaments. However, limited information is available on cardiac MLCP. In this study, we hypothesized that the striated muscle-specific MLCP regulatory subunit, MYPT2, targets the phosphatase catalytic subunit to cardiac myosin, contributing to the maintenance of cardiac function in vivo through the regulation of RLC-phosphorylation. To test this hypothesis, we generated a floxed-PPP1R12B mouse model crossed with a cardiac-specific Mer-Cre-Mer to conditionally ablate MYPT2 in adult cardiomyocytes. Immunofluorescence microscopy using the gene-ablated tissue as a control confirmed the localization of MYPT2 to regions where it overlaps with a subset of RLC. Biochemical analysis revealed an increase in RLC-phosphorylation in vivo. The loss of MYPT2 demonstrated significant protection against pressure overload-induced hypertrophy, as evidenced by heart weight, qPCR of hypertrophy-associated genes, measurements of myocyte diameters, and expression of β-MHC protein. Furthermore, mantATP chase assays revealed an increased ratio of myosin heads distributed to the interfilament space in MYPT2-ablated heart muscle fibers, confirming that RLC-phosphorylation regulated by MLCP, enhances cardiac performance in vivo. Our findings establish MYPT2 as the regulatory subunit of cardiac MLCP, distinct from the ubiquitously expressed canonical smooth muscle MLCP. Targeting MYPT2 to increase cardiac RLC-phosphorylation in vivo may improve baseline cardiac performance, thereby attenuating pathological hypertrophy.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Herman May
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nhu Nguyen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A Hill
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G Gillette
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Audrey N Chang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Pak Center for Mineral Metabolism and Clinical Research, UTSW Medical Center, Dallas, Texas, USA.
| |
Collapse
|
24
|
Liu X, Shu S. Suggesting Dictyostelium as a Model for Disease-Related Protein Studies through Myosin II Polymerization Pathway. Cells 2024; 13:263. [PMID: 38334655 PMCID: PMC10854627 DOI: 10.3390/cells13030263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/10/2024] Open
Abstract
Dictyostelium myosin II displays remarkable dynamism within the cell, continually undergoing polymerization and depolymerization processes. Under low-ion conditions, it assumes a folded structure like muscle myosins and forms thick filaments through polymerization. In our study, we presented intermediate structures observed during the early stages of polymerization of purified myosin via negative staining electron microscopy, immediately crosslinked with glutaraldehyde at the onset of polymerization. We identified folded monomers, dimers, and tetramers in the process. Our findings suggest that Dictyostelium myosin II follows a polymerization pathway in vitro akin to muscle myosin, with folded monomers forming folded parallel and antiparallel dimers that subsequently associate to create folded tetramers. These folded tetramers eventually unfold and associate with other tetramers to produce long filaments. Furthermore, our research revealed that ATP influences filament size, reducing it regardless of the status of RLC phosphorylation while significantly increasing the critical polymerization concentrations from 0.2 to 9 nM. In addition, we demonstrate the morphology of fully matured Dictyostelium myosin II filaments.
Collapse
|
25
|
Shirai Y, Miura K, Hamada R, Ishikura K, Kunishima S, Hattori M. A nationwide survey of MYH9-related disease in Japan. Clin Exp Nephrol 2024; 28:40-49. [PMID: 37733142 DOI: 10.1007/s10157-023-02404-3] [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: 05/03/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND MYH9-related disease (MYH9-RD) is characterized by congenital macrothrombocytopenia, Döhle body-like granulocyte inclusions, and nephropathy, which may progress to end-stage kidney disease (ESKD). However, information on the effects of renin-angiotensin system (RAS) inhibitors on kidney survival is currently lacking and the outcomes of kidney replacement therapy (KRT) in MYH9-RD are largely unknown. METHODS We conducted a cross-sectional nationwide survey by sending questionnaires to 145 institutions in Japan and analyzed data for 49 patients. RESULTS The median patient age was 27 years. Genetic analysis was performed in 37 (76%) patients. Twenty-four patients (65%) had MYH9 variants affecting the motor domain of non-muscle myosin heavy chain-IIA, and these patients had poorer kidney survival than those with variants affecting the tail domain (P = 0.02). There was no significant difference in kidney survival between patients treated with and without RAS inhibitors. Hemodialysis and peritoneal dialysis were performed in 16 and 7 patients, respectively. There were no major bleeding complications during the perioperative period or during follow-up, except for one patient. Most of the 11 patients who underwent kidney transplantation required perioperative red cell concentrate transfusions, but there was no graft loss during the median posttransplant observational period of 2.0 (interquartile range, 1.3-6.8) years. CONCLUSION Our study demonstrated no beneficial effect of RAS inhibitors on kidney function in patients with MYH9-RD, indicating the need for further studies with more patients. All modalities of KRT are feasible options for MYH9-RD patients who progress to ESKD, with adequate attention to bleeding complications.
Collapse
Affiliation(s)
- Yoko Shirai
- Department of Pediatric Nephrology, Tokyo Women's Medical University, 8-1, Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women's Medical University, 8-1, Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Riku Hamada
- Department of Nephrology and Rheumatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Kenji Ishikura
- Department of Pediatrics, Kitasato University School of Medicine, Kanagawa, Japan
| | - Shinji Kunishima
- School of Health Science, Gifu University of Medical Science, Seki, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women's Medical University, 8-1, Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan.
| |
Collapse
|
26
|
Clements R, Smith T, Cowart L, Zhumi J, Sherrod A, Cahill A, Hunter GL. Myosin XV is a negative regulator of signaling filopodia during long-range lateral inhibition. Dev Biol 2024; 505:110-121. [PMID: 37956923 PMCID: PMC10767839 DOI: 10.1016/j.ydbio.2023.11.002] [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: 08/17/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/20/2023]
Abstract
The self-organization of cells during development is essential for the formation of healthy tissues and requires the coordination of cell activities at local scales. Cytonemes, or signaling filopodia, are dynamic actin-based cellular protrusions that allow cells to engage in contact mediated signaling at a distance. While signaling filopodia have been shown to support several signaling paradigms during development, less is understood about how these protrusions are regulated. We investigated the role of the plus-end directed, unconventional MyTH4-FERM myosins in regulating signaling filopodia during sensory bristle patterning on the dorsal thorax of the fruit fly Drosophila melanogaster. We found that Myosin XV is required for regulating signaling filopodia dynamics and, as a consequence, lateral inhibition more broadly throughout the patterning epithelium. We found that Myosin XV is required for limiting the length and number of signaling filopodia generated by bristle precursor cells. Cells with additional and longer signaling filopodia due to loss of Myosin XV are not signaling competent, due to altered levels of Delta ligand and Notch receptor along their lengths. We conclude that Myosin XV acts to negatively regulate signaling filopodia, as well as promote the ability of signaling filopodia to engage in long-range Notch signaling. Since Myosin XV isoforms are present across several vertebrate and invertebrate systems, this may have significance for other long-range signaling mechanisms.
Collapse
Affiliation(s)
- Rhiannon Clements
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States
| | - Tyler Smith
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States
| | - Luke Cowart
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States
| | - Jennifer Zhumi
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States
| | - Alan Sherrod
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States
| | - Aidan Cahill
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States
| | - Ginger L Hunter
- Department of Biology, Clarkson University, Potsdam, NY, 13699, United States.
| |
Collapse
|
27
|
Endo T. Postnatal skeletal muscle myogenesis governed by signal transduction networks: MAPKs and PI3K-Akt control multiple steps. Biochem Biophys Res Commun 2023; 682:223-243. [PMID: 37826946 DOI: 10.1016/j.bbrc.2023.09.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Skeletal muscle myogenesis represents one of the most intensively and extensively examined systems of cell differentiation, tissue formation, and regeneration. Muscle regeneration provides an in vivo model system of postnatal myogenesis. It comprises multiple steps including muscle stem cell (or satellite cell) quiescence, activation, migration, myogenic determination, myoblast proliferation, myocyte differentiation, myofiber maturation, and hypertrophy. A variety of extracellular signaling and subsequent intracellular signal transduction pathways or networks govern the individual steps of postnatal myogenesis. Among them, MAPK pathways (the ERK, JNK, p38 MAPK, and ERK5 pathways) and PI3K-Akt signaling regulate multiple steps of myogenesis. Ca2+, cytokine, and Wnt signaling also participate in several myogenesis steps. These signaling pathways often control cell cycle regulatory proteins or the muscle-specific MyoD family and the MEF2 family of transcription factors. This article comprehensively reviews molecular mechanisms of the individual steps of postnatal skeletal muscle myogenesis by focusing on signal transduction pathways or networks. Nevertheless, no or only a partial signaling molecules or pathways have been identified in some responses during myogenesis. The elucidation of these unidentified signaling molecules and pathways leads to an extensive understanding of the molecular mechanisms of myogenesis.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
28
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566646. [PMID: 38014187 PMCID: PMC10680644 DOI: 10.1101/2023.11.10.566646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (3-fold) the maximum steady-state actin-activated ATPase activity (kcat) and decreases (6-fold) the actin concentration at which ATPase is one-half maximal (KATPase). We also found a 3 to 4-fold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio 3-fold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt-bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt-bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M. L. Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Wen Ma
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - David V. Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
29
|
Wollscheid HP, Ulrich HD. Chromatin meets the cytoskeleton: the importance of nuclear actin dynamics and associated motors for genome stability. DNA Repair (Amst) 2023; 131:103571. [PMID: 37738698 DOI: 10.1016/j.dnarep.2023.103571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
The actin cytoskeleton is of fundamental importance for numerous cellular processes, including intracellular transport, cell plasticity, and cell migration. However, functions of filamentous actin (F-actin) in the nucleus remain understudied due to the comparatively low abundance of nuclear actin and the resulting experimental limitations to its visualization. Owing to recent technological advances such as super-resolution microscopy and the development of nuclear-specific actin probes, essential roles of the actin cytoskeleton in the context of genome maintenance are now emerging. In addition to the contributions of monomeric actin as a component of multiple important nuclear protein complexes, nuclear actin has been found to undergo polymerization in response to DNA damage and DNA replication stress. Consequently, nuclear F-actin plays important roles in the regulation of intra-nuclear mobility of repair and replication foci as well as the maintenance of nuclear shape, two important aspects of efficient stress tolerance. Beyond actin itself, there is accumulating evidence for the participation of multiple actin-binding proteins (ABPs) in the surveillance of genome integrity, including nucleation factors and motor proteins of the myosin family. Here we summarize recent findings highlighting the importance of actin cytoskeletal factors within the nucleus in key genome maintenance pathways.
Collapse
Affiliation(s)
- Hans-Peter Wollscheid
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, Mainz D - 55128, Germany.
| | - Helle D Ulrich
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, Mainz D - 55128, Germany.
| |
Collapse
|
30
|
Zhang Q, Feng P, Zhu XH, Zhou SQ, Ye ML, Yang XJ, Gong S, Huang SY, Tan XR, He SW, Li YQ. DNAJA4 suppresses epithelial-mesenchymal transition and metastasis in nasopharyngeal carcinoma via PSMD2-mediated MYH9 degradation. Cell Death Dis 2023; 14:697. [PMID: 37875476 PMCID: PMC10598267 DOI: 10.1038/s41419-023-06225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023]
Abstract
Emerging evidence indicates that DNA methylation plays an important role in the initiation and progression of nasopharyngeal carcinoma (NPC). DNAJA4 is hypermethylated in NPC, while its role in regulating NPC progression remains unclear. Here, we revealed that the promoter of DNAJA4 was hypermethylated and its expression was downregulated in NPC tissues and cells. Overexpression of DNAJA4 significantly suppressed NPC cell migration, invasion, and EMT in vitro, and markedly inhibited the inguinal lymph node metastasis and lung metastatic colonization in vivo, while it did not affect NPC cell viability and proliferation capability. Mechanistically, DNAJA4 facilitated MYH9 protein degradation via the ubiquitin-proteasome pathway by recruiting PSMD2. Furthermore, the suppressive effects of DNAJA4 on NPC cell migration, invasion, and EMT were reversed by overexpression of MYH9 in NPC cells. Clinically, a low level of DNAJA4 indicated poor prognosis and an increased probability of distant metastasis in NPC patients. Collectively, DNAJA4 serves as a crucial driver for NPC invasion and metastasis, and the DNAJA4-PSMD2-MYH9 axis might contain potential targets for NPC treatments.
Collapse
Affiliation(s)
- Qun Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, PR China
| | - Ping Feng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xun-Hua Zhu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Shi-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Ming-Liang Ye
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xiao-Jing Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Sha Gong
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Sheng-Yan Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xi-Rong Tan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Shi-Wei He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Ying-Qing Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| |
Collapse
|
31
|
Colin A, Orhant-Prioux M, Guérin C, Savinov M, Cao W, Vianay B, Scarfone I, Roux A, De La Cruz EM, Mogilner A, Théry M, Blanchoin L. Friction patterns guide actin network contraction. Proc Natl Acad Sci U S A 2023; 120:e2300416120. [PMID: 37725653 PMCID: PMC10523593 DOI: 10.1073/pnas.2300416120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/09/2023] [Indexed: 09/21/2023] Open
Abstract
The shape of cells is the outcome of the balance of inner forces produced by the actomyosin network and the resistive forces produced by cell adhesion to their environment. The specific contributions of contractile, anchoring and friction forces to network deformation rate and orientation are difficult to disentangle in living cells where they influence each other. Here, we reconstituted contractile actomyosin networks in vitro to study specifically the role of the friction forces between the network and its anchoring substrate. To modulate the magnitude and spatial distribution of friction forces, we used glass or lipids surface micropatterning to control the initial shape of the network. We adapted the concentration of Nucleating Promoting Factor on each surface to induce the assembly of actin networks of similar densities and compare the deformation of the network toward the centroid of the pattern shape upon myosin-induced contraction. We found that actin network deformation was faster and more coordinated on lipid bilayers than on glass, showing the resistance of friction to network contraction. To further study the role of the spatial distribution of these friction forces, we designed heterogeneous micropatterns made of glass and lipids. The deformation upon contraction was no longer symmetric but biased toward the region of higher friction. Furthermore, we showed that the pattern of friction could robustly drive network contraction and dominate the contribution of asymmetric distributions of myosins. Therefore, we demonstrate that during contraction, both the active and resistive forces are essential to direct the actin network deformation.
Collapse
Affiliation(s)
- Alexandra Colin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Magali Orhant-Prioux
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Christophe Guérin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Mariya Savinov
- Courant Institute of Mathematical Sciences, New York University, New York, NY10012
| | - Wenxiang Cao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520-8114
| | - Benoit Vianay
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| | - Ilaria Scarfone
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, CH-1211Geneva, Switzerland
| | - Enrique M. De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520-8114
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, NY10012
| | - Manuel Théry
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| | - Laurent Blanchoin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| |
Collapse
|
32
|
Hojjatian A, Taylor DW, Daneshparvar N, Fagnant PM, Trybus KM, Taylor KA. Double-headed binding of myosin II to F-actin shows the effect of strain on head structure. J Struct Biol 2023; 215:107995. [PMID: 37414375 PMCID: PMC10544818 DOI: 10.1016/j.jsb.2023.107995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Force production in muscle is achieved through the interaction of myosin and actin. Strong binding states in active muscle are associated with Mg·ADP bound to the active site; release of Mg·ADP allows rebinding of ATP and dissociation from actin. Thus, Mg·ADP binding is positioned for adaptation as a force sensor. Mechanical loads on the lever arm can affect the ability of myosin to release Mg·ADP but exactly how this is done is poorly defined. Here we use F-actin decorated with double-headed smooth muscle myosin fragments in the presence of Mg·ADP to visualize the effect of internally supplied tension on the paired lever arms using cryoEM. The interaction of the paired heads with two adjacent actin subunits is predicted to place one lever arm under positive and the other under negative strain. The converter domain is believed to be the most flexible domain within myosin head. Our results, instead, point to the segment of heavy chain between the essential and regulatory light chains as the location of the largest structural change. Moreover, our results suggest no large changes in the myosin coiled coil tail as the locus of strain relief when both heads bind F-actin. The method would be adaptable to double-headed members of the myosin family. We anticipate that the study of actin-myosin interaction using double-headed fragments enables visualization of domains that are typically noisy in decoration with single-headed fragments.
Collapse
Affiliation(s)
- Alimohammad Hojjatian
- Inst. of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, United States
| | - Dianne W Taylor
- Inst. of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, United States
| | - Nadia Daneshparvar
- Inst. of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, United States
| | - Patricia M Fagnant
- Dept of Molecular Physiology & Biophysics, University of Vermont College of Medicine, Burlington, VT 05405, United States
| | - Kathleen M Trybus
- Dept of Molecular Physiology & Biophysics, University of Vermont College of Medicine, Burlington, VT 05405, United States
| | - Kenneth A Taylor
- Inst. of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
33
|
Steffensen KE, Dawson JF. Actin's C-terminus coordinates actin structural changes and functions. Cytoskeleton (Hoboken) 2023; 80:313-329. [PMID: 37036084 DOI: 10.1002/cm.21757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/17/2023] [Accepted: 03/30/2023] [Indexed: 04/11/2023]
Abstract
Actin is essential to eukaryotic cellular processes. Actin's C-terminus appears to play a direct role in modulating actin's structure and properties, facilitating the binding and function of actin-binding proteins (ABPs). The structural and functional characterization of filamentous actin's C-terminus has been impeded by its inherent flexibility, as well as actin's resistance to crystallization for x-ray diffraction and the historical resolution constraints associated with electron microscopy. Many biochemical studies have established that actin's C-terminus must retain its flexibility and structural integrity to modulate actin's structure and functions. For example, C-terminal structural changes are known to affect nucleotide binding and exchange, as well as propagate actin structural changes throughout extensive allosteric networks, facilitating the binding and function of ABPs. Advances in electron microscopy have resulted in high-resolution structures of filamentous actin, providing insights into subtle structural changes that are mediated by actin's C-terminus. Here, we review existing knowledge establishing the importance of actin's C-terminus within actin structural changes and functions and discuss how modern structural characterization techniques provide the tools to understand the role of actin's C-terminus in cellular processes.
Collapse
Affiliation(s)
- Karl E Steffensen
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - John F Dawson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
34
|
Häfner L, Brockmeyer J, Haase I, Kranz B, Jira W. Identification of Cross-Species Marker Peptides for the Detection of Mammalian and Poultry Meat in Vegan and Vegetarian Foods. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:12597-12608. [PMID: 37561394 DOI: 10.1021/acs.jafc.3c01100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Authentication of vegan and vegetarian foods is important since these increasingly popular food items could be adulterated with cheap meat to increase profit margins. In this study, nine marker peptides for the detection of meat (several species) were identified applying liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). These marker peptides enable the crucial differentiation of beef versus milk and chicken meat versus egg, demonstrated by the investigation of 19 commercial vegetarian meat substitutes containing milk and egg. Extensive experimental testing proved the presence of the cross-species meat marker peptides in 19 food-relevant types of mammals and poultry as well as their absence in more than 136 plant-based ingredients for the production of vegan and vegetarian foods. An authentic vegan sausage matrix based on an actual retail product was produced and spiked with 5.0%, w/w meat to confirm the high signal intensities and the heat stability of the marker peptides.
Collapse
Affiliation(s)
- Lukas Häfner
- National Reference Centre for Authentic Food, Max Rubner-Institut (MRI), E.-C.-Baumann-Straße 20, Kulmbach 95326, Germany
| | - Jens Brockmeyer
- Department of Food Chemistry, Institute for Biochemistry and Technical Biochemistry, University of Stuttgart, Allmandring 5B, Stuttgart 70569, Germany
| | - Ilka Haase
- National Reference Centre for Authentic Food, Max Rubner-Institut (MRI), E.-C.-Baumann-Straße 20, Kulmbach 95326, Germany
| | - Bertolt Kranz
- National Reference Centre for Authentic Food, Max Rubner-Institut (MRI), E.-C.-Baumann-Straße 20, Kulmbach 95326, Germany
| | - Wolfgang Jira
- Department of Safety and Quality of Meat, Max Rubner-Institut (MRI), E.-C.-Baumann-Straße 20, Kulmbach 95326, Germany
| |
Collapse
|
35
|
Leanza L, Perego C, Pesce L, Salvalaglio M, von Delius M, Pavan GM. Into the dynamics of rotaxanes at atomistic resolution. Chem Sci 2023; 14:6716-6729. [PMID: 37350834 PMCID: PMC10283497 DOI: 10.1039/d3sc01593a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/04/2023] [Indexed: 06/24/2023] Open
Abstract
Mechanically-interlocked molecules (MIMs) are at the basis of artificial molecular machines and are attracting increasing interest for various applications, from catalysis to drug delivery and nanoelectronics. MIMs are composed of mechanically-interconnected molecular sub-parts that can move with respect to each other, imparting these systems innately dynamical behaviors and interesting stimuli-responsive properties. The rational design of MIMs with desired functionalities requires studying their dynamics at sub-molecular resolution and on relevant timescales, which is challenging experimentally and computationally. Here, we combine molecular dynamics and metadynamics simulations to reconstruct the thermodynamics and kinetics of different types of MIMs at atomistic resolution under different conditions. As representative case studies, we use rotaxanes and molecular shuttles substantially differing in structure, architecture, and dynamical behavior. Our computational approach provides results in agreement with the available experimental evidence and a direct demonstration of the critical effect of the solvent on the dynamics of the MIMs. At the same time, our simulations unveil key factors controlling the dynamics of these systems, providing submolecular-level insights into the mechanisms and kinetics of shuttling. Reconstruction of the free-energy profiles from the simulations reveals details of the conformations of macrocycles on the binding site that are difficult to access via routine experiments and precious for understanding the MIMs' behavior, while their decomposition in enthalpic and entropic contributions unveils the mechanisms and key transitions ruling the intermolecular movements between metastable states within them. The computational framework presented herein is flexible and can be used, in principle, to study a variety of mechanically-interlocked systems.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Applied Science and Technology, Politecnico di Torino Corso Duca degli Abruzzi, 24 10129 Torino Italy
| | - Claudio Perego
- Department of Innovative Technologies, University of Applied Sciences and Arts of Southern Switzerland, Polo Universitario Lugano Campus Est, Via la Santa 1 6962 Lugano-Viganello Switzerland
| | - Luca Pesce
- Department of Innovative Technologies, University of Applied Sciences and Arts of Southern Switzerland, Polo Universitario Lugano Campus Est, Via la Santa 1 6962 Lugano-Viganello Switzerland
| | - Matteo Salvalaglio
- Department of Chemical Engineering, University College London London WC1E 7JE UK
| | - Max von Delius
- Institute of Organic Chemistry, Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Giovanni M Pavan
- Department of Applied Science and Technology, Politecnico di Torino Corso Duca degli Abruzzi, 24 10129 Torino Italy
- Department of Innovative Technologies, University of Applied Sciences and Arts of Southern Switzerland, Polo Universitario Lugano Campus Est, Via la Santa 1 6962 Lugano-Viganello Switzerland
| |
Collapse
|
36
|
Singhania A, Kalita S, Chettri P, Ghosh S. Accounts of applied molecular rotors and rotary motors: recent advances. NANOSCALE ADVANCES 2023; 5:3177-3208. [PMID: 37325522 PMCID: PMC10262963 DOI: 10.1039/d3na00010a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Molecular machines are nanoscale devices capable of performing mechanical works at molecular level. These systems could be a single molecule or a collection of component molecules that interrelate with one another to produce nanomechanical movements and resulting performances. The design of the components of molecular machine with bioinspired traits results in various nanomechanical motions. Some known molecular machines are rotors, motors, nanocars, gears, elevators, and so on based on their nanomechanical motion. The conversion of these individual nanomechanical motions to collective motions via integration into suitable platforms yields impressive macroscopic output at varied sizes. Instead of limited experimental acquaintances, the researchers demonstrated several applications of molecular machines in chemical transformation, energy conversion, gas/liquid separation, biomedical use, and soft material fabrication. As a result, the development of new molecular machines and their applications has accelerated over the previous two decades. This review highlights the design principles and application scopes of several rotors and rotary motor systems because these machines are used in real applications. This review also offers a systematic and thorough overview of current advancements in rotary motors, providing in-depth knowledge and predicting future problems and goals in this area.
Collapse
Affiliation(s)
- Anup Singhania
- Natural Product Chemistry Group, Chemical Sciences & Technology Division, CSIR-North East Institute of Science & Technology Jorhat 785006 Assam India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Sudeshna Kalita
- Natural Product Chemistry Group, Chemical Sciences & Technology Division, CSIR-North East Institute of Science & Technology Jorhat 785006 Assam India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Prerna Chettri
- Natural Product Chemistry Group, Chemical Sciences & Technology Division, CSIR-North East Institute of Science & Technology Jorhat 785006 Assam India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Subrata Ghosh
- Natural Product Chemistry Group, Chemical Sciences & Technology Division, CSIR-North East Institute of Science & Technology Jorhat 785006 Assam India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| |
Collapse
|
37
|
Rajan S, Kudryashov DS, Reisler E. Actin Bundles Dynamics and Architecture. Biomolecules 2023; 13:450. [PMID: 36979385 PMCID: PMC10046292 DOI: 10.3390/biom13030450] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Cells use the actin cytoskeleton for many of their functions, including their division, adhesion, mechanosensing, endo- and phagocytosis, migration, and invasion. Actin bundles are the main constituent of actin-rich structures involved in these processes. An ever-increasing number of proteins that crosslink actin into bundles or regulate their morphology is being identified in cells. With recent advances in high-resolution microscopy and imaging techniques, the complex process of bundles formation and the multiple forms of physiological bundles are beginning to be better understood. Here, we review the physiochemical and biological properties of four families of highly conserved and abundant actin-bundling proteins, namely, α-actinin, fimbrin/plastin, fascin, and espin. We describe the similarities and differences between these proteins, their role in the formation of physiological actin bundles, and their properties-both related and unrelated to their bundling abilities. We also review some aspects of the general mechanism of actin bundles formation, which are known from the available information on the activity of the key actin partners involved in this process.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
38
|
Smith JD, Brawley J, Bordenave KC, Olsen RK, Intasiri A, Cremo CR, Bell TW. Isoform selectivities of novel 4-hydroxycoumarin imines as inhibitors of myosin II. Eur J Med Chem 2023; 247:115008. [PMID: 36543032 PMCID: PMC9889102 DOI: 10.1016/j.ejmech.2022.115008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Muscle myosin inhibition could be used to treat many medical conditions involving hypercontractile states, including muscle spasticity, chronic musculoskeletal pain, and hypertrophic cardiomyopathy. A series of 13 advanced analogs of 3-(N-butylethanimidoyl)ethyl)-4-hydroxy-2H-chromen-2-one (BHC) were synthesized to explore extended imine nitrogen side chains and compare aldimines vs. ketimines. None of the new analogs inhibit nonmuscle myosin in a cytokinesis assay. ATPase structure-activity relationships reveal that selectivity for cardiac vs. skeletal myosin can be tuned with subtle structural changes. None of the compounds inhibited smooth muscle myosin II. Docking the compounds to homology models of cardiac and skeletal myosin II gave rationales for the effects of side arm length on inhibition selectivity and for cardiac vs. skeletal myosin. Properties including solubility, stability and toxicity, suggest that certain BHC analogs may be useful as candidates for preclinical studies or as lead compounds for advanced candidates for drugs with cardiac or skeletal muscle myosin selectivity.
Collapse
Affiliation(s)
- Joshua D Smith
- Department of Pharmacology, University of Nevada, School of Medicine, Reno, NV, 89557-0318, USA
| | - Jhonnathan Brawley
- Department of Chemistry, University of Nevada, Reno, NV, 89557-0216, USA
| | - Kate C Bordenave
- Department of Pharmacology, University of Nevada, School of Medicine, Reno, NV, 89557-0318, USA
| | - Ryan K Olsen
- Department of Chemistry, University of Nevada, Reno, NV, 89557-0216, USA
| | - Amarawan Intasiri
- Department of Chemistry, University of Nevada, Reno, NV, 89557-0216, USA
| | - Christine R Cremo
- Department of Pharmacology, University of Nevada, School of Medicine, Reno, NV, 89557-0318, USA.
| | - Thomas W Bell
- Department of Chemistry, University of Nevada, Reno, NV, 89557-0216, USA.
| |
Collapse
|
39
|
Dotts AJ, Reiman D, Yin P, Kujawa S, Grobman WA, Dai Y, Bulun SE. In Vivo Genome-Wide PGR Binding in Pregnant Human Myometrium Identifies Potential Regulators of Labor. Reprod Sci 2023; 30:544-559. [PMID: 35732928 PMCID: PMC9988762 DOI: 10.1007/s43032-022-01002-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/03/2022] [Indexed: 12/22/2022]
Abstract
The alterations in myometrial biology during labor are not well understood. The myometrium is the contractile portion of the uterus and contributes to labor, a process that may be regulated by the steroid hormone progesterone. Thus, human myometrial tissues from term pregnant in-active-labor (TIL) and term pregnant not-in-labor (TNIL) subjects were used for genome-wide analyses to elucidate potential future preventive or therapeutic targets involved in the regulation of labor. Using myometrial tissues directly subjected to RNA sequencing (RNA-seq), progesterone receptor (PGR) chromatin immunoprecipitation sequencing (ChIP-seq), and histone modification ChIP-seq, we profiled genome-wide changes associated with gene expression in myometrial smooth muscle tissue in vivo. In TIL myometrium, PGR predominantly occupied promoter regions, including the classical progesterone response element, whereas it bound mainly to intergenic regions in TNIL myometrial tissue. Differential binding analysis uncovered over 1700 differential PGR-bound sites between TIL and TNIL, with 1361 sites gained and 428 lost in labor. Functional analysis identified multiple pathways involved in cAMP-mediated signaling enriched in labor. A three-way integration of the data for ChIP-seq, RNA-seq, and active histone marks uncovered the following genes associated with PGR binding, transcriptional activation, and altered mRNA levels: ATP11A, CBX7, and TNS1. In vitro studies showed that ATP11A, CBX7, and TNS1 are progesterone responsive. We speculate that these genes may contribute to the contractile phenotype of the myometrium during various stages of labor. In conclusion, we provide novel labor-associated genome-wide events and PGR-target genes that can serve as targets for future mechanistic studies.
Collapse
Affiliation(s)
- Ariel J Dotts
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Derek Reiman
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ping Yin
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Stacy Kujawa
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - William A Grobman
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Yang Dai
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Serdar E Bulun
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
40
|
Fitz GN, Weck ML, Bodnya C, Perkins OL, Tyska MJ. Protrusion growth driven by myosin-generated force. Dev Cell 2023; 58:18-33.e6. [PMID: 36626869 PMCID: PMC9940483 DOI: 10.1016/j.devcel.2022.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/10/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023]
Abstract
Actin-based protrusions extend from the surface of all eukaryotic cells, where they support diverse activities essential for life. Models of protrusion growth hypothesize that actin filament assembly exerts force for pushing the plasma membrane outward. However, membrane-associated myosin motors are also abundant in protrusions, although their potential for contributing, growth-promoting force remains unexplored. Using an inducible system that docks myosin motor domains to membrane-binding modules with temporal control, we found that application of myosin-generated force to the membrane is sufficient for driving robust protrusion elongation in human, mouse, and pig cell culture models. Protrusion growth scaled with motor accumulation, required barbed-end-directed force, and was independent of cargo delivery or recruitment of canonical elongation factors. Application of growth-promoting force was also supported by structurally distinct myosin motors and membrane-binding modules. Thus, myosin-generated force can drive protrusion growth, and this mechanism is likely active in diverse biological contexts.
Collapse
Affiliation(s)
- Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Meredith L Weck
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Caroline Bodnya
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Olivia L Perkins
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
41
|
Lobo GP, Radhakrishnan R, Leung M, Gruesen A, Knölker HJ, van Kuijk FJ, Montezuma SR. In Silico Prediction of MYO1C-Rhodopsin Interactions and Its Significance in Protein Localization and Visual Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:499-505. [PMID: 37440078 DOI: 10.1007/978-3-031-27681-1_73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Rods and cones are photoreceptor neurons in the retina that are required for visual sensation in vertebrates, where proper protein localization and compartmentalization are critical for phototransduction and visual function. In human retinal diseases, improper protein transport to the outer segment (OS) or mislocalization of proteins to the inner segment (IS) could lead to impaired visual responses and photoreceptor cell degeneration, causing a loss of visual function. We showed involvement of an unconventional motor protein, MYO1C, in the proper localization of rhodopsin to the OS, where loss of MYO1C in a mammalian model caused mislocalization of rhodopsin to IS and cell bodies, leading to progressively severe retinal phenotypes. In this study, using modeling and docking analysis, we aimed to identify the protein-protein interaction sites between MYO1C and Rhodopsin to establish a hypothesis that a physical interaction between these proteins is necessary for the proper trafficking of rhodopsin and visual function.
Collapse
Affiliation(s)
- Glenn P Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA.
| | - Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA
| | - Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA
| | | | - Frederik J van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
42
|
Souissi A, Abdelmalek Driss D, Chakchouk I, Ben Said M, Ben Ayed I, Mosrati MA, Elloumi I, Tlili A, Aifa S, Masmoudi S. Molecular insights into MYO3A kinase domain variants explain variability in both severity and progression of DFNB30 hearing impairment. J Biomol Struct Dyn 2022; 40:10940-10951. [PMID: 34423747 DOI: 10.1080/07391102.2021.1953600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Hereditary hearing impairment (HI) is a common disease with the highest incidence among sensory defects. Several genes have been identified to affect stereocilia structure causing HI, including the unconventional myosin3A. Interestingly, we noticed that variants in MYO3A gene have been previously found to cause variable HI onset and severity. Using clinical exome sequencing, we identified a novel pathogenic variant p.(Lys50Arg) in the MYO3A kinase domain (MYO3A-KD). Previous in vitro studies supported its damaging effect as a 'kinase-dead' mutant. We further analyzed this variation through molecular dynamics which predicts that changes in flexibility of MYO3A structure would influence the protein-ATP binding properties. This Lys50Arg mutation segregated with congenital profound non-syndromic HI. To better investigate this variability, we collected previously identified MYO3A-KDs variants, p.(Tyr129Cys), p.(His142Gln) and p.(Pro189Thr), and built both wild type and mutant 3 D MYO3A-KD models to assess their impact on the protein structure and function. Our results suggest that KD mutations could either cause a congenital profound form of HI, when particularly affecting the kinase activity and preventing the auto-phosphorylation of the motor, or a late onset and progressive form, when partially or completely inactivating the MYO3A protein. In conclusion, we report a novel pathogenic variant affecting the ATP-binding site within the MYO3A-KD causing congenital profound HI. Through computational approaches we provide a deeper understanding on the correlation between the effects of MYO3A-KD mutations and the variable hearing phenotypes. To the best of our knowledge this is the first study to correlate mutations' genotypes with the variable phenotypes of DFNB30.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Amal Souissi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Dorra Abdelmalek Driss
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Imen Chakchouk
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Mariem Ben Said
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ikhlas Ben Ayed
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia.,Medical Genetic Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
| | - Mohamed Ali Mosrati
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ines Elloumi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Sami Aifa
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Saber Masmoudi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
43
|
Mohan IK, Baba KSSS, Iyyapu R, Thirumalasetty S, Satish OS. Advances in congestive heart failure biomarkers. Adv Clin Chem 2022; 112:205-248. [PMID: 36642484 DOI: 10.1016/bs.acc.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Congestive heart failure (CHF) is the leading cause of morbidity and mortality in the elderly worldwide. Although many biomarkers associated with in heart failure, these are generally prognostic and identify patients with moderate and severe disease. Unfortunately, the role of biomarkers in decision making for early and advanced heart failure remains largely unexplored. Previous studies suggest the natriuretic peptides have the potential to improve the diagnosis of heart failure, but they still have significant limitations related to cut-off values. Although some promising cardiac biomarkers have emerged, comprehensive data from large cohort studies is lacking. The utility of multiple biomarkers that reflect various pathophysiologic pathways are increasingly being explored in heart failure risk stratification and to diagnose disease conditions promptly and accurately. MicroRNAs serve as mediators and/or regulators of renin-angiotensin-induced cardiac remodeling by directly targeting enzymes, receptors and signaling molecules. The role of miRNA in HF diagnosis is a promising area of research and further exploration may offer both diagnostic and prognostic applications and phenotype-specific targets. In this review, we provide insight into the classification of different biochemical and molecular markers associated with CHF, examine clinical usefulness in CHF and highlight the most clinically relevant.
Collapse
Affiliation(s)
| | - K S S Sai Baba
- Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| | - Rohit Iyyapu
- Katuri Medical College & Hospital, Guntur, Andhra Pradesh, India
| | | | - O Sai Satish
- Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| |
Collapse
|
44
|
Cartón-García F, Brotons B, Anguita E, Dopeso H, Tarragona J, Nieto R, García-Vidal E, Macaya I, Zagyva Z, Dalmau M, Sánchez-Martín M, van Ijzendoorn SCD, Landolfi S, Hernandez-Losa J, Schwartz Jr S, Matias-Guiu X, Ramón y Cajal S, Martínez-Barriocanal Á, Arango D. Myosin Vb as a tumor suppressor gene in intestinal cancer. Oncogene 2022; 41:5279-5288. [DOI: 10.1038/s41388-022-02508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
|
45
|
Ahmed RO, Ali A, Al-Tobasei R, Leeds T, Kenney B, Salem M. Weighted Single-Step GWAS Identifies Genes Influencing Fillet Color in Rainbow Trout. Genes (Basel) 2022; 13:genes13081331. [PMID: 35893068 PMCID: PMC9332390 DOI: 10.3390/genes13081331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 02/04/2023] Open
Abstract
The visual appearance of the fish fillet is a significant determinant of consumers' purchase decisions. Depending on the rainbow trout diet, a uniform bright white or reddish/pink fillet color is desirable. Factors affecting fillet color are complex, ranging from the ability of live fish to accumulate carotenoids in the muscle to preharvest environmental conditions, early postmortem muscle metabolism, and storage conditions. Identifying genetic markers of fillet color is a desirable goal but a challenging task for the aquaculture industry. This study used weighted, single-step GWAS to explore the genetic basis of fillet color variation in rainbow trout. We identified several SNP windows explaining up to 3.5%, 2.5%, and 1.6% of the additive genetic variance for fillet redness, yellowness, and whiteness, respectively. SNPs are located within genes implicated in carotenoid metabolism (β,β-carotene 15,15'-dioxygenase, retinol dehydrogenase) and myoglobin homeostasis (ATP synthase subunit β, mitochondrial (ATP5F1B)). These genes are involved in processes that influence muscle pigmentation and postmortem flesh coloration. Other identified genes are involved in the maintenance of muscle structural integrity (kelch protein 41b (klh41b), collagen α-1(XXVIII) chain (COL28A1), and cathepsin K (CTSK)) and protection against lipid oxidation (peroxiredoxin, superoxide dismutase 2 (SOD2), sestrin-1, Ubiquitin carboxyl-terminal hydrolase-10 (USP10)). A-to-G single-nucleotide polymorphism in β,β-carotene 15,15'-dioxygenase, and USP10 result in isoleucine-to-valine and proline-to-leucine non-synonymous amino acid substitutions, respectively. Our observation confirms that fillet color is a complex trait regulated by many genes involved in carotenoid metabolism, myoglobin homeostasis, protection against lipid oxidation, and maintenance of muscle structural integrity. The significant SNPs identified in this study could be prioritized via genomic selection in breeding programs to improve fillet color in rainbow trout.
Collapse
Affiliation(s)
- Ridwan O. Ahmed
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (R.O.A.); (A.A.)
| | - Ali Ali
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (R.O.A.); (A.A.)
| | - Rafet Al-Tobasei
- Computational Science Program, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Tim Leeds
- United States Department of Agriculture Kearneysville, National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, Kearneysville, WV 25430, USA;
| | - Brett Kenney
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV 26506, USA;
| | - Mohamed Salem
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (R.O.A.); (A.A.)
- Correspondence:
| |
Collapse
|
46
|
Najafabadi FR, Leaver M, Grill SW. Orchestrating nonmuscle myosin II filament assembly at the onset of cytokinesis. Mol Biol Cell 2022; 33:ar74. [PMID: 35544301 PMCID: PMC9635286 DOI: 10.1091/mbc.e21-12-0599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/14/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
Contractile forces in the actomyosin cortex are required for cellular morphogenesis. This includes the invagination of the cell membrane during division, where filaments of nonmuscle myosin II (NMII) are responsible for generating contractile forces in the cortex. However, how NMII heterohexamers form filaments in vivo is not well understood. To quantify NMII filament assembly dynamics, we imaged the cortex of Caenorhabditis elegans embryos at high spatial resolution around the time of the first division. We show that during the assembly of the cytokinetic ring, the number of NMII filaments in the cortex increases and more NMII motors are assembled into each filament. These dynamics are influenced by two proteins in the RhoA GTPase pathway, the RhoA-dependent kinase LET-502 and the myosin phosphatase MEL-11. We find that these two proteins differentially regulate NMII activity at the anterior and at the division site. We show that the coordinated action of these regulators generates a gradient of free NMII in the cytoplasm driving a net diffusive flux of NMII motors toward the cytokinetic ring. Our work highlights how NMII filament assembly and disassembly dynamics are orchestrated over space and time to facilitate the up-regulation of cortical contractility during cytokinesis.
Collapse
Affiliation(s)
- Fereshteh R. Najafabadi
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
- Biotechnology Centre, Technische Universität Dresden, Tatzberg 47/49, Dresden 01307
| | - Mark Leaver
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
- Biotechnology Centre, Technische Universität Dresden, Tatzberg 47/49, Dresden 01307
| | - Stephan W. Grill
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden 01307, Germany
- Biotechnology Centre, Technische Universität Dresden, Tatzberg 47/49, Dresden 01307
- Excellence Cluster Physics of Life, Technische Universität, Dresden 01307, Germany
| |
Collapse
|
47
|
Wu Y, Zhang Y, Xu X, Wang W. Effect of Ca 2+ binding states of calmodulin on the conformational dynamics and force responses of myosin lever arm. J Chem Phys 2022; 157:035101. [DOI: 10.1063/5.0095842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The mechanochemical coupling and biological function of myosin motors are regulated by Ca2+ concentrations. As one of the regulation pathways, Ca2+ binding induces conformational change of the light chain calmodulin and its binding modes with myosin lever arm, which can affect the stiffness of the lever arm and force transmission. However, the underlying molecular mechanism of the Ca2+ regulated stiffness change is not fully understood. Here we study the effect of Ca2+ binding on the conformational dynamics and stiffness of the myosin VIIa lever arm bound with calmodulin by performing molecular dynamics simulations and dynamic correlation network analysis. The results showed that the calmodulin bound lever arm at apo state can sample three different conformations. In addition to the conformation observed in crystal structure, calmodulin bound lever arm at apo condition can also adopt another two conformations featured by different extents of small-angle bending of the lever arm. However, large-angle bending is strongly prohibited. Such results suggest that the calmodulin bound lever arm without Ca2+ binding is plastic for small-angle deformation but shows high stiffness for large-angle deformation. In comparison, after the binding of Ca2+, although the calmodulin bound lever arm is locally more rigid, it can adopt largely deformed or even unfolded conformations, which may render the lever arm incompetent for force transmission. The conformational plasticity of the lever arm for small-angle deformation at apo condition may be utilized as force buffer to prevent the lever arm from unfolding during the power stroke action of the motor domain.
Collapse
Affiliation(s)
- Yichao Wu
- Department of Physics, Nanjing University, China
| | | | | | - Wei Wang
- Department of Physics, Nanjing University, China
| |
Collapse
|
48
|
Defective VWF secretion due to the expression of MYH9-RD E1841K mutant in endothelial cells disrupts hemostasis. Blood Adv 2022; 6:4537-4552. [PMID: 35764499 DOI: 10.1182/bloodadvances.2022008011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/08/2022] [Indexed: 11/20/2022] Open
Abstract
Mutations in MYH9, the gene encoding the heavy chain of non-muscle myosin IIa (NMII-A), cause MYH9-related disease (MYH9-RD) that is an autosomal-dominant thrombocytopenia with bleeding tendency. Previously, we showed that NMII-A in endothelial cells (ECs) is critical for hemostasis via regulating von Willebrand factor (VWF) release from Weibel-Palade bodies (WPBs). The aim of this study was to determine the role of the expression of MYH9 mutants in ECs in the pathogenesis of the MYH9-RD bleeding symptom. First, we expressed the 5 most common NMII-A mutants in ECs, and found that E1841K mutant-expressing ECs secreted less VWF than the controls in response to a cAMP signaling agonist. Then, we generated 2 knockin mouse lines, one with Myh9 E1841K in ECs and the other in megakaryocytes. Endothelium-specific E1841K mice exhibited impaired cAMP-induced VWF release and a prolonged bleeding time with normal platelets, while megakaryocyte-specific E1841K mice exhibited macrothrombocytopenia and a prolonged bleeding time with normal VWF release. Finally, we present mechanistic findings that E1841K mutation not only interferes with S1943 phosphorylation and impairs the peripheral distribution of Rab27a positive WPBs in ECs under quiescent condition, but also interferes with S1916 phosphorylation by disrupting the interaction with zyxin and CKIIα, and reduces actin framework formation around WPBs and subsequent VWF secretion under the stimulation by a cAMP agonist. Altogether, our results suggest that impaired cAMP-induced endothelial VWF secretion by E1841K mutant expression may contribute to the MYH9-RD bleeding phenotype.
Collapse
|
49
|
Coleman JR, Deguchi H, Deguchi TK, Cohen MJ, Moore EE, Griffin JH. Full-length plasma skeletal muscle myosin isoform deficiency is associated with coagulopathy in acutely injured patients. J Thromb Haemost 2022; 20:1385-1389. [PMID: 35253989 PMCID: PMC9310574 DOI: 10.1111/jth.15695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/01/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Skeletal muscle myosin (SkM) molecules are procoagulant both in vitro and in vivo. The association of plasma SkM isoforms with blood coagulability and hemostatic capacity has not been defined. OBJECTIVES We hypothesized that coagulopathy in acutely injured patients is associated with procoagulant plasma SkM heavy chain levels. METHODS To test this hypothesis, citrated whole blood and plasma from 104 trauma patients were collected and studied to obtain data for rapid thrombelastography, international normalized ratios, and plasma SkM levels. Coagulability parameters were dichotomized by the threshold for the hypercoagulable trauma-induced coagulopathy. RESULTS Lower plasma full-length SkM heavy chain (full-SkM) levels were associated with higher international normalized ratio values (>1.3) (p = .03). The full-SkM levels were also associated with a lower rate of clot propagation (thrombelastography angle <65°) (p = .004), and plasma full-SkM levels were positively correlated with the thrombelastography angle (r2 = .32, p = .0007). The trauma patient group with the lower plasma full-SkM levels showed an association with lower clot strength (maximum amplitude <55 mm) (p = .002), and plasma full-SkM levels positively correlated with maximum amplitude (r2 = .27, p = .005). Hyperfibrinolysis was associated with significantly decreased full-SkM levels (p = .03). Trauma patients who required red blood cells and fresh frozen plasma transfusions had lower plasma full-SkM levels compared with those without transfusions (p = .04 and .02, respectively). CONCLUSIONS In acutely injured trauma patients, lower levels of plasma full-SkM levels are linked to hypocoagulability in trauma-induced coagulopathy, implying that SkM plays a role in the hemostatic capacity in trauma patients and may contribute to trauma-induced coagulopathy.
Collapse
Affiliation(s)
| | - Hiroshi Deguchi
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Taichi K. Deguchi
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Mitchel J. Cohen
- Department of SurgeryUniversity of ColoradoAuroraColoradoUSA
- Ernest E Moore Shock Trauma Center at Denver HealthDenverColoradoUSA
| | - Ernest E. Moore
- Department of SurgeryUniversity of ColoradoAuroraColoradoUSA
- Ernest E Moore Shock Trauma Center at Denver HealthDenverColoradoUSA
| | - John H. Griffin
- Department of Molecular MedicineThe Scripps Research InstituteLa JollaCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan DiegoCaliforniaUSA
| |
Collapse
|
50
|
Chen Y, Zhang T, Xian M, Zhang R, Yang W, Su B, Yang G, Sun L, Xu W, Xu S, Gao H, Xu L, Gao X, Li J. A draft genome of Drung cattle reveals clues to its chromosomal fusion and environmental adaptation. Commun Biol 2022; 5:353. [PMID: 35418663 PMCID: PMC9008013 DOI: 10.1038/s42003-022-03298-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 03/21/2022] [Indexed: 12/02/2022] Open
Abstract
Drung cattle (Bos frontalis) have 58 chromosomes, differing from the Bos taurus 2n = 60 karyotype. To date, its origin and evolution history have not been proven conclusively, and the mechanisms of chromosome fusion and environmental adaptation have not been clearly elucidated. Here, we assembled a high integrity and good contiguity genome of Drung cattle with 13.7-fold contig N50 and 4.1-fold scaffold N50 improvements over the recently published Indian mithun assembly, respectively. Speciation time estimation and phylogenetic analysis showed that Drung cattle diverged from Bos taurus into an independent evolutionary clade. Sequence evidence of centromere regions provides clues to the breakpoints in BTA2 and BTA28 centromere satellites. We furthermore integrated a circulation and contraction-related biological process involving 43 evolutionary genes that participated in pathways associated with the evolution of the cardiovascular system. These findings may have important implications for understanding the molecular mechanisms of chromosome fusion, alpine valleys adaptability and cardiovascular function.
Collapse
Affiliation(s)
- Yan Chen
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Tianliu Zhang
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Ming Xian
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Rui Zhang
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Weifei Yang
- 1 Gene Co., Ltd, 310051, Hangzhou, P.R. China
- Annoroad Gene Technology (Beijing) Co., Ltd, 100176, Beijing, P.R. China
| | - Baqi Su
- Drung Cattle Conservation Farm in Jiudang Wood, Drung and Nu Minority Autonomous County, Gongshan, 673500, Kunming, Yunnan, P.R. China
| | - Guoqiang Yang
- Livestock and Poultry Breed Improvement Center, Nujiang Lisu Minority Autonomous Prefecture, 673199, Kunming, Yunnan, P.R. China
| | - Limin Sun
- Yunnan Animal Husbandry Service, 650224, Kunming, Yunnan, P.R. China
| | - Wenkun Xu
- Yunnan Animal Husbandry Service, 650224, Kunming, Yunnan, P.R. China
| | - Shangzhong Xu
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Huijiang Gao
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Lingyang Xu
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China
| | - Xue Gao
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China.
| | - Junya Li
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193, Beijing, P.R. China.
| |
Collapse
|