1
|
Shangguan J, Rock RS. Hundreds of myosin 10s are pushed to the tips of filopodia and could cause traffic jams on actin. eLife 2024; 12:RP90603. [PMID: 39480891 PMCID: PMC11527427 DOI: 10.7554/elife.90603] [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] [Indexed: 11/02/2024] Open
Abstract
Myosin 10 (Myo10) is a motor protein known for its role in filopodia formation. Although Myo10-driven filopodial dynamics have been characterized, there is no information about the absolute number of Myo10 molecules during the filopodial lifecycle. To better understand molecular stoichiometries and packing restraints in filopodia, we measured Myo10 abundance in these structures. We combined SDS-PAGE densitometry with epifluorescence microscopy to quantitate HaloTag-labeled Myo10 in U2OS cells. About 6% of total intracellular Myo10 localizes to filopodia, where it enriches at opposite cellular ends. Hundreds of Myo10s are in a typical filopodium, and their distribution across filopodia is log-normal. Some filopodial tips even contain more Myo10 than accessible binding sites on the actin filament bundle. Live-cell movies reveal a dense cluster of over a hundred Myo10 molecules that initiates filopodial elongation. Hundreds of Myo10 molecules continue to accumulate during filopodial growth, but accumulation ceases when retraction begins. Rates of filopodial elongation, second-phase elongation, and retraction are inversely related to Myo10 quantities. Our estimates of Myo10 molecules in filopodia provide insight into the physics of packing Myo10, its cargo, and other filopodia-associated proteins in narrow membrane compartments. Our protocol provides a framework for future work analyzing Myo10 abundance and distribution upon perturbation.
Collapse
Affiliation(s)
- Julia Shangguan
- Department of Biochemistry and Molecular Biology, University of ChicagoChicagoUnited States
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The Institute for Biophysical Dynamics, University of ChicagoChicagoUnited States
| |
Collapse
|
2
|
Shangguan J, Rock RS. Hundreds of myosin 10s are pushed to the tips of filopodia and could cause traffic jams on actin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.26.546598. [PMID: 37425746 PMCID: PMC10327019 DOI: 10.1101/2023.06.26.546598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Myosin 10 (Myo10) is a motor protein known for its role in filopodia formation. Although Myo10-driven filopodial dynamics have been characterized, there is no information about the absolute number of Myo10 molecules during the filopodial lifecycle. To better understand molecular stoichiometries and packing restraints in filopodia, we measured Myo10 abundance in these structures. We combined SDS-PAGE densitometry with epifluorescence microscopy to quantitate HaloTag-labeled Myo10 in U2OS cells. About 6% of total intracellular Myo10 localizes to filopodia, where it enriches at opposite cellular ends. Hundreds of Myo10s are in a typical filopodium, and their distribution across filopodia is log-normal. Some filopodial tips even contain more Myo10 than accessible binding sites on the actin filament bundle. Live-cell movies reveal a dense cluster of over a hundred Myo10 molecules that initiates filopodial elongation. Hundreds of Myo10 molecules continue to accumulate during filopodial growth, but accumulation ceases when retraction begins. Rates of filopodial elongation, second-phase elongation, and retraction are inversely related to Myo10 quantities. Our estimates of Myo10 molecules in filopodia provide insight into the physics of packing Myo10, its cargo, and other filopodia-associated proteins in narrow membrane compartments. Our protocol provides a framework for future work analyzing Myo10 abundance and distribution upon perturbation.
Collapse
Affiliation(s)
- Julia Shangguan
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois, USA
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Wang L, Bu T, Wu X, Li L, Sun F, Cheng CY. Motor proteins, spermatogenesis and testis function. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:381-445. [PMID: 38960481 DOI: 10.1016/bs.apcsb.2024.03.003] [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: 07/05/2024]
Abstract
The role of motor proteins in supporting intracellular transports of vesicles and organelles in mammalian cells has been known for decades. On the other hand, the function of motor proteins that support spermatogenesis is also well established since the deletion of motor protein genes leads to subfertility and/or infertility. Furthermore, mutations and genetic variations of motor protein genes affect fertility in men, but also a wide range of developmental defects in humans including multiple organs besides the testis. In this review, we seek to provide a summary of microtubule and actin-dependent motor proteins based on earlier and recent findings in the field. Since these two cytoskeletons are polarized structures, different motor proteins are being used to transport cargoes to different ends of these cytoskeletons. However, their involvement in germ cell transport across the blood-testis barrier (BTB) and the epithelium of the seminiferous tubules remains relatively unknown. It is based on recent findings in the field, we have provided a hypothetical model by which motor proteins are being used to support germ cell transport across the BTB and the seminiferous epithelium during the epithelial cycle of spermatogenesis. In our discussion, we have highlighted the areas of research that deserve attention to bridge the gap of research in relating the function of motor proteins to spermatogenesis.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
4
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Mechanisms underlying Myosin 10's contribution to the maintenance of mitotic spindle bipolarity. Mol Biol Cell 2024; 35:ar14. [PMID: 38019611 PMCID: PMC10881153 DOI: 10.1091/mbc.e23-07-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Myosin 10 (Myo10) couples microtubules and integrin-based adhesions to movement along actin filaments via its microtubule-binding MyTH4 domain and integrin-binding FERM domain, respectively. Here we show that Myo10-depleted HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in Myo10-depleted MEFs and in Myo10-depleted HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates y-tubulin-positive acentriolar foci that serve as extra spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both microtubules and integrins to promote PCM/pole integrity. Conversely, Myo10 only needs interact with integrins to promote supernumerary centrosome clustering. Importantly, images of metaphase Halo-Myo10 knockin cells show that the myosin localizes exclusively to the spindle and the tips of adhesive retraction fibers. We conclude that Myo10 promotes PCM/pole integrity in part by interacting with spindle microtubules, and that it promotes supernumerary centrosome clustering by supporting retraction fiber-based cell adhesion, which likely serves to anchor the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
5
|
Hall ET, Dillard ME, Cleverdon ER, Zhang Y, Daly CA, Ansari SS, Wakefield R, Stewart DP, Pruett-Miller SM, Lavado A, Carisey AF, Johnson A, Wang YD, Selner E, Tanes M, Ryu YS, Robinson CG, Steinberg J, Ogden SK. Cytoneme signaling provides essential contributions to mammalian tissue patterning. Cell 2024; 187:276-293.e23. [PMID: 38171360 PMCID: PMC10842732 DOI: 10.1016/j.cell.2023.12.003] [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] [Received: 07/26/2022] [Revised: 09/06/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
During development, morphogens pattern tissues by instructing cell fate across long distances. Directly visualizing morphogen transport in situ has been inaccessible, so the molecular mechanisms ensuring successful morphogen delivery remain unclear. To tackle this longstanding problem, we developed a mouse model for compromised sonic hedgehog (SHH) morphogen delivery and discovered that endocytic recycling promotes SHH loading into signaling filopodia called cytonemes. We optimized methods to preserve in vivo cytonemes for advanced microscopy and show endogenous SHH localized to cytonemes in developing mouse neural tubes. Depletion of SHH from neural tube cytonemes alters neuronal cell fates and compromises neurodevelopment. Mutation of the filopodial motor myosin 10 (MYO10) reduces cytoneme length and density, which corrupts neuronal signaling activity of both SHH and WNT. Combined, these results demonstrate that cytoneme-based signal transport provides essential contributions to morphogen dispersion during mammalian tissue development and suggest MYO10 is a key regulator of cytoneme function.
Collapse
Affiliation(s)
- Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Miriam E Dillard
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elizabeth R Cleverdon
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yan Zhang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christina A Daly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shariq S Ansari
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Randall Wakefield
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel P Stewart
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alfonso Lavado
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Pediatric Neurological Disease Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alex F Carisey
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amanda Johnson
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Emma Selner
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael Tanes
- Center for In Vivo Imaging and Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Young Sang Ryu
- Center for In Vivo Imaging and Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Camenzind G Robinson
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jeffrey Steinberg
- Center for In Vivo Imaging and Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
6
|
Davoudi P, Do DN, Colombo S, Rathgeber B, Sargolzaei M, Plastow G, Wang Z, Hu G, Valipour S, Miar Y. Genome-wide association studies for economically important traits in mink using copy number variation. Sci Rep 2024; 14:24. [PMID: 38167844 PMCID: PMC10762091 DOI: 10.1038/s41598-023-50497-3] [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: 10/04/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Copy number variations (CNVs) are structural variants consisting of duplications and deletions of DNA segments, which are known to play important roles in the genetics of complex traits in livestock species. However, CNV-based genome-wide association studies (GWAS) have remained unexplored in American mink. Therefore, the purpose of the current study was to investigate the association between CNVs and complex traits in American mink. A CNV-based GWAS was performed with the ParseCNV2 software program using deregressed estimated breeding values of 27 traits as pseudophenotypes, categorized into traits of growth and feed efficiency, reproduction, pelt quality, and Aleutian disease tests. The study identified a total of 10,137 CNVs (6968 duplications and 3169 deletions) using the Affymetrix Mink 70K single nucleotide polymorphism (SNP) array in 2986 American mink. The association analyses identified 250 CNV regions (CNVRs) associated with at least one of the studied traits. These CNVRs overlapped with a total of 320 potential candidate genes, and among them, several genes have been known to be related to the traits such as ARID1B, APPL1, TOX, and GPC5 (growth and feed efficiency traits); GRM1, RNASE10, WNT3, WNT3A, and WNT9B (reproduction traits); MYO10, and LIMS1 (pelt quality traits); and IFNGR2, APEX1, UBE3A, and STX11 (Aleutian disease tests). Overall, the results of the study provide potential candidate genes that may regulate economically important traits and therefore may be used as genetic markers in mink genomic breeding programs.
Collapse
Affiliation(s)
- Pourya Davoudi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Stefanie Colombo
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Bruce Rathgeber
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Mehdi Sargolzaei
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Select Sires Inc., Plain City, OH, USA
| | - Graham Plastow
- Livestock Gentec, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Zhiquan Wang
- Livestock Gentec, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Guoyu Hu
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Shafagh Valipour
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Younes Miar
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada.
| |
Collapse
|
7
|
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
|
8
|
Brand CM, Kuang S, Gilbertson EN, McArthur E, Pollard KS, Webster TH, Capra JA. Sequence-based machine learning reveals 3D genome differences between bonobos and chimpanzees. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564272. [PMID: 37961120 PMCID: PMC10634871 DOI: 10.1101/2023.10.26.564272] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Phenotypic divergence between closely related species, including bonobos and chimpanzees (genus Pan), is largely driven by variation in gene regulation. The 3D structure of the genome mediates gene expression; however, genome folding differences in Pan are not well understood. Here, we apply machine learning to predict genome-wide 3D genome contact maps from DNA sequence for 56 bonobos and chimpanzees, encompassing all five extant lineages. We use a pairwise approach to estimate 3D divergence between individuals from the resulting contact maps in 4,420 1 Mb genomic windows. While most pairs were similar, ∼17% were predicted to be substantially divergent in genome folding. The most dissimilar maps were largely driven by single individuals with rare variants that produce unique 3D genome folding in a region. We also identified 89 genomic windows where bonobo and chimpanzee contact maps substantially diverged, including several windows harboring genes associated with traits implicated in Pan phenotypic divergence. We used in silico mutagenesis to identify 51 3D-modifying variants in these bonobo-chimpanzee divergent windows, finding that 34 or 66.67% induce genome folding changes via CTCF binding motif disruption. Our results reveal 3D genome variation at the population-level and identify genomic regions where changes in 3D folding may contribute to phenotypic differences in our closest living relatives.
Collapse
Affiliation(s)
- Colin M. Brand
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Shuzhen Kuang
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA
| | - Erin N. Gilbertson
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA
- Biomedical Informatics Graduate Program, University of California San Francisco, San Francisco, CA
| | - Evonne McArthur
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Katherine S. Pollard
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA
- Biomedical Informatics Graduate Program, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| | | | - John A. Capra
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
- Biomedical Informatics Graduate Program, University of California San Francisco, San Francisco, CA
| |
Collapse
|
9
|
Powis G, Meuillet EJ, Indarte M, Booher G, Kirkpatrick L. Pleckstrin Homology [PH] domain, structure, mechanism, and contribution to human disease. Biomed Pharmacother 2023; 165:115024. [PMID: 37399719 DOI: 10.1016/j.biopha.2023.115024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023] Open
Abstract
The pleckstrin homology [PH] domain is a structural fold found in more than 250 proteins making it the 11th most common domain in the human proteome. 25% of family members have more than one PH domain and some PH domains are split by one, or several other, protein domains although still folding to give functioning PH domains. We review mechanisms of PH domain activity, the role PH domain mutation plays in human disease including cancer, hyperproliferation, neurodegeneration, inflammation, and infection, and discuss pharmacotherapeutic approaches to regulate PH domain activity for the treatment of human disease. Almost half PH domain family members bind phosphatidylinositols [PIs] that attach the host protein to cell membranes where they interact with other membrane proteins to give signaling complexes or cytoskeleton scaffold platforms. A PH domain in its native state may fold over other protein domains thereby preventing substrate access to a catalytic site or binding with other proteins. The resulting autoinhibition can be released by PI binding to the PH domain, or by protein phosphorylation thus providing fine tuning of the cellular control of PH domain protein activity. For many years the PH domain was thought to be undruggable until high-resolution structures of human PH domains allowed structure-based design of novel inhibitors that selectively bind the PH domain. Allosteric inhibitors of the Akt1 PH domain have already been tested in cancer patients and for proteus syndrome, with several other PH domain inhibitors in preclinical development for treatment of other human diseases.
Collapse
Affiliation(s)
- Garth Powis
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA.
| | | | - Martin Indarte
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| | - Garrett Booher
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| | - Lynn Kirkpatrick
- PHusis Therapeutics Inc., 6019 Folsom Drive, La Jolla, CA 92037, USA
| |
Collapse
|
10
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.547992. [PMID: 37461640 PMCID: PMC10350058 DOI: 10.1101/2023.07.07.547992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
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 is present across several vertebrate and invertebrate systems, this may have significance for other long-range signaling mechanisms.
Collapse
Affiliation(s)
| | - Tyler Smith
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| | - Luke Cowart
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| | - Jennifer Zhumi
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| | - Alan Sherrod
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| | - Aidan Cahill
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| | - Ginger L Hunter
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| |
Collapse
|
11
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Myosin 10 uses its MyTH4 and FERM domains differentially to support two aspects of spindle pole biology required for mitotic spindle bipolarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545002. [PMID: 37398378 PMCID: PMC10312724 DOI: 10.1101/2023.06.15.545002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Myosin 10 (Myo10) has the ability to link actin filaments to integrin-based adhesions and to microtubules by virtue of its integrin-binding FERM domain and microtubule-binding MyTH4 domain, respectively. Here we used Myo10 knockout cells to define Myo10's contribution to the maintenance of spindle bipolarity, and complementation to quantitate the relative contributions of its MyTH4 and FERM domains. Myo10 knockout HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in knockout MEFs and knockout HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates γ-tubulin-positive acentriolar foci that serve as additional spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both integrins and microtubules to promote PCM/pole integrity. Conversely, Myo10's ability to promote the clustering of supernumerary centrosomes only requires that it interact with integrins. Importantly, images of Halo-Myo10 knock-in cells show that the myosin localizes exclusively within adhesive retraction fibers during mitosis. Based on these and other results, we conclude that Myo10 promotes PCM/pole integrity at a distance, and that it facilitates supernumerary centrosome clustering by promoting retraction fiber-based cell adhesion, which likely provides an anchor for the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
12
|
Crozet F, Letort G, Bulteau R, Da Silva C, Eichmuller A, Tortorelli AF, Blévinal J, Belle M, Dumont J, Piolot T, Dauphin A, Coulpier F, Chédotal A, Maître JL, Verlhac MH, Clarke HJ, Terret ME. Filopodia-like protrusions of adjacent somatic cells shape the developmental potential of oocytes. Life Sci Alliance 2023; 6:e202301963. [PMID: 36944420 PMCID: PMC10029974 DOI: 10.26508/lsa.202301963] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023] Open
Abstract
The oocyte must grow and mature before fertilization, thanks to a close dialogue with the somatic cells that surround it. Part of this communication is through filopodia-like protrusions, called transzonal projections (TZPs), sent by the somatic cells to the oocyte membrane. To investigate the contribution of TZPs to oocyte quality, we impaired their structure by generating a full knockout mouse of the TZP structural component myosin-X (MYO10). Using spinning disk and super-resolution microscopy combined with a machine-learning approach to phenotype oocyte morphology, we show that the lack of Myo10 decreases TZP density during oocyte growth. Reduction in TZPs does not prevent oocyte growth but impairs oocyte-matrix integrity. Importantly, we reveal by transcriptomic analysis that gene expression is altered in TZP-deprived oocytes and that oocyte maturation and subsequent early embryonic development are partially affected, effectively reducing mouse fertility. We propose that TZPs play a role in the structural integrity of the germline-somatic complex, which is essential for regulating gene expression in the oocyte and thus its developmental potential.
Collapse
Affiliation(s)
- Flora Crozet
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Paris, France
| | - Gaëlle Letort
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Paris, France
| | - Rose Bulteau
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Christelle Da Silva
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Adrien Eichmuller
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | - Anna Francesca Tortorelli
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | | | - Morgane Belle
- Institut de la Vision, UMRS968/UMR7210/UM80, Paris, France
| | - Julien Dumont
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Tristan Piolot
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Aurélien Dauphin
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | - Fanny Coulpier
- Genomics Core Facility, Institut de Biologie de l'ENS, Département de biologie, Ecole normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Alain Chédotal
- Institut de la Vision, UMRS968/UMR7210/UM80, Paris, France
| | - Jean-Léon Maître
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | - Marie-Hélène Verlhac
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada
| | - Marie-Emilie Terret
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
13
|
Hein JI, Scholz J, Körber S, Kaufmann T, Faix J. Unleashed Actin Assembly in Capping Protein-Deficient B16-F1 Cells Enables Identification of Multiple Factors Contributing to Filopodium Formation. Cells 2023; 12:cells12060890. [PMID: 36980231 PMCID: PMC10047565 DOI: 10.3390/cells12060890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Filopodia are dynamic, finger-like actin-filament bundles that overcome membrane tension by forces generated through actin polymerization at their tips to allow extension of these structures a few microns beyond the cell periphery. Actin assembly of these protrusions is regulated by accessory proteins including heterodimeric capping protein (CP) or Ena/VASP actin polymerases to either terminate or promote filament growth. Accordingly, the depletion of CP in B16-F1 melanoma cells was previously shown to cause an explosive formation of filopodia. In Ena/VASP-deficient cells, CP depletion appeared to result in ruffling instead of inducing filopodia, implying that Ena/VASP proteins are absolutely essential for filopodia formation. However, this hypothesis was not yet experimentally confirmed. Methods: Here, we used B16-F1 cells and CRISPR/Cas9 technology to eliminate CP either alone or in combination with Ena/VASP or other factors residing at filopodia tips, followed by quantifications of filopodia length and number. Results: Unexpectedly, we find massive formations of filopodia even in the absence of CP and Ena/VASP proteins. Notably, combined inactivation of Ena/VASP, unconventional myosin-X and the formin FMNL3 was required to markedly impair filopodia formation in CP-deficient cells. Conclusions: Taken together, our results reveal that, besides Ena/VASP proteins, numerous other factors contribute to filopodia formation.
Collapse
Affiliation(s)
| | | | | | | | - Jan Faix
- Correspondence: ; Tel.: +49-511-532-2928
| |
Collapse
|
14
|
Popović A, Miihkinen M, Ghimire S, Saup R, Grönloh MLB, Ball NJ, Goult BT, Ivaska J, Jacquemet G. Myosin-X recruits lamellipodin to filopodia tips. J Cell Sci 2023; 136:293507. [PMID: 36861887 PMCID: PMC10022686 DOI: 10.1242/jcs.260574] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/12/2023] [Indexed: 03/03/2023] Open
Abstract
Myosin-X (MYO10), a molecular motor localizing to filopodia, is thought to transport various cargo to filopodia tips, modulating filopodia function. However, only a few MYO10 cargoes have been described. Here, using GFP-Trap and BioID approaches combined with mass spectrometry, we identified lamellipodin (RAPH1) as a novel MYO10 cargo. We report that the FERM domain of MYO10 is required for RAPH1 localization and accumulation at filopodia tips. Previous studies have mapped the RAPH1 interaction domain for adhesome components to its talin-binding and Ras-association domains. Surprisingly, we find that the RAPH1 MYO10-binding site is not within these domains. Instead, it comprises a conserved helix located just after the RAPH1 pleckstrin homology domain with previously unknown functions. Functionally, RAPH1 supports MYO10 filopodia formation and stability but is not required to activate integrins at filopodia tips. Taken together, our data indicate a feed-forward mechanism whereby MYO10 filopodia are positively regulated by MYO10-mediated transport of RAPH1 to the filopodium tip.
Collapse
Affiliation(s)
- Ana Popović
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Mitro Miihkinen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Sujan Ghimire
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Rafael Saup
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Max L B Grönloh
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Neil J Ball
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Department of Life Technologies, University of Turku, 20520 Turku, Finland.,InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Western Finnish Cancer Center (FICAN West), University of Turku, 20520 Turku, Finland
| | - Guillaume Jacquemet
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland.,InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Turku Bioimaging, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| |
Collapse
|
15
|
Pokrant T, Hein JI, Körber S, Disanza A, Pich A, Scita G, Rottner K, Faix J. Ena/VASP clustering at microspike tips involves lamellipodin but not I-BAR proteins, and absolutely requires unconventional myosin-X. Proc Natl Acad Sci U S A 2023; 120:e2217437120. [PMID: 36598940 PMCID: PMC9926217 DOI: 10.1073/pnas.2217437120] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023] Open
Abstract
Sheet-like membrane protrusions at the leading edge, termed lamellipodia, drive 2D-cell migration using active actin polymerization. Microspikes comprise actin-filament bundles embedded within lamellipodia, but the molecular mechanisms driving their formation and their potential functional relevance have remained elusive. Microspike formation requires the specific activity of clustered Ena/VASP proteins at their tips to enable processive actin assembly in the presence of capping protein, but the factors and mechanisms mediating Ena/VASP clustering are poorly understood. Systematic analyses of B16-F1 melanoma mutants lacking potential candidate proteins revealed that neither inverse BAR-domain proteins, nor lamellipodin or Abi is essential for clustering, although they differentially contribute to lamellipodial VASP accumulation. In contrast, unconventional myosin-X (MyoX) identified here as proximal to VASP was obligatory for Ena/VASP clustering and microspike formation. Interestingly, and despite the invariable distribution of other relevant marker proteins, the width of lamellipodia in MyoX-KO mutants was significantly reduced as compared with B16-F1 control, suggesting that microspikes contribute to lamellipodium stability. Consistently, MyoX removal caused marked defects in protrusion and random 2D-cell migration. Strikingly, Ena/VASP-deficiency also uncoupled MyoX cluster dynamics from actin assembly in lamellipodia, establishing their tight functional association in microspike formation.
Collapse
Affiliation(s)
- Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| | - Jens Ingo Hein
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| | - Andrea Disanza
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139Milan, Italy
| | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School, 30625Hannover, Germany
| | - Giorgio Scita
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, 20139 Milan, Italy
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, 38124Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| |
Collapse
|
16
|
Pokrant T, Hein JI, Körber S, Disanza A, Pich A, Scita G, Rottner K, Faix J. Ena/VASP clustering at microspike tips involves lamellipodin but not I-BAR proteins, and absolutely requires unconventional myosin-X. Proc Natl Acad Sci U S A 2023. [PMID: 36598940 DOI: 10.1101/2022.05.12.491613] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Sheet-like membrane protrusions at the leading edge, termed lamellipodia, drive 2D-cell migration using active actin polymerization. Microspikes comprise actin-filament bundles embedded within lamellipodia, but the molecular mechanisms driving their formation and their potential functional relevance have remained elusive. Microspike formation requires the specific activity of clustered Ena/VASP proteins at their tips to enable processive actin assembly in the presence of capping protein, but the factors and mechanisms mediating Ena/VASP clustering are poorly understood. Systematic analyses of B16-F1 melanoma mutants lacking potential candidate proteins revealed that neither inverse BAR-domain proteins, nor lamellipodin or Abi is essential for clustering, although they differentially contribute to lamellipodial VASP accumulation. In contrast, unconventional myosin-X (MyoX) identified here as proximal to VASP was obligatory for Ena/VASP clustering and microspike formation. Interestingly, and despite the invariable distribution of other relevant marker proteins, the width of lamellipodia in MyoX-KO mutants was significantly reduced as compared with B16-F1 control, suggesting that microspikes contribute to lamellipodium stability. Consistently, MyoX removal caused marked defects in protrusion and random 2D-cell migration. Strikingly, Ena/VASP-deficiency also uncoupled MyoX cluster dynamics from actin assembly in lamellipodia, establishing their tight functional association in microspike formation.
Collapse
Affiliation(s)
- Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Ingo Hein
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Andrea Disanza
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
| | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Giorgio Scita
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, 20139 Milan, Italy
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
17
|
Douceau S, Deutsch Guerrero T, Ferent J. Establishing Hedgehog Gradients during Neural Development. Cells 2023; 12:225. [PMID: 36672161 PMCID: PMC9856818 DOI: 10.3390/cells12020225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/07/2023] Open
Abstract
A morphogen is a signaling molecule that induces specific cellular responses depending on its local concentration. The concept of morphogenic gradients has been a central paradigm of developmental biology for decades. Sonic Hedgehog (Shh) is one of the most important morphogens that displays pleiotropic functions during embryonic development, ranging from neuronal patterning to axon guidance. It is commonly accepted that Shh is distributed in a gradient in several tissues from different origins during development; however, how these gradients are formed and maintained at the cellular and molecular levels is still the center of a great deal of research. In this review, we first explored all of the different sources of Shh during the development of the nervous system. Then, we detailed how these sources can distribute Shh in the surrounding tissues via a variety of mechanisms. Finally, we addressed how disrupting Shh distribution and gradients can induce severe neurodevelopmental disorders and cancers. Although the concept of gradient has been central in the field of neurodevelopment since the fifties, we also describe how contemporary leading-edge techniques, such as organoids, can revisit this classical model.
Collapse
Affiliation(s)
- Sara Douceau
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| | - Tanya Deutsch Guerrero
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| | - Julien Ferent
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| |
Collapse
|
18
|
Washington-Hughes CL, Roy S, Seneviratne HK, Karuppagounder SS, Morel Y, Jones JW, Zak A, Xiao T, Boronina TN, Cole RN, Bumpus NN, Chang CJ, Dawson TM, Lutsenko S. Atp7b-dependent choroid plexus dysfunction causes transient copper deficit and metabolic changes in the developing mouse brain. PLoS Genet 2023; 19:e1010558. [PMID: 36626371 PMCID: PMC9870141 DOI: 10.1371/journal.pgen.1010558] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 01/23/2023] [Accepted: 12/07/2022] [Indexed: 01/11/2023] Open
Abstract
Copper (Cu) has a multifaceted role in brain development, function, and metabolism. Two homologous Cu transporters, Atp7a (Menkes disease protein) and Atp7b (Wilson disease protein), maintain Cu homeostasis in the tissue. Atp7a mediates Cu entry into the brain and activates Cu-dependent enzymes, whereas the role of Atp7b is less clear. We show that during postnatal development Atp7b is necessary for normal morphology and function of choroid plexus (ChPl). Inactivation of Atp7b causes reorganization of ChPl' cytoskeleton and cell-cell contacts, loss of Slc31a1 from the apical membrane, and a decrease in the length and number of microvilli and cilia. In ChPl lacking Atp7b, Atp7a is upregulated but remains intracellular, which limits Cu transport into the brain and results in significant Cu deficit, which is reversed only in older animals. Cu deficiency is associated with down-regulation of Atp7a in locus coeruleus and catecholamine imbalance, despite normal expression of dopamine-β-hydroxylase. In addition, there are notable changes in the brain lipidome, which can be attributed to inhibition of diacylglyceride-to-phosphatidylethanolamine conversion. These results identify the new role for Atp7b in developing brain and identify metabolic changes that could be exacerbated by Cu chelation therapy.
Collapse
Affiliation(s)
| | - Shubhrajit Roy
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Herana Kamal Seneviratne
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Senthilkumar S. Karuppagounder
- Neurodegeneration and Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yulemni Morel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Alex Zak
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tong Xiao
- Department of Chemistry, University of California Berkeley, California, United States of America
| | - Tatiana N. Boronina
- Department of Biological Chemistry Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert N. Cole
- Department of Biological Chemistry Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Namandjé N. Bumpus
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher J. Chang
- Department of Chemistry, University of California Berkeley, California, United States of America
- Department of Molecular and Cell Biology, University of California Berkeley, California
- Helen Wills Neuroscience Institute, University of California Berkeley, California
| | - Ted M. Dawson
- Neurodegeneration and Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland; United States of America
| | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Li R, Sun Y, Cui R, Zhang X. Comprehensive Transcriptome Analysis of Different Skin Colors to Evaluate Genes Related to the Production of Pigment in Celestial Goldfish. BIOLOGY 2022; 12:biology12010007. [PMID: 36671700 PMCID: PMC9854719 DOI: 10.3390/biology12010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Skin color is an important phenotypic feature of vertebrate fitness under natural conditions. Celestial goldfish, a common goldfish breed in China, mainly shows three kinds of skin colors including white, yellow and brown. However, the molecular genetic basis of this phenotype is still unclear. In this study, high-throughput sequencing was carried out on the back skin tissues of celestial goldfish with different skin colors. About 58.46 Gb of original data were generated, filtered and blasted, and 74,297 mRNAs were obtained according to the reference transcriptome. A total of 4653 differentially expressed genes were screened out among the brown, yellow and white groups, and the expression of melanogenesis related genes in brown goldfish was significantly higher than the other two groups. There are 19 common differentially expressed genes among three groups, of which eight genes are related to pigment production, including tyrp1a, slc2a11b, mlana, gch2, loc113060382, loc113079820, loc113068772 and loc113059134. RT-qPCR verified that the expression patterns of randomly selected differentially expressed transcripts were highly consistent with those obtained by RNA sequencing. GO and KEGG annotation revealed that these differentially expressed genes were mostly enriched in pathways of the production of pigment, including melanogenesis, tyrosine metabolism, Wnt signaling pathway, MAPK signaling pathway etc. These results indicated that the external characteristics of goldfish are consistent with the analysis results at transcriptome level. The results of this study will lay a foundation for further study on the expression characteristics and gene network analysis of pigment related genes.
Collapse
Affiliation(s)
| | | | - Ran Cui
- Correspondence: (R.C.); (X.Z.)
| | | |
Collapse
|
20
|
Omelchenko T. Cellular protrusions in 3D: Orchestrating early mouse embryogenesis. Semin Cell Dev Biol 2022; 129:63-74. [PMID: 35577698 DOI: 10.1016/j.semcdb.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 12/26/2022]
Abstract
Cellular protrusions generated by the actin cytoskeleton are central to the process of building the body of the embryo. Problems with cellular protrusions underlie human diseases and syndromes, including implantation defects and pregnancy loss, congenital birth defects, and cancer. Cells use protrusive activity together with actin-myosin contractility to create an ordered body shape of the embryo. Here, I review how actin-rich protrusions are used by two major morphological cell types, epithelial and mesenchymal cells, during collective cell migration to sculpt the mouse embryo body. Pre-gastrulation epithelial collective migration of the anterior visceral endoderm is essential for establishing the anterior-posterior body axis. Gastrulation mesenchymal collective migration of the mesoderm wings is crucial for body elongation, and somite and heart formation. Analysis of mouse mutants with disrupted cellular protrusions revealed the key role of protrusions in embryonic morphogenesis and embryo survival. Recent technical approaches have allowed examination of the mechanisms that control cell and tissue movements in vivo in the complex 3D microenvironment of living mouse embryos. Advancing our understanding of protrusion-driven morphogenesis should provide novel insights into human developmental disorders and cancer metastasis.
Collapse
Affiliation(s)
- Tatiana Omelchenko
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York 10065, USA.
| |
Collapse
|
21
|
Granados-Aparici S, Volodarsky-Perel A, Yang Q, Anam S, Tulandi T, Buckett W, Son WY, Younes G, Chung JT, Jin S, Terret MÉ, Clarke HJ. MYO10 promotes transzonal projection (TZP)-dependent germ line-somatic contact during mammalian folliculogenesis. Biol Reprod 2022; 107:474-487. [PMID: 35470858 PMCID: PMC9382396 DOI: 10.1093/biolre/ioac078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/17/2022] [Accepted: 04/15/2022] [Indexed: 11/12/2022] Open
Abstract
Granulosa cells of growing ovarian follicles elaborate filopodia-like structures termed transzonal projections (TZPs) that supply the enclosed oocyte with factors essential for its development. Little is known, however, of the mechanisms underlying the generation of TZPs. We show in mouse and human that filopodia, defined by an actin backbone, emerge from granulosa cells in early-stage primary follicles and that actin-rich TZPs become detectable as soon as a space corresponding to the zona pellucida appears. mRNA encoding Myosin10 (MYO10), a motor protein that accumulates at the base and tips of filopodia and has been implicated in their initiation and elongation, is present in granulosa cells and oocytes of growing follicles. MYO10 protein accumulates in foci located mainly between the oocyte and innermost layer of granulosa cells, where it co-localizes with actin. In both mouse and human, the number of MYO10 foci increases as oocytes grow, corresponding to the increase in the number of actin-TZPs. RNAi-mediated depletion of MYO10 in cultured mouse granulosa cell-oocyte complexes is associated with a 52% reduction in the number of MYO10 foci and a 28% reduction in the number of actin-TZPs. Moreover, incubation of cumulus-oocyte complexes in the presence of epidermal growth factor, which triggers a 93% reduction in the number of actin-TZPs, is associated with a 55% reduction in the number of MYO10 foci. These results suggest that granulosa cells possess an ability to elaborate filopodia, which when directed towards the oocyte become actin-TZPs, and that MYO10 increases the efficiency of formation or maintenance of actin-TZPs.
Collapse
Affiliation(s)
- Sofia Granados-Aparici
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.,Research Institute of the McGill University Health Center, Montreal, Canada
| | - Alexander Volodarsky-Perel
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.,Research Institute of the McGill University Health Center, Montreal, Canada
| | - Qin Yang
- Research Institute of the McGill University Health Center, Montreal, Canada
| | - Sibat Anam
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Togas Tulandi
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.,Research Institute of the McGill University Health Center, Montreal, Canada
| | - William Buckett
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.,Research Institute of the McGill University Health Center, Montreal, Canada
| | - Weon-Young Son
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada
| | - Grace Younes
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.,Research Institute of the McGill University Health Center, Montreal, Canada
| | - Jin-Tae Chung
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada
| | - Shaoguang Jin
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada
| | | | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.,Research Institute of the McGill University Health Center, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
22
|
Mayca Pozo F, Geng X, Tamagno I, Jackson MW, Heimsath EG, Hammer JA, Cheney RE, Zhang Y. MYO10 drives genomic instability and inflammation in cancer. SCIENCE ADVANCES 2021; 7:eabg6908. [PMID: 34524844 PMCID: PMC8443186 DOI: 10.1126/sciadv.abg6908] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/26/2021] [Indexed: 05/29/2023]
Abstract
Genomic instability is a hallmark of human cancer; yet the underlying mechanisms remain poorly understood. Here, we report that the cytoplasmic unconventional Myosin X (MYO10) regulates genome stability, through which it mediates inflammation in cancer. MYO10 is an unstable protein that undergoes ubiquitin-conjugating enzyme H7 (UbcH7)/β-transducin repeat containing protein 1 (β-TrCP1)–dependent degradation. MYO10 is upregulated in both human and mouse tumors and its expression level predisposes tumor progression and response to immune therapy. Overexpressing MYO10 increased genomic instability, elevated the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING)–dependent inflammatory response, and accelerated tumor growth in mice. Conversely, depletion of MYO10 ameliorated genomic instability and reduced the inflammation signaling. Further, inhibiting inflammation or disrupting Myo10 significantly suppressed the growth of both human and mouse breast tumors in mice. Our data suggest that MYO10 promotes tumor progression through inducing genomic instability, which, in turn, creates an immunogenic environment for immune checkpoint blockades.
Collapse
Affiliation(s)
- Franklin Mayca Pozo
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xinran Geng
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ilaria Tamagno
- Department of Pathology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mark W. Jackson
- Department of Pathology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ernest G. Heimsath
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Youwei Zhang
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
23
|
Hammers DW, Hart CC, Matheny MK, Heimsath EG, Lee YI, Hammer JA, Cheney RE, Sweeney HL. Filopodia powered by class x myosin promote fusion of mammalian myoblasts. eLife 2021; 10:e72419. [PMID: 34519272 PMCID: PMC8500716 DOI: 10.7554/elife.72419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/13/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle fibers are multinucleated cellular giants formed by the fusion of mononuclear myoblasts. Several molecules involved in myoblast fusion have been discovered, and finger-like projections coincident with myoblast fusion have also been implicated in the fusion process. The role of these cellular projections in muscle cell fusion was investigated herein. We demonstrate that these projections are filopodia generated by class X myosin (Myo10), an unconventional myosin motor protein specialized for filopodia. We further show that Myo10 is highly expressed by differentiating myoblasts, and Myo10 ablation inhibits both filopodia formation and myoblast fusion in vitro. In vivo, Myo10 labels regenerating muscle fibers associated with Duchenne muscular dystrophy and acute muscle injury. In mice, conditional loss of Myo10 from muscle-resident stem cells, known as satellite cells, severely impairs postnatal muscle regeneration. Furthermore, the muscle fusion proteins Myomaker and Myomixer are detected in myoblast filopodia. These data demonstrate that Myo10-driven filopodia facilitate multinucleated mammalian muscle formation.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Fusion
- Cell Line
- Cell Proliferation
- Disease Models, Animal
- Humans
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle Development
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- Myosins/genetics
- Myosins/metabolism
- Pseudopodia/genetics
- Pseudopodia/metabolism
- Regeneration
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Time Factors
- Mice
Collapse
Affiliation(s)
- David W Hammers
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Cora C Hart
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Michael K Matheny
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Ernest G Heimsath
- Department of Cell Biology & Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of MedicineChapel HillUnited States
| | - Young il Lee
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung and Blood InstituteBethesdaUnited States
| | - Richard E Cheney
- Department of Cell Biology & Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of MedicineChapel HillUnited States
| | - H Lee Sweeney
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| |
Collapse
|
24
|
Abstract
Filopodia, microvilli and stereocilia represent an important group of plasma membrane protrusions. These specialized projections are supported by parallel bundles of actin filaments and have critical roles in sensing the external environment, increasing cell surface area, and acting as mechanosensors. While actin-associated proteins are essential for actin-filament elongation and bundling in these protrusions, myosin motors have a surprising role in the formation and extension of filopodia and stereocilia and in the organization of microvilli. Actin regulators and specific myosins collaborate in controlling the length of these structures. Myosins can transport cargoes along the length of these protrusions, and, in the case of stereocilia and microvilli, interactions with adaptors and cargoes can also serve to anchor adhesion receptors to the actin-rich core via functionally conserved motor-adaptor complexes. This review highlights recent progress in understanding the diverse roles myosins play in filopodia, microvilli and stereocilia.
Collapse
Affiliation(s)
- Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75005 Paris, France.
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
Maniou E, Staddon MF, Marshall AR, Greene NDE, Copp AJ, Banerjee S, Galea GL. Hindbrain neuropore tissue geometry determines asymmetric cell-mediated closure dynamics in mouse embryos. Proc Natl Acad Sci U S A 2021; 118:e2023163118. [PMID: 33941697 PMCID: PMC8126771 DOI: 10.1073/pnas.2023163118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gap closure is a common morphogenetic process. In mammals, failure to close the embryonic hindbrain neuropore (HNP) gap causes fatal anencephaly. We observed that surface ectoderm cells surrounding the mouse HNP assemble high-tension actomyosin purse strings at their leading edge and establish the initial contacts across the embryonic midline. Fibronectin and laminin are present, and tensin 1 accumulates in focal adhesion-like puncta at this leading edge. The HNP gap closes asymmetrically, faster from its rostral than caudal end, while maintaining an elongated aspect ratio. Cell-based physical modeling identifies two closure mechanisms sufficient to account for tissue-level HNP closure dynamics: purse-string contraction and directional cell motion implemented through active crawling. Combining both closure mechanisms hastens gap closure and produces a constant rate of gap shortening. Purse-string contraction reduces, whereas crawling increases gap aspect ratio, and their combination maintains it. Closure rate asymmetry can be explained by asymmetric embryo tissue geometry, namely a narrower rostral gap apex, whereas biomechanical tension inferred from laser ablation is equivalent at the gaps' rostral and caudal closure points. At the cellular level, the physical model predicts rearrangements of cells at the HNP rostral and caudal extremes as the gap shortens. These behaviors are reproducibly live imaged in mouse embryos. Thus, mammalian embryos coordinate cellular- and tissue-level mechanics to achieve this critical gap closure event.
Collapse
Affiliation(s)
- Eirini Maniou
- Department of Developmental Biology and Cancer Researching and Teaching, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom
| | - Michael F Staddon
- Department of Physics and Astronomy, University College London, WC1E 6BT London, United Kingdom
| | - Abigail R Marshall
- Department of Developmental Biology and Cancer Researching and Teaching, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom
| | - Nicholas D E Greene
- Department of Developmental Biology and Cancer Researching and Teaching, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom
| | - Andrew J Copp
- Department of Developmental Biology and Cancer Researching and Teaching, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom
| | | | - Gabriel L Galea
- Department of Developmental Biology and Cancer Researching and Teaching, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom;
- Department of Comparative Bioveterinary Sciences, Royal Veterinary College, NW1 0TU London, United Kingdom
| |
Collapse
|
26
|
Hall ET, Dillard ME, Stewart DP, Zhang Y, Wagner B, Levine RM, Pruett-Miller SM, Sykes A, Temirov J, Cheney RE, Mori M, Robinson CG, Ogden SK. Cytoneme delivery of Sonic Hedgehog from ligand-producing cells requires Myosin 10 and a Dispatched-BOC/CDON co-receptor complex. eLife 2021; 10:61432. [PMID: 33570491 PMCID: PMC7968926 DOI: 10.7554/elife.61432] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Morphogens function in concentration-dependent manners to instruct cell fate during tissue patterning. The cytoneme morphogen transport model posits that specialized filopodia extend between morphogen-sending and responding cells to ensure that appropriate signaling thresholds are achieved. How morphogens are transported along and deployed from cytonemes, how quickly a cytoneme-delivered, receptor-dependent signal is initiated, and whether these processes are conserved across phyla are not known. Herein, we reveal that the actin motor Myosin 10 promotes vesicular transport of Sonic Hedgehog (SHH) morphogen in mouse cell cytonemes, and that SHH morphogen gradient organization is altered in neural tubes of Myo10-/- mice. We demonstrate that cytoneme-mediated deposition of SHH onto receiving cells induces a rapid, receptor-dependent signal response that occurs within seconds of ligand delivery. This activity is dependent upon a novel Dispatched (DISP)-BOC/CDON co-receptor complex that functions in ligand-producing cells to promote cytoneme occurrence and facilitate ligand delivery for signal activation.
Collapse
Affiliation(s)
- Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Miriam E Dillard
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Daniel P Stewart
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Yan Zhang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Ben Wagner
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, United States
| | - Rachel M Levine
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - April Sykes
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, United States
| | - Jamshid Temirov
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Richard E Cheney
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, United States
| | - Motomi Mori
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, United States
| | - Camenzind G Robinson
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, United States
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| |
Collapse
|
27
|
Zhang Z, Denans N, Liu Y, Zhulyn O, Rosenblatt HD, Wernig M, Barna M. Optogenetic manipulation of cellular communication using engineered myosin motors. Nat Cell Biol 2021; 23:198-208. [PMID: 33526902 PMCID: PMC7880895 DOI: 10.1038/s41556-020-00625-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
Cells achieve highly efficient and accurate communication through cellular projections such as neurites and filopodia, yet there is a lack of genetically encoded tools that can selectively manipulate their composition and dynamics. Here, we present a versatile optogenetic toolbox of artificial multi-headed myosin motors that can move bidirectionally within long cellular extensions and allow for the selective transport of GFP-tagged cargo with light. Utilizing these engineered motors, we could transport bulky transmembrane receptors and organelles as well as actin remodellers to control the dynamics of both filopodia and neurites. Using an optimized in vivo imaging scheme, we further demonstrate that, upon limb amputation in axolotls, a complex array of filopodial extensions is formed. We selectively modulated these filopodial extensions and showed that they re-establish a Sonic Hedgehog signalling gradient during regeneration. Considering the ubiquitous existence of actin-based extensions, this toolbox shows the potential to manipulate cellular communication with unprecedented accuracy.
Collapse
Affiliation(s)
- Zijian Zhang
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Nicolas Denans
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Yingfei Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Olena Zhulyn
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Hannah D Rosenblatt
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Maria Barna
- Department of Developmental Biology, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
28
|
Wu S, Li H, Wang L, Mak N, Wu X, Ge R, Sun F, Cheng CY. Motor Proteins and Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:131-159. [PMID: 34453735 DOI: 10.1007/978-3-030-77779-1_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Unlike the intermediate filament- and septin-based cytoskeletons which are apolar structures, the microtubule (MT) and actin cytoskeletons are polarized structures in mammalian cells and tissues including the testis, most notable in Sertoli cells. In the testis, these cytoskeletons that stretch across the epithelium of seminiferous tubules and lay perpendicular to the basement membrane of tunica propria serve as tracks for corresponding motor proteins to support cellular cargo transport. These cargoes include residual bodies, phagosomes, endocytic vesicles and most notably developing spermatocytes and haploid spermatids which lack the ultrastructures of motile cells (e.g., lamellipodia, filopodia). As such, these developing germ cells require the corresponding motor proteins to facilitate their transport across the seminiferous epithelium during the epithelial cycle of spermatogenesis. Due to the polarized natures of these cytoskeletons with distinctive plus (+) and minus (-) end, directional cargo transport can take place based on the use of corresponding actin- or MT-based motor proteins. These include the MT-based minus (-) end directed motor proteins: dyneins, and the plus (+) end directed motor proteins: kinesins, as well as the actin-based motor proteins: myosins, many of which are plus (+) end directed but a few are also minus (-) end directed motor proteins. Recent studies have shown that these motor proteins are essential to support spermatogenesis. In this review, we briefly summarize and evaluate these recent findings so that this information will serve as a helpful guide for future studies and for planning functional experiments to better understand their role mechanistically in supporting spermatogenesis.
Collapse
Affiliation(s)
- Siwen Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA
| | - Huitao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA
| | - Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA.,Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Nathan Mak
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA
| | - Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China
| | - Fei Sun
- Sir Run Run Shaw Hospital (SRRSH), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - C Yan Cheng
- Sir Run Run Shaw Hospital (SRRSH), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Kenchappa RS, Mistriotis P, Wisniewski E, Bhattacharya S, Kulkarni T, West R, Luu A, Conlon M, Heimsath E, Crish JF, Picariello HS, Dovas A, Zarco N, Lara-Velazquez M, Quiñones-Hinojosa A, Hammer JA, Mukhopadhyay D, Cheney RE, Konstantopoulos K, Canoll P, Rosenfeld SS. Myosin 10 Regulates Invasion, Mitosis, and Metabolic Signaling in Glioblastoma. iScience 2020; 23:101802. [PMID: 33299973 PMCID: PMC7702012 DOI: 10.1016/j.isci.2020.101802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/18/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
Invasion and proliferation are defining phenotypes of cancer, and in glioblastoma blocking one stimulates the other, implying that effective therapy must inhibit both, ideally through a single target that is also dispensable for normal tissue function. The molecular motor myosin 10 meets these criteria. Myosin 10 knockout mice can survive to adulthood, implying that normal cells can compensate for its loss; its deletion impairs invasion, slows proliferation, and prolongs survival in murine models of glioblastoma. Myosin 10 deletion also enhances tumor dependency on the DNA damage and the metabolic stress responses and induces synthetic lethality when combined with inhibitors of these processes. Our results thus demonstrate that targeting myosin 10 is active against glioblastoma by itself, synergizes with other clinically available therapeutics, may have acceptable side effects in normal tissues, and has potential as a heretofore unexplored therapeutic approach for this disease.
Collapse
Affiliation(s)
- Rajappa S. Kenchappa
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Emily Wisniewski
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Santanu Bhattacharya
- Departments of Biochemistry and Molecular Biology and Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tanmay Kulkarni
- Departments of Biochemistry and Molecular Biology and Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rita West
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Amanda Luu
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Meghan Conlon
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Ernest Heimsath
- Department of Cell Biology and Physiology, and the Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - James F. Crish
- Department of Cancer Biology, Lerner Research Institute, Cleveland, OH 44106, USA
| | - Hannah S. Picariello
- Department of Cancer Biology, Lerner Research Institute, Cleveland, OH 44106, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Alfredo Quiñones-Hinojosa
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Debrabrata Mukhopadhyay
- Departments of Biochemistry and Molecular Biology and Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, and the Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Steven S. Rosenfeld
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
30
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
31
|
Blangy A, Bompard G, Guerit D, Marie P, Maurin J, Morel A, Vives V. The osteoclast cytoskeleton - current understanding and therapeutic perspectives for osteoporosis. J Cell Sci 2020; 133:133/13/jcs244798. [PMID: 32611680 DOI: 10.1242/jcs.244798] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Osteoclasts are giant multinucleated myeloid cells specialized for bone resorption, which is essential for the preservation of bone health throughout life. The activity of osteoclasts relies on the typical organization of osteoclast cytoskeleton components into a highly complex structure comprising actin, microtubules and other cytoskeletal proteins that constitutes the backbone of the bone resorption apparatus. The development of methods to differentiate osteoclasts in culture and manipulate them genetically, as well as improvements in cell imaging technologies, has shed light onto the molecular mechanisms that control the structure and dynamics of the osteoclast cytoskeleton, and thus the mechanism of bone resorption. Although essential for normal bone physiology, abnormal osteoclast activity can cause bone defects, in particular their hyper-activation is commonly associated with many pathologies, hormonal imbalance and medical treatments. Increased bone degradation by osteoclasts provokes progressive bone loss, leading to osteoporosis, with the resulting bone frailty leading to fractures, loss of autonomy and premature death. In this context, the osteoclast cytoskeleton has recently proven to be a relevant therapeutic target for controlling pathological bone resorption levels. Here, we review the present knowledge on the regulatory mechanisms of the osteoclast cytoskeleton that control their bone resorption activity in normal and pathological conditions.
Collapse
Affiliation(s)
- Anne Blangy
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Guillaume Bompard
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - David Guerit
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Pauline Marie
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Justine Maurin
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Anne Morel
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Virginie Vives
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| |
Collapse
|
32
|
Summerbell ER, Mouw JK, Bell JSK, Knippler CM, Pedro B, Arnst JL, Khatib TO, Commander R, Barwick BG, Konen J, Dwivedi B, Seby S, Kowalski J, Vertino PM, Marcus AI. Epigenetically heterogeneous tumor cells direct collective invasion through filopodia-driven fibronectin micropatterning. SCIENCE ADVANCES 2020; 6:eaaz6197. [PMID: 32832657 PMCID: PMC7439406 DOI: 10.1126/sciadv.aaz6197] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 06/11/2020] [Indexed: 06/11/2023]
Abstract
Tumor heterogeneity drives disease progression, treatment resistance, and patient relapse, yet remains largely underexplored in invasion and metastasis. Here, we investigated heterogeneity within collective cancer invasion by integrating DNA methylation and gene expression analysis in rare purified lung cancer leader and follower cells. Our results showed global DNA methylation rewiring in leader cells and revealed the filopodial motor MYO10 as a critical gene at the intersection of epigenetic heterogeneity and three-dimensional (3D) collective invasion. We further identified JAG1 signaling as a previously unknown upstream activator of MYO10 expression in leader cells. Using live-cell imaging, we found that MYO10 drives filopodial persistence necessary for micropatterning extracellular fibronectin into linear tracks at the edge of 3D collective invasion exclusively in leaders. Our data fit a model where epigenetic heterogeneity and JAG1 signaling jointly drive collective cancer invasion through MYO10 up-regulation in epigenetically permissive leader cells, which induces filopodia dynamics necessary for linearized fibronectin micropatterning.
Collapse
Affiliation(s)
| | - Janna K. Mouw
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Joshua S. K. Bell
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA, USA
| | - Christina M. Knippler
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Brian Pedro
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - Jamie L. Arnst
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Tala O. Khatib
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA, USA
| | - Rachel Commander
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jessica Konen
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - Bhakti Dwivedi
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sandra Seby
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jeanne Kowalski
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Paula M. Vertino
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Adam I. Marcus
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
33
|
|
34
|
Affiliation(s)
- Kate E Keller
- Department of Ophthalmology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
35
|
Tunneling Nanotubes and the Eye: Intercellular Communication and Implications for Ocular Health and Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7246785. [PMID: 32352005 PMCID: PMC7171654 DOI: 10.1155/2020/7246785] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
Cellular communication is an essential process for the development and maintenance of all tissues including the eye. Recently, a new method of cellular communication has been described, which relies on formation of tubules, called tunneling nanotubes (TNTs). These structures connect the cytoplasm of adjacent cells and allow the direct transport of cellular cargo between cells without the need for secretion into the extracellular milieu. TNTs may be an important mechanism for signaling between cells that reside long distances from each other or for cells in aqueous environments, where diffusion-based signaling is challenging. Given the wide range of cargoes transported, such as lysosomes, endosomes, mitochondria, viruses, and miRNAs, TNTs may play a role in normal homeostatic processes in the eye as well as function in ocular disease. This review will describe TNT cellular communication in ocular cell cultures and the mammalian eye in vivo, the role of TNTs in mitochondrial transport with an emphasis on mitochondrial eye diseases, and molecules involved in TNT biogenesis and their function in eyes, and finally, we will describe TNT formation in inflammation, cancer, and stem cells, focusing on pathological processes of particular interest to vision scientists.
Collapse
|
36
|
Wang B, Pan JX, Yu H, Xiong L, Zhao K, Xiong S, Guo JP, Lin S, Sun D, Zhao L, Guo H, Mei L, Xiong WC. Lack of Myosin X Enhances Osteoclastogenesis and Increases Cell Surface Unc5b in Osteoclast-Lineage Cells. J Bone Miner Res 2019; 34:939-954. [PMID: 30645777 PMCID: PMC7105956 DOI: 10.1002/jbmr.3667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/23/2018] [Accepted: 01/05/2019] [Indexed: 01/08/2023]
Abstract
Normal bone mass is maintained by balanced bone formation and resorption. Myosin X (Myo10), an unconventional "myosin tail homology 4-band 4.1, ezrin, radixin, moesin" (MyTH4-FERM) domain containing myosin, is implicated in regulating osteoclast (OC) adhesion, podosome positioning, and differentiation in vitro. However, evidence is lacking for Myo10 in vivo function. Here we show that mice with Myo10 loss of function, Myo10m/m , exhibit osteoporotic deficits, which are likely due to the increased OC genesis and bone resorption because bone formation is unchanged. Similar deficits are detected in OC-selective Myo10 conditional knockout (cko) mice, indicating a cell autonomous function of Myo10. Further mechanistic studies suggest that Unc-5 Netrin receptor B (Unc5b) protein levels, in particular its cell surface level, are higher in the mutant OCs, but lower in RAW264.7 cells or HEK293 cells expressing Myo10. Suppressing Unc5b expression in bone marrow macrophages (BMMs) from Myo10m/m mice by infection with lentivirus of Unc5b shRNA markedly impaired RANKL-induced OC genesis. Netrin-1, a ligand of Unc5b, increased RANKL-induced OC formation in BMMs from both wild-type and Myo10m/m mice. Taken together, these results suggest that Myo10 plays a negative role in OC formation, likely by inhibiting Unc5b cell-surface targeting, and suppressing Netrin-1 promoted OC genesis. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Bo Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.,Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jin-Xiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Huali Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.,Key laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Kai Zhao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shan Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jun-Peng Guo
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sen Lin
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Lu Zhao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.,Key laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Haohan Guo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
37
|
Baena V, Terasaki M. Three-dimensional organization of transzonal projections and other cytoplasmic extensions in the mouse ovarian follicle. Sci Rep 2019; 9:1262. [PMID: 30718581 PMCID: PMC6362238 DOI: 10.1038/s41598-018-37766-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
Each mammalian oocyte is nurtured by its own multi-cellular structure, the ovarian follicle. We used new methods for serial section electron microscopy to examine entire cumulus and mural granulosa cells and their projections in mouse antral ovarian follicles. Transzonal projections (TZPs) are thin cytoplasmic projections that connect cumulus cells to the oocyte and are crucial for normal oocyte development. We studied these projections in detail and found that most TZPs do not reach the oocyte, and that they often branch and make gap junctions with each other. Furthermore, the TZPs that connect to the oocyte are usually contacted on their shaft by oocyte microvilli. Mural granulosa cells were found to possess randomly oriented cytoplasmic projections that are strikingly similar to the free-ended TZPs. We propose that granulosa cells use cytoplasmic projections to search for the oocyte, and cumulus cell differentiation results from a contact-mediated paracrine interaction with the oocyte.
Collapse
Affiliation(s)
- Valentina Baena
- Department of Cell Biology, UConn Health, Farmington, CT, USA
| | - Mark Terasaki
- Department of Cell Biology, UConn Health, Farmington, CT, USA.
| |
Collapse
|
38
|
Bachg AC, Horsthemke M, Skryabin BV, Klasen T, Nagelmann N, Faber C, Woodham E, Machesky LM, Bachg S, Stange R, Jeong HW, Adams RH, Bähler M, Hanley PJ. Phenotypic analysis of Myo10 knockout (Myo10 tm2/tm2) mice lacking full-length (motorized) but not brain-specific headless myosin X. Sci Rep 2019; 9:597. [PMID: 30679680 PMCID: PMC6345916 DOI: 10.1038/s41598-018-37160-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023] Open
Abstract
We investigated the physiological functions of Myo10 (myosin X) using Myo10 reporter knockout (Myo10tm2) mice. Full-length (motorized) Myo10 protein was deleted, but the brain-specific headless (Hdl) isoform (Hdl-Myo10) was still expressed in homozygous mutants. In vitro, we confirmed that Hdl-Myo10 does not induce filopodia, but it strongly localized to the plasma membrane independent of the MyTH4-FERM domain. Filopodia-inducing Myo10 is implicated in axon guidance and mice lacking the Myo10 cargo protein DCC (deleted in colorectal cancer) have severe commissural defects, whereas MRI (magnetic resonance imaging) of isolated brains revealed intact commissures in Myo10tm2/tm2 mice. However, reminiscent of Waardenburg syndrome, a neural crest disorder, Myo10tm2/tm2 mice exhibited pigmentation defects (white belly spots) and simple syndactyly with high penetrance (>95%), and 24% of mutant embryos developed exencephalus, a neural tube closure defect. Furthermore, Myo10tm2/tm2 mice consistently displayed bilateral persistence of the hyaloid vasculature, revealed by MRI and retinal whole-mount preparations. In principle, impaired tissue clearance could contribute to persistence of hyaloid vasculature and syndactyly. However, Myo10-deficient macrophages exhibited no defects in the phagocytosis of apoptotic or IgG-opsonized cells. RNA sequence analysis showed that Myo10 was the most strongly expressed unconventional myosin in retinal vascular endothelial cells and expression levels increased 4-fold between P6 and P15, when vertical sprouting angiogenesis gives rise to deeper layers. Nevertheless, imaging of isolated adult mutant retinas did not reveal vascularization defects. In summary, Myo10 is important for both prenatal (neural tube closure and digit formation) and postnatal development (hyaloid regression, but not retinal vascularization).
Collapse
Affiliation(s)
- Anne C Bachg
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Markus Horsthemke
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Boris V Skryabin
- Department of Medicine, Transgenic Animal and Genetic Engineering Models (TRAM), Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Tim Klasen
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Nina Nagelmann
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Emma Woodham
- Cancer Research UK Beatson Institute, Glasgow University College of Medical, Veterinary and Life Sciences Garscube Estate, Glasgow, G61 1BD, United Kingdom
| | - Laura M Machesky
- Cancer Research UK Beatson Institute, Glasgow University College of Medical, Veterinary and Life Sciences Garscube Estate, Glasgow, G61 1BD, United Kingdom
| | - Sandra Bachg
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149, Münster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149, Münster, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, 48149, Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, 48149, Münster, Germany
| | - Martin Bähler
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Peter J Hanley
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany.
| |
Collapse
|
39
|
Young LE, Latario CJ, Higgs HN. Roles for Ena/VASP proteins in FMNL3-mediated filopodial assembly. J Cell Sci 2018; 131:131/21/jcs220814. [PMID: 30373894 DOI: 10.1242/jcs.220814] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/25/2018] [Indexed: 02/01/2023] Open
Abstract
Filopodia are actin-dependent finger-like structures that protrude from the plasma membrane. Actin filament barbed-end-binding proteins localized to filopodial tips are key to filopodial assembly. Two classes of barbed-end-binding proteins are formins and Ena/VASP proteins, and both classes have been localized to filopodial tips in specific cellular contexts. Here, we examine the filopodial roles of the FMNL formins and Ena/VASP proteins in U2OS cells. FMNL3 suppression reduces filopodial assembly by 90%, and FMNL3 is enriched at >95% of filopodial tips. Suppression of VASP or Mena (also known as ENAH) reduces filopodial assembly by >75%. However, VASP and Mena do not display consistent filopodial tip localization, but are enriched in focal adhesions (FAs). Interestingly, >85% of FMNL3-containing filopodia are associated with FAs. Two situations increase Ena/VASP filopodial localization: (1) expression of myosin-X, and (2) actively spreading cells. In spreading cells, filopodia often mark sites of nascent adhesions. Interestingly, VASP suppression in spreading cells causes a significant increase in adhesion assembly at filopodial tips. This work demonstrates that, in U2OS cells, Ena/VASP proteins play roles in filopodia beyond those at filopodial tips.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lorna E Young
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Casey J Latario
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| |
Collapse
|
40
|
Fischer RS, Lam PY, Huttenlocher A, Waterman CM. Filopodia and focal adhesions: An integrated system driving branching morphogenesis in neuronal pathfinding and angiogenesis. Dev Biol 2018; 451:86-95. [PMID: 30193787 DOI: 10.1016/j.ydbio.2018.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/08/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022]
Abstract
Single cell branching during development in vertebrates is typified by neuronal branching to form neurites and vascular branches formed by sprouting angiogenesis. Neurons and endothelial tip cells possess subcellular protrusions that share many common features from the morphological to the molecular level. Both systems utilize filopodia as their cellular protrusion organelles and depend on specific integrin-mediated adhesions to the local extracellular matrix for guidance in their pathfinding. We discuss the similar molecular machineries involved in these two types of cell branch formation and use their analogy to propose a new mechanism for angiogenic filopodia function, namely as adhesion assembly sites. In support of this model we provide primary data of angiogenesis in zebrafish in vivo showing that the actin assembly factor VASP participates in both filopodia formation and adhesion assembly at the base of the filopodia, enabling forward progress of the tip cell. The use of filopodia and their associated adhesions provide a common mechanism for neuronal and endothelial pathfinding during development in response to extracellular matrix cues.
Collapse
Affiliation(s)
- Robert S Fischer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, United States
| | - Pui-Ying Lam
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, United States
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, United States
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, United States.
| |
Collapse
|
41
|
Insights into the Etiology of Mammalian Neural Tube Closure Defects from Developmental, Genetic and Evolutionary Studies. J Dev Biol 2018; 6:jdb6030022. [PMID: 30134561 PMCID: PMC6162505 DOI: 10.3390/jdb6030022] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023] Open
Abstract
The human neural tube defects (NTD), anencephaly, spina bifida and craniorachischisis, originate from a failure of the embryonic neural tube to close. Human NTD are relatively common and both complex and heterogeneous in genetic origin, but the genetic variants and developmental mechanisms are largely unknown. Here we review the numerous studies, mainly in mice, of normal neural tube closure, the mechanisms of failure caused by specific gene mutations, and the evolution of the vertebrate cranial neural tube and its genetic processes, seeking insights into the etiology of human NTD. We find evidence of many regions along the anterior–posterior axis each differing in some aspect of neural tube closure—morphology, cell behavior, specific genes required—and conclude that the etiology of NTD is likely to be partly specific to the anterior–posterior location of the defect and also genetically heterogeneous. We revisit the hypotheses explaining the excess of females among cranial NTD cases in mice and humans and new developments in understanding the role of the folate pathway in NTD. Finally, we demonstrate that evidence from mouse mutants strongly supports the search for digenic or oligogenic etiology in human NTD of all types.
Collapse
|
42
|
Boyer NP, Gupton SL. Revisiting Netrin-1: One Who Guides (Axons). Front Cell Neurosci 2018; 12:221. [PMID: 30108487 PMCID: PMC6080411 DOI: 10.3389/fncel.2018.00221] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/09/2018] [Indexed: 12/28/2022] Open
Abstract
Proper patterning of the nervous system requires that developing axons find appropriate postsynaptic partners; this entails microns to meters of extension through an extracellular milieu exhibiting a wide range of mechanical and chemical properties. Thus, the elaborate networks of fiber tracts and non-fasciculated axons evident in mature organisms are formed via complex pathfinding. The macroscopic structures of axon projections are highly stereotyped across members of the same species, indicating precise mechanisms guide their formation. The developing axon exhibits directionally biased growth toward or away from external guidance cues. One of the most studied guidance cues is netrin-1, however, its presentation in vivo remains debated. Guidance cues can be secreted to form soluble or chemotactic gradients or presented bound to cells or the extracellular matrix to form haptotactic gradients. The growth cone, a highly specialized dynamic structure at the end of the extending axon, detects these guidance cues via transmembrane receptors, such as the netrin-1 receptors deleted in colorectal cancer (DCC) and UNC5. These receptors orchestrate remodeling of the cytoskeleton and cell membrane through both chemical and mechanotransductive pathways, which result in traction forces generated by the cytoskeleton against the extracellular environment and translocation of the growth cone. Through intracellular signaling responses, netrin-1 can trigger either attraction or repulsion of the axon. Here we review the mechanisms by which the classical guidance cue netrin-1 regulates intracellular effectors to respond to the extracellular environment in the context of axon guidance during development of the central nervous system and discuss recent findings that demonstrate the critical importance of mechanical forces in this process.
Collapse
Affiliation(s)
- Nicholas P. Boyer
- Neurobiology Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
43
|
Tokuo H, Bhawan J, Coluccio LM. Myosin X is required for efficient melanoblast migration and melanoma initiation and metastasis. Sci Rep 2018; 8:10449. [PMID: 29993000 PMCID: PMC6041326 DOI: 10.1038/s41598-018-28717-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/15/2018] [Indexed: 12/20/2022] Open
Abstract
Myosin X (Myo10), an actin-associated molecular motor, has a clear role in filopodia induction and cell migration in vitro, but its role in vivo in mammals is not well understood. Here, we investigate the role of Myo10 in melanocyte lineage and melanoma induction. We found that Myo10 knockout (Myo10KO) mice exhibit a white spot on their belly caused by reduced melanoblast migration. Myo10KO mice crossed with available mice that conditionally express in melanocytes the BRAFV600E mutation combined with Pten silencing exhibited reduced melanoma development and metastasis, which extended medial survival time. Knockdown of Myo10 (Myo10kd) in B16F1 mouse melanoma cell lines decreased lung colonization after tail-vein injection. Myo10kd also inhibited long protrusion (LP) formation by reducing the transportation of its cargo molecule vasodilator-stimulated phosphoprotein (VASP) to the leading edge of migrating cells. These findings provide the first genetic evidence for the involvement of Myo10 not only in melanoblast migration, but also in melanoma development and metastasis.
Collapse
Affiliation(s)
- Hiroshi Tokuo
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Lynne M Coluccio
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
| |
Collapse
|
44
|
Patel N, Khan AO, Alsahli S, Abdel-Salam G, Nowilaty SR, Mansour AM, Nabil A, Al-Owain M, Sogati S, Salih MA, Kamal AM, Alsharif H, Alsaif HS, Alzahrani SS, Abdulwahab F, Ibrahim N, Hashem M, Faquih T, Shah ZA, Abouelhoda M, Monies D, Dasouki M, Shaheen R, Wakil SM, Aldahmesh MA, Alkuraya FS. Genetic investigation of 93 families with microphthalmia or posterior microphthalmos. Clin Genet 2018; 93:1210-1222. [PMID: 29450879 DOI: 10.1111/cge.13239] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 01/24/2023]
Abstract
Microphthalmia is a developmental eye defect that is highly variable in severity and in its potential for systemic association. Despite the discovery of many disease genes in microphthalmia, at least 50% of patients remain undiagnosed genetically. Here, we describe a cohort of 147 patients (93 families) from our highly consanguineous population with various forms of microphthalmia (including the distinct entity of posterior microphthalmos) that were investigated using a next-generation sequencing multi-gene panel (i-panel) as well as whole exome sequencing and molecular karyotyping. A potentially causal mutation was identified in the majority of the cohort with microphthalmia (61%) and posterior microphthalmos (82%). The identified mutations (55 point mutations, 15 of which are novel) spanned 24 known disease genes, some of which have not or only very rarely been linked to microphthalmia (PAX6, SLC18A2, DSC3 and CNKSR1). Our study has also identified interesting candidate variants in 2 genes that have not been linked to human diseases (MYO10 and ZNF219), which we present here as novel candidates for microphthalmia. In addition to revealing novel phenotypic aspects of microphthalmia, this study expands its allelic and locus heterogeneity and highlights the need for expanded testing of patients with this condition.
Collapse
Affiliation(s)
- N Patel
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - A O Khan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.,Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - S Alsahli
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | | | - S R Nowilaty
- Vitreo-retinal Division, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - A M Mansour
- Department of Ophthalmology, American University of Beirut, Beirut, Lebanon
| | - A Nabil
- Human Genetics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - M Al-Owain
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - S Sogati
- Department of Medical Genetics, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - M A Salih
- Division of Pediatrics Neurology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - A M Kamal
- Department of Ophthalmology, Cairo University, Cairo, Egypt
| | - H Alsharif
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - H S Alsaif
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - S S Alzahrani
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - F Abdulwahab
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - N Ibrahim
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - M Hashem
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - T Faquih
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.,Saudi Human Genome Project, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Z A Shah
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.,Saudi Human Genome Project, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - M Abouelhoda
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.,Saudi Human Genome Project, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - D Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.,Saudi Human Genome Project, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - M Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - R Shaheen
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - S M Wakil
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - M A Aldahmesh
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - F S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.,Saudi Human Genome Project, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|