Non-Muscle Myosin II A: Friend or Foe in Cancer?
Abstract
:1. Introduction
2. Structure of NM IIA
3. Regulation of NM IIA
3.1. Role of RLC Phosphorylation in Regulating NM IIA Activity
3.2. Role of NMHC Phosphorylation in Regulating NM IIA Activity
3.3. Role of Protein Interactions in Regulating NM IIA Activity
4. Physiological Functions of NM IIA
5. MYH9 as an Oncogene
5.1. Head and Neck Squamous Cell Carcinoma
5.2. Gliomas
5.3. Nasopharyngeal Carcinoma (NPC)
5.4. Non-Small Cell Lung Cancer (NSCLC)
5.5. Hepatocellular Carcinoma (HCC)
5.6. Pancreatic Cancer (PC)
5.7. Esophageal Cancer
5.8. Gastric Cancer (GC)
5.9. Colorectal Cancer (CRC)
5.10. Breast Cancer
5.11. Renal Cell Carcinomas (RCCs)
5.12. Prostate Cancer
5.13. Osteosarcoma
5.14. Papillary Thyroid Carcinoma (PTC)
5.15. Acute Myeloid Leukemia (AML)
5.16. Diffuse Large B Cell Lymphoma (DLBCL)
6. The Role of NM IIA as a Tumor Suppressor in Mice
6.1. Squamous Cell Carcinoma of the Skin
6.2. Melanoma
7. Dual Role of NM IIA in Humans
8. Role of NM IIA in Other Pathological Conditions
9. Targeting MYH9
Targeting the ROCK-Myosin II Signaling Pathway
10. Conclusion and Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
MYH9 | Myosin heavy chain 9 |
MYH10 | Myosin heavy chain 10 |
MYH14 | Myosin heavy chain 14 |
NM IIA | Non-muscle myosin IIA |
NMHC IIA | Non-muscle myosin IIA heavy chain |
NMHC IIB | Non-muscle myosin IIB heavy chain |
NMHC IIC | Non-muscle myosin IIC heavy chain |
MHA | May–Hegglin anomaly |
EPS | Epstein syndrome |
FTS | Fechtner syndrome |
SPS | Sebastian platelet syndrome |
ELC | Essential light chain |
RLC | Regulatory light chain |
MHC | Myosin heavy chain |
MLC | Myosin light chain |
MHC IIA | Myosin heavy chain IIA |
ROCK | Rho-associated protein kinase |
ORF | Open reading frame |
MYL6 | Myosin light chain 6 |
MYL6B | Myosin light chain 6B |
MYL 12A | Myosin light chain 12A |
MYL 12B | Myosin light chain 12B |
MLCK | Myosin light chain kinase |
MyBP-C | Myosin-binding protein C |
MyBP-H | Myosin-binding protein H |
PKA | Protein kinase A |
PKC | Protein kinase C |
PKCβ | Protein kinase Cβ |
TRPM7 | Transient receptor potential melastatin 7 |
CK II | Casein kinase II |
MRCK | Myotonic dystrophy kinase-related CDC42-binding kinase |
MYPT1 | Myosin phosphatase target subunit 1 |
PDGF | Platelet-derived growth factor |
PAK1 | p21-activated kinase 1 |
DAPK | Death-associated protein kinase |
NHT | Non-helical tail |
EMT | Epithelial-to-mesenchymal transition |
TGF-β | Transforming growth factor-β |
EGF | Epidermal growth factor |
Lgl | Lethal (2) giant larvae |
aPKCζ | Atypical protein kinase C |
LIMCH1 | LIM and calponin-homology domains 1 |
MYBPHL | Myosin-binding protein H-like |
Tpm4.2 | Tropomyosin 4.2 |
NMY-2 | Non-muscle myosin II heavy chain in C. elegans |
ECM | Extracellular matrix |
MyoGEF | Myosin-interacting guanine nucleotide exchange factor |
GSIS | Glucose-stimulated insulin secretion |
APPL1 | Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 |
PP2A | Protein phosphatase 2A |
FAK | Focal adhesion kinase |
MHB | Midbrain–hindbrain boundary |
HNSCC | Head and neck squamous cell carcinoma |
NEDD9 | Neural precursor cell expressed, developmentally downregulated 9 |
CAS | Crk-associated substrate |
MMPs | Matrix metalloproteinases |
HMGA1 | High mobility group AT-jook 1 |
PDOX | Patient-derived orthotopic xenograft |
GBM | Glioblastoma |
THBS1 | Thrombospondin 1 |
VEGFA | Vascular endothelial growth factor A |
HIF-1α | Hypoxia-inducible factor-1α |
NPC | Nasopharyngeal carcinoma |
EBV | Epstein–Barr virus |
BART | BamHI A rightward transcript |
FKHR | Forkhead in rhabdomyosarcoma |
FoxO | Forkhead box O-class |
TRAF6 | TNF receptor-associated factor 6 |
GSK3β | Glycogen synthase kinase-3β |
FNDC3B | Fibronectin type III domain containing 3B |
NSCLC | Non-small cell lung cancer |
CSC | Cancer stem cell |
LCCs | Lung cancer cells |
EGFR | Epidermal growth factor receptor |
TKIs | Tyrosine kinase inhibitors |
TME | Tumor microenvironment |
CAFs | Cancer-associated fibroblasts |
MICAL2 | Microtubule-associated monooxygenase, calponin, and LIM domain containing 2 |
HCC | Hepatocellular carcinoma |
APC | Adenomatous polyposis coli |
AXIN1 | Axis inhibition protein 1 |
HBX | Hepatitis B virus X protein |
TRPCs | Canonical transient receptor potential channels |
NAP1L5 | Nucleosome assembly protein 1-like 5 |
PC | Pancreatic cancer |
GEO | Gene expression omnibus |
PACC | Pancreatic acinar cell carcinoma |
SCC | Squamous cell carcinoma |
ADCA | Adenocarcinoma |
NGS | Next-generation sequencing |
ESCC | Esophageal squamous cell carcinoma |
PTP1B | Protein tyrosine phosphatase 1B |
GC | Gastric cancer |
FBXW7 | F-Box and WD repeat domain containing 7 |
USP2 | Ubiquitin-specific peptidase 2 |
HULC | Hepatocellular carcinoma upregulated long non-coding RNA |
SLAMF6 | SLAM family member 6 |
SRF | Serum response factor |
S100A4 | S100 calcium binding protein A4 |
S100A1 | S100 calcium binding protein A1 |
S100P | S100 calcium binding protein P |
CRC | Colorectal cancer |
AMPK | AMP-activated protein kinase |
mTOR | Mechanistic target of rapamycin kinase |
MAPK | Mitogen-activated protein kinase |
AKT | AKT serine/threonine kinase |
ATG9B | Autophagy-related protein 9B |
STUB1 | STIP1 homology and U-Box containing protein 1 |
ACTN4 | Actinin-4 |
LIMK1 | LIM kinase 1 |
TIMELESS | Timeless circadian regulator |
CRY | Cryptochrome proteins |
PER | Period proteins |
PLAGL2 | Polymorphic adenoma-like protein 2 |
HNRNPA2B1 | Heterogeneous nuclear ribonucleoprotein A2/B1 |
HIF1α | Hypoxia inducible factor 1 subunit-α |
EIF6 | Eukaryotic translation initiation factor 6 |
TFPI-2 | Tissue factor pathway inhibitor 2 |
ISG15 | Interferon-stimulated gene 15 |
IFN-γ | Interferon γ |
HSP | Heat shock protein |
ERN1 | Endoplasmic reticulum to nucleus signaling 1 |
HBXIP | Hepatitis B X-interacting protein |
ACD | Assembly-competent domain |
PKCβII | Protein kinase C βII |
HER2 | Human epidermal growth factor receptor 2 |
HER3 | Human epidermal growth factor receptor 3 |
RCC | Renal cell carcinoma |
CXCR4 | C-X-C motif chemokine receptor 4 |
ccRCC | Clear cell renal cell carcinoma |
TUBB4A | Tubulin beta class IVA |
OS | Osteosarcoma |
lncRNA | Long non-coding RNA |
PTC | Papillary thyroid carcinoma |
CRLF1 | Cytokine receptor-like factor 1 |
ETV4 | ETS variant transcription factor 4 |
MMP1 | Matrix metalloproteinase 1 |
DTC | Differentiated thyroid carcinoma |
NEK6 | NIMA-related kinase 6 |
FOXE1 | Forkhead Box E1 |
PTCSC2 | Papillary thyroid carcinoma susceptibility candidate 2 |
AML | Acute myeloid leukemia |
CML | Chronic myeloid leukemia |
DLBCL | Diffuse large B cell lymphoma |
PTGDS | Glycoprotein prostaglandin D2 synthase |
ROS | Reactive oxygen species |
MYH9-RD | MYH9-related diseases |
SBS | Sebastian syndrome |
FTNS | Fechtner syndrome |
ESRD | End-stage renal disease |
WES | Whole-exome sequencing |
ALS | Amyotrophic lateral sclerosis |
MS | Multiple sclerosis |
shRNA | Short hairpin RNA |
KSHV | Kaposi’s sarcoma-associated herpesvirus |
HHV-8 | Human herpesvirus 8 |
PFTs | Pore-forming toxins |
LLO | Listeriolysin O |
PFO | Perfringolysin O |
VEGF | Vascular endothelial growth factor |
VEGFR2 | Vascular endothelial growth factor receptor-2 |
IQGAP1 | IQ motif containing GTPase activating protein 1 |
TNBC | Triple-negative breast cancer |
DARTS | Drug affinity responsive target stability |
HSPA9 | Heat shock protein A9 |
HHT | Homoharringtonine |
APE2 | Apurinic/apyrimidinic endonuclease 2 |
MLC2 | Myosin light chain 2 |
EF-1α | Elongation factor 1 α |
SLC9A1/NHE1 | Solute carrier family 9 member A1 |
TAMs | Tumor-associated macrophages |
CCL2 | C-C motif chemokine ligand 2 |
PD-1/PD-L1 | Programmed death-1/programmed death ligand-1 |
GVHD | Graft versus host disease |
MSN | Moesin |
NDRG1 | N-Myc downstream regulated 1 |
CA-4 | Combretastatin |
References
- Fletcher, D.A.; Mullins, R.D. Cell mechanics and the cytoskeleton. Nature 2010, 463, 485–492. [Google Scholar] [CrossRef]
- Ross, J.L.; Ali, M.Y.; Warshaw, D.M. Cargo transport: Molecular motors navigate a complex cytoskeleton. Curr. Opin. Cell Biol. 2008, 20, 41–47. [Google Scholar] [CrossRef] [PubMed]
- Pollard, T.D.; Goldman, R.D. Overview of the Cytoskeleton from an Evolutionary Perspective. Cold Spring Harb. Perspect. Biol. 2018, 10, a030288. [Google Scholar] [CrossRef] [PubMed]
- Richards, T.A.; Cavalier-Smith, T. Myosin domain evolution and the primary divergence of eukaryotes. Nature 2005, 436, 1113–1118. [Google Scholar] [CrossRef]
- Odronitz, F.; Kollmar, M. Drawing the tree of eukaryotic life based on the analysis of 2269 manually annotated myosins from 328 species. Genome Biol. 2007, 8, R196. [Google Scholar] [CrossRef]
- Sebé-Pedrós, A.; Grau-Bové, X.; Richards, T.A.; Ruiz-Trillo, I. Evolution and Classification of Myosins, a Paneukaryotic Whole-Genome Approach. Genome Biol. Evol. 2014, 6, 290–305. [Google Scholar] [CrossRef]
- Swailes, N.T.; Colegrave, M.; Knight, P.J.; Peckham, M. Non-muscle myosins 2A and 2B drive changes in cell morphology that occur as myoblasts align and fuse. J. Cell Sci. 2006, 119, 3561–3570. [Google Scholar] [CrossRef] [PubMed]
- Yuen, S.L.; Ogut, O.; Brozovich, F.V. Nonmuscle myosin is regulated during smooth muscle contraction. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H191–H199. [Google Scholar] [CrossRef]
- Morano, I.; Chai, G.-X.; Baltas, L.G.; Lamounier-Zepter, V.; Lutsch, G.; Kott, M.; Haase, H.; Bader, M. Smooth-muscle contraction without smooth-muscle myosin. Nat. Cell Biol. 2000, 2, 371–375. [Google Scholar] [CrossRef]
- Heissler, S.M.; Sellers, J.R. Kinetic Adaptations of Myosins for Their Diverse Cellular Functions. Traffic 2016, 17, 839–859. [Google Scholar] [CrossRef]
- Masters, T.A.; Kendrick-Jones, J.; Buss, F. Myosins: Domain Organisation, Motor Properties, Physiological Roles and Cellular Functions; Springer International Publishing: New York, NY, USA, 2016; pp. 77–122. [Google Scholar]
- Marigo, V.; Nigro, A.; Pecci, A.; Montanaro, D.; Di Stazio, M.; Balduini, C.L.; Savoia, A. Correlation between the clinical phenotype of MYH9-related disease and tissue distribution of class II nonmuscle myosin heavy chains. Genomics 2004, 83, 1125–1133. [Google Scholar] [CrossRef]
- Brown, S.S. Myosins in yeast. Curr. Opin. Cell Biol. 1997, 9, 44–48. [Google Scholar] [CrossRef] [PubMed]
- Mansfield, S.G.; al-Shirawi, D.Y.; Ketchum, A.S.; Newbern, E.C.; Kiehart, D.P. Molecular organization and alternative splicing in zipper, the gene that encodes the Drosophila non-muscle myosin II heavy chain. J. Mol. Biol. 1996, 255, 98–109. [Google Scholar] [CrossRef] [PubMed]
- Soldati, T.; Geissler, H.; Schwarz, E.C. How many is enough? Exploring the myosin repertoire in the model eukaryoteDictyostelium discoideum. Cell Biochem. Biophys. 1999, 30, 389–411. [Google Scholar] [CrossRef] [PubMed]
- Wang, A.; Ma, X.; Conti, M.A.; Adelstein, R.S. Distinct and redundant roles of the non-muscle myosin II isoforms and functional domains. Biochem. Soc. Trans. 2011, 39, 1131–1135. [Google Scholar] [CrossRef]
- Burridge, K.; Bray, D. Purification and structural analysis of myosins from brain and other non-muscle tissues. J. Mol. Biol. 1975, 99, 33398–33410. [Google Scholar] [CrossRef]
- Billington, N.; Wang, A.; Mao, J.; Adelstein, R.S.; Sellers, J.R. Characterization of Three Full-length Human Nonmuscle Myosin II Paralogs. J. Biol. Chem. 2013, 288, 33398–33410. [Google Scholar] [CrossRef]
- Golomb, E.; Ma, X.; Jana, S.S.; Preston, Y.A.; Kawamoto, S.; Shoham, N.G.; Goldin, E.; Conti, M.A.; Sellers, J.R.; Adelstein, R.S. Identification and Characterization of Nonmuscle Myosin II-C, a New Member of the Myosin II Family. J. Biol. Chem. 2004, 279, 2800–2808. [Google Scholar] [CrossRef] [PubMed]
- Nagy, A.; Takagi, Y.; Billington, N.; Sun, S.A.; Hong, D.K.T.; Homsher, E.; Wang, A.; Sellers, J.R. Kinetic Characterization of Nonmuscle Myosin IIB at the Single Molecule Level. J. Biol. Chem. 2013, 288, 709–722. [Google Scholar] [CrossRef]
- Li, Y.; Lalwani, A.K.; Mhatre, A.N. Alternative Splice Variants of MYH9. DNA Cell Biol. 2008, 27, 117–125. [Google Scholar] [CrossRef]
- Takahashi, M.; Kawamoto, S.; Adelstein, R.S. Evidence for inserted sequences in the head region of nonmuscle myosin specific to the nervous system. Cloning of the cDNA encoding the myosin heavy chain-B isoform of vertebrate nonmuscle myosin. J. Biol. Chem. 1992, 267, 17864–17871. [Google Scholar] [CrossRef] [PubMed]
- Wang, A.; Ma, X.; Conti, M.A.; Liu, C.; Kawamoto, S.; Adelstein, R.S. Nonmuscle myosin II isoform and domain specificity during early mouse development. Proc. Natl. Acad. Sci. USA 2010, 107, 14645–14650. [Google Scholar] [CrossRef]
- Kovacs, M.; Wang, F.; Hu, A.; Zhang, Y.; Sellers, J.R. Functional divergence of human cytoplasmic myosin II: Kinetic characterization of the non-muscle IIA isoform. J. Biol. Chem. 2003, 278, 38132–38140. [Google Scholar] [CrossRef] [PubMed]
- Kolega, J. Cytoplasmic dynamics of myosin IIA and IIB: Spatial ‘sorting’ of isoforms in locomoting cells. J. Cell Sci. 1998, 111, 2085–2095. [Google Scholar] [CrossRef] [PubMed]
- Maupin, P.; Phillips, C.L.; Adelstein, R.S.; Pollard, T.D. Differential localization of myosin-II isozymes in human cultured cells and blood cells. J. Cell Sci. 1994, 107, 3077–3090. [Google Scholar] [CrossRef] [PubMed]
- Bao, J.; Jana, S.S.; Adelstein, R.S. Vertebrate Nonmuscle Myosin II Isoforms Rescue Small Interfering RNA-induced Defects in COS-7 Cell Cytokinesis. J. Biol. Chem. 2005, 280, 19594–19599. [Google Scholar] [CrossRef]
- Pecci, A.; Ma, X.; Savoia, A.; Adelstein, R.S. MYH9: Structure, functions and role of non-muscle myosin IIA in human disease. Gene 2018, 664, 152–167. [Google Scholar] [CrossRef]
- Gazda, L.; Pokrzywa, W.; Hellerschmied, D.; Löwe, T.; Forné, I.; Mueller-Planitz, F.; Hoppe, T.; Clausen, T. The Myosin Chaperone UNC-45 Is Organized in Tandem Modules to Support Myofilament Formation in C. elegans. Cell 2013, 152, 183–195. [Google Scholar] [CrossRef]
- Rayment, I.; Rypniewski, W.R.; Schmidt-Base, K.; Smith, R.; Tomchick, D.R.; Benning, M.M.; Winkelmann, D.A.; Wesenberg, G.; Holden, H.M. Three-dimensional structure of myosin subfragment-1: A molecular motor. Science 1993, 261, 50–58. [Google Scholar] [CrossRef]
- Rayment, I.; Holden, H.M.; Whittaker, M.; Yohn, C.B.; Lorenz, M.; Holmes, K.C.; Milligan, R.A. Structure of the actin-myosin complex and its implications for muscle contraction. Science 1993, 261, 58–65. [Google Scholar] [CrossRef]
- Winkelmann, D.A.; Almeda, S.; Vibert, P.; Cohen, C. A new myosin fragment: Visualization of the regulatory domain. Nature 1984, 307, 758–760. [Google Scholar] [CrossRef]
- Cote, G.P.; Robinson, E.A.; Appella, E.; Korn, E.D. Amino acid sequence of a segment of the Acanthamoeba myosin II heavy chain containing all three regulatory phosphorylation sites. J. Biol. Chem. 1984, 259, 12781–12787. [Google Scholar] [CrossRef]
- Sellers, J.R. Myosins: A diverse superfamily. Biochim. Biophys. Acta 2000, 1496, 3–22. [Google Scholar] [CrossRef] [PubMed]
- Cheney, R.E.; Mooseker, M.S. Unconventional myosins. Curr. Opin. Cell Biol. 1992, 4, 27–35. [Google Scholar] [CrossRef]
- Uyeda, T.Q.; Abramson, P.D.; Spudich, J.A. The neck region of the myosin motor domain acts as a lever arm to generate movement. Proc. Natl. Acad. Sci. USA 1996, 93, 4459–4464. [Google Scholar] [CrossRef] [PubMed]
- Heissler, S.M.; Sellers, J.R. Myosin light chains: Teaching old dogs new tricks. BioArchitecture 2014, 4, 169–188. [Google Scholar] [CrossRef]
- Maliga, Z.; Junqueira, M.; Toyoda, Y.; Ettinger, A.; Mora-Bermúdez, F.; Klemm, R.W.; Vasilj, A.; Guhr, E.; Ibarlucea-Benitez, I.; Poser, I.; et al. A genomic toolkit to investigate kinesin and myosin motor function in cells. Nat. Cell Biol. 2013, 15, 325–334. [Google Scholar] [CrossRef]
- Dasbiswas, K.; Hu, S.; Schnorrer, F.; Safran, S.A.; Bershadsky, A.D. Ordering of myosin II filaments driven by mechanical forces: Experiments and theory. Philos. Trans. R Soc. B Biol. Sci. 2018, 373, 20170114. [Google Scholar] [CrossRef] [PubMed]
- Craig, R.; Woodhead, J.L. Structure and function of myosin filaments. Curr. Opin. Struct. Biol. 2006, 16, 204–212. [Google Scholar] [CrossRef]
- Onishi, H.; Wakabayashi, T. Electron microscopic studies of myosin molecules from chicken gizzard muscle I: The formation of the intramolecular loop in the myosin tail. J. Biochem. 1982, 92, 871–879. [Google Scholar] [CrossRef]
- Trybus, K.M.; Huiatt, T.W.; Lowey, S. A bent monomeric conformation of myosin from smooth muscle. Proc. Natl. Acad. Sci. USA 1982, 79, 6151–6155. [Google Scholar] [CrossRef] [PubMed]
- Cross, R.A.; Cross, K.E.; Sobieszek, A. ATP-linked monomer-polymer equilibrium of smooth muscle myosin: The free folded monomer traps ADP.Pi. EMBO J. 1986, 5, 2637–2641. [Google Scholar] [CrossRef] [PubMed]
- Cross, R.A.; Jackson, A.P.; Citi, S.; Kendrick-Jones, J.; Bagshaw, C.R. Active site trapping of nucleotide by smooth and non-muscle myosins. J. Mol. Biol. 1988, 203, 173–181. [Google Scholar] [CrossRef]
- Scholey, J.M.; Taylor, K.A.; Kendrick-Jones, J. Regulation of non-muscle myosin assembly by calmodulin-dependent light chain kinase. Nature 1980, 287, 233–235. [Google Scholar] [CrossRef] [PubMed]
- Rottbauer, W.; Wessels, G.; Dahme, T.; Just, S.; Trano, N.; Hassel, D.; Burns, C.G.; Katus, H.A.; Fishman, M.C. Cardiac Myosin Light Chain-2. Circ. Res. 2006, 99, 323–331. [Google Scholar] [CrossRef]
- Coluccio, L.M. Myosins; Springer: New York, NY, USA, 2012; pp. 1177–1182. [Google Scholar]
- Heissler, S.M.; Manstein, D.J. Nonmuscle myosin-2: Mix and match. Cell. Mol. Life Sci. 2013, 70, 1–21. [Google Scholar] [CrossRef]
- Vicente-Manzanares, M.; Newell-Litwa, K.; Bachir, A.I.; Whitmore, L.A.; Horwitz, A.R. Myosin IIA/IIB restrict adhesive and protrusive signaling to generate front–back polarity in migrating cells. J. Cell Biol. 2011, 193, 381–396. [Google Scholar] [CrossRef]
- Schiffhauer, E.S.; Luo, T.; Mohan, K.; Srivastava, V.; Qian, X.; Griffis, E.R.; Iglesias, P.A.; Robinson, D.N. Mechanoaccumulative Elements of the Mammalian Actin Cytoskeleton. Curr. Biol. 2016, 26, 1473–1479. [Google Scholar] [CrossRef]
- Schiffhauer, E.S.; Ren, Y.; Iglesias, V.A.; Kothari, P.; Iglesias, P.A.; Robinson, D.N. Myosin IIB assembly state determines its mechanosensitive dynamics. J. Cell Biol. 2019, 218, 895–908. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, C.; Adelstein, R.S.; Ma, X. Replacing nonmuscle myosin 2A with myosin 2C1 permits gastrulation but not placenta vascular development in mice. Mol. Biol. Cell 2018, 29, 2326–2335. [Google Scholar] [CrossRef]
- Lin, B.L.; Li, A.; Mun, J.Y.; Previs, M.J.; Previs, S.B.; Campbell, S.G.; Dos Remedios, C.G.; Tombe, P.D.P.; Craig, R.; Warshaw, D.M.; et al. Skeletal myosin binding protein-C isoforms regulate thin filament activity in a Ca2+-dependent manner. Sci. Rep. 2018, 8, 2604. [Google Scholar] [CrossRef] [PubMed]
- Jung, H.S.; Burgess, S.A.; Billington, N.; Colegrave, M.; Patel, H.; Chalovich, J.M.; Chantler, P.D.; Knight, P.J. Conservation of the regulated structure of folded myosin 2 in species separated by at least 600 million years of independent evolution. Proc. Natl. Acad. Sci. USA 2008, 105, 6022–6026. [Google Scholar] [CrossRef] [PubMed]
- Burgess, S.A.; Yu, S.; Walker, M.L.; Hawkins, R.J.; Chalovich, J.M.; Knight, P.J. Structures of Smooth Muscle Myosin and Heavy Meromyosin in the Folded, Shutdown State. J. Mol. Biol. 2007, 372, 1165–1178. [Google Scholar] [CrossRef]
- Jung, H.S.; Komatsu, S.; Ikebe, M.; Craig, R. Head–Head and Head–Tail Interaction: A General Mechanism for Switching Off Myosin II Activity in Cells. Mol. Biol. Cell 2008, 19, 3234–3242. [Google Scholar] [CrossRef]
- Milton, D.L.; Schneck, A.N.; Ziech, D.A.; Ba, M.; Facemyer, K.C.; Halayko, A.J.; Baker, J.E.; Gerthoffer, W.T.; Cremo, C.R. Direct evidence for functional smooth muscle myosin II in the 10S self-inhibited monomeric conformation in airway smooth muscle cells. Proc. Natl. Acad. Sci. USA 2011, 108, 1421–1426. [Google Scholar] [CrossRef]
- Casadei, J.M.; Gordon, R.D.; Lampson, L.A.; Schotland, D.L.; Barchi, R.L. Monoclonal antibodies against the voltage-sensitive Na+ channel from mammalian skeletal muscle. Proc. Natl. Acad. Sci. USA 1984, 81, 6227–6231. [Google Scholar] [CrossRef]
- Craig, R.; Smith, R.; Kendrick-Jones, J. Light-chain phosphorylation controls the conformation of vertebrate non-muscle and smooth muscle myosin molecules. Nature 1983, 302, 436–439. [Google Scholar] [CrossRef]
- Yang, S.; Lee, K.H.; Woodhead, J.L.; Sato, O.; Ikebe, M.; Craig, R. The central role of the tail in switching off 10S myosin II activity. J. Gen. Physiol. 2019, 151, 1081–1093. [Google Scholar] [CrossRef]
- Yang, S.; Tiwari, P.; Lee, K.H.; Sato, O.; Ikebe, M.; Padrón, R.; Craig, R. Cryo-EM structure of the inhibited (10S) form of myosin II. Nature 2020, 588, 521–525. [Google Scholar] [CrossRef] [PubMed]
- Adelstein, R.S.; Conti, M.A. Phosphorylation of platelet myosin increases actin-activated myosin ATPase activity. Nature 1975, 256, 597–598. [Google Scholar] [CrossRef]
- Trybus, K.M.; Lowey, S. Conformational states of smooth muscle myosin. Effects of light chain phosphorylation and ionic strength. J. Biol. Chem. 1984, 259, 8564–8571. [Google Scholar] [CrossRef]
- Kimura, K.; Ito, M.; Amano, M.; Chihara, K.; Fukata, Y.; Nakafuku, M.; Yamamori, B.; Feng, J.; Nakano, T.; Okawa, K.; et al. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science 1996, 273, 245–248. [Google Scholar] [CrossRef] [PubMed]
- Vicente-Manzanares, M.; Horwitz, A.R. Myosin light chain mono-and di-phosphorylation differentially regulate adhesion and polarity in migrating cells. Biochem. Biophys. Res. Commun. 2010, 402, 537–542. [Google Scholar] [CrossRef]
- Ikebe, M.; Hartshorne, D.J.; Elzinga, M. Identification, phosphorylation, and dephosphorylation of a second site for myosin light chain kinase on the 20,000-dalton light chain of smooth muscle myosin. J. Biol. Chem. 1986, 261, 36–39. [Google Scholar] [CrossRef] [PubMed]
- Aguilar-Cuenca, R.; Llorente-González, C.; Chapman, J.R.; Talayero, V.C.; Garrido-Casado, M.; Delgado-Arévalo, C.; Millán-Salanova, M.; Shabanowitz, J.; Hunt, D.F.; Sellers, J.R.; et al. Tyrosine Phosphorylation of the Myosin Regulatory Light Chain Controls Non-muscle Myosin II Assembly and Function in Migrating Cells. Curr. Biol. 2020, 30, 2446–2458.e2446. [Google Scholar] [CrossRef] [PubMed]
- Matsumura, F.; Yamakita, Y.; Yamashiro, S. Myosin light chain kinases and phosphatase in mitosis and cytokinesis. Arch. Biochem. Biophys. 2011, 510, 76–82. [Google Scholar] [CrossRef] [PubMed]
- Tan, C.H.; Gasic, I.; Huber-Reggi, S.P.; Dudka, D.; Barisic, M.; Maiato, H.; Meraldi, P. The equatorial position of the metaphase plate ensures symmetric cell divisions. elife 2015, 4, e05124. [Google Scholar] [CrossRef]
- Nishikawa, M.; Sellers, J.R.; Adelstein, R.S.; Hidaka, H. Protein kinase C modulates in vitro phosphorylation of the smooth muscle heavy meromyosin by myosin light chain kinase. J. Biol. Chem. 1984, 259, 8808–8814. [Google Scholar] [CrossRef]
- Ikebe, M.; Reardon, S. Phosphorylation of bovine platelet myosin by protein kinase C. Biochemistry 1990, 29, 2713–2720. [Google Scholar] [CrossRef]
- Beach, J.R.; Licate, L.S.; Crish, J.F.; Egelhoff, T.T. Analysis of the role of Ser1/Ser2/Thr9 phosphorylation on myosin II assembly and function in live cells. BMC Cell Biol. 2011, 12, 52. [Google Scholar] [CrossRef]
- Komatsu, S.; Ikebe, M. The Phosphorylation of Myosin II at the Ser1 and Ser2 Is Critical for Normal Platelet-derived Growth Factor–induced Reorganization of Myosin Filaments. Mol. Biol. Cell 2007, 18, 5081–5090. [Google Scholar] [CrossRef]
- Asokan, S.B.; Johnson, H.E.; Rahman, A.; King, S.J.; Rotty, J.D.; Lebedeva, I.P.; Haugh, J.M.; Bear, J.E. Mesenchymal Chemotaxis Requires Selective Inactivation of Myosin II at the Leading Edge via a Noncanonical PLCγ/PKCα Pathway. Dev. Cell 2014, 31, 747–760. [Google Scholar] [CrossRef]
- Totsukawa, G.; Wu, Y.; Sasaki, Y.; Hartshorne, D.J.; Yamakita, Y.; Yamashiro, S.; Matsumura, F. Distinct roles of MLCK and ROCK in the regulation of membrane protrusions and focal adhesion dynamics during cell migration of fibroblasts. J. Cell Biol. 2004, 164, 427–439. [Google Scholar] [CrossRef] [PubMed]
- Sanders, L.C.; Matsumura, F.; Bokoch, G.M.; de Lanerolle, P. Inhibition of myosin light chain kinase by p21-activated kinase. Science 1999, 283, 2083–2085. [Google Scholar] [CrossRef] [PubMed]
- Dudek, S.M.; Jacobson, J.R.; Chiang, E.T.; Birukov, K.G.; Wang, P.; Zhan, X.; Garcia, J.G.N. Pulmonary Endothelial Cell Barrier Enhancement by Sphingosine 1-Phosphate. J. Biol. Chem. 2004, 279, 24692–24700. [Google Scholar] [CrossRef]
- Shin, D.H.; Chun, Y.-S.; Lee, D.S.; Huang, L.E.; Park, J.-W. Bortezomib inhibits tumor adaptation to hypoxia by stimulating the FIH-mediated repression of hypoxia-inducible factor-1. Blood 2008, 111, 3131–3136. [Google Scholar] [CrossRef] [PubMed]
- Haigo, S.L.; Hildebrand, J.D.; Harland, R.M.; Wallingford, J.B. Shroom Induces Apical Constriction and Is Required for Hingepoint Formation during Neural Tube Closure. Curr. Biol. 2003, 13, 2125–2137. [Google Scholar] [CrossRef]
- Hildebrand, J.D. Shroom regulates epithelial cell shape via the apical positioning of an actomyosin network. J. Cell Sci. 2005, 118, 5191–5203. [Google Scholar] [CrossRef]
- Murata-Hori, M.; Suizu, F.; Iwasaki, T.; Kikuchi, A.; Hosoya, H. ZIP kinase identified as a novel myosin regulatory light chain kinase in HeLa cells. FEBS Lett. 1999, 451, 81–84. [Google Scholar] [CrossRef]
- Dulyaninova, N.G.; Bresnick, A.R. The heavy chain has its day. BioArchitecture 2013, 3, 77–85. [Google Scholar] [CrossRef]
- Murakami, N.; Chauhan, V.P.; Elzinga, M. Two nonmuscle myosin II heavy chain isoforms expressed in rabbit brains: Filament forming properties, the effects of phosphorylation by protein kinase C and casein kinase II, and location of the phosphorylation sites. Biochemistry 1998, 37, 1989–2003. [Google Scholar] [CrossRef] [PubMed]
- Dulyaninova, N.G.; Malashkevich, V.N.; Almo, S.C.; Bresnick, A.R. Regulation of myosin-IIA assembly and Mts1 binding by heavy chain phosphorylation. Biochemistry 2005, 44, 6867–6876. [Google Scholar] [CrossRef] [PubMed]
- Clark, K.; Middelbeek, J.; Dorovkov, M.V.; Figdor, C.G.; Ryazanov, A.G.; Lasonder, E.; Van Leeuwen, F.N. The α-kinases TRPM6 and TRPM7, but not eEF-2 kinase, phosphorylate the assembly domain of myosin IIA, IIB and IIC. FEBS Lett. 2008, 582, 2993–2997. [Google Scholar] [CrossRef]
- Clark, K.; Middelbeek, J.; Lasonder, E.; Dulyaninova, N.G.; Morrice, N.A.; Ryazanov, A.G.; Bresnick, A.R.; Figdor, C.G.; Van Leeuwen, F.N. TRPM7 Regulates Myosin IIA Filament Stability and Protein Localization by Heavy Chain Phosphorylation. J. Mol. Biol. 2008, 378, 790–803. [Google Scholar] [CrossRef] [PubMed]
- Ronen, D.; Ravid, S. Myosin II Tailpiece Determines Its Paracrystal Structure, Filament Assembly Properties, and Cellular Localization. J. Biol. Chem. 2009, 284, 24948–24957. [Google Scholar] [CrossRef] [PubMed]
- Conti, M.A.; Sellers, J.R.; Adelstein, R.S.; Elzinga, M. Identification of the serine residue phosphorylated by protein kinase C in vertebrate nonmuscle myosin heavy chains. Biochemistry 1991, 30, 966–970. [Google Scholar] [CrossRef]
- Even-Faitelson, L.; Ravid, S. PAK1 and aPKCζ Regulate Myosin II-B Phosphorylation: A Novel Signaling Pathway Regulating Filament Assembly. Mol. Biol. Cell 2006, 17, 2869–2881. [Google Scholar] [CrossRef]
- Kawamoto, S.; Bengur, A.R.; Sellers, J.R.; Adelstein, R.S. In situ phosphorylation of human platelet myosin heavy and light chains by protein kinase C. J. Biol. Chem. 1989, 264, 2258–2265. [Google Scholar] [CrossRef]
- Moussavi, R.S.; Kelley, C.A.; Adelstein, R.S. Phosphorylation of vertebrate nonmuscle and smooth muscle myosin heavy chains and light chains. Mol. Cell. Biochem. 1993, 127–128, 219–227. [Google Scholar] [CrossRef]
- Ludowyke, R.I.; Elgundi, Z.; Kranenburg, T.; Stehn, J.R.; Schmitz-Peiffer, C.; Hughes, W.E.; Biden, T.J. Phosphorylation of Nonmuscle Myosin Heavy Chain IIA on Ser1917 Is Mediated by Protein Kinase CβII and Coincides with the Onset of Stimulated Degranulation of RBL-2H3 Mast Cells. J. Immunol. 2006, 177, 1492–1499. [Google Scholar] [CrossRef]
- Ruzzene, M.; Tosoni, K.; Zanin, S.; Cesaro, L.; Pinna, L.A. Protein kinase CK2 accumulation in “oncophilic” cells: Causes and effects. Mol. Cell. Biochem. 2011, 356, 5–10. [Google Scholar] [CrossRef]
- Borgo, C.; D’Amore, C.; Sarno, S.; Salvi, M.; Ruzzene, M. Protein kinase CK2: A potential therapeutic target for diverse human diseases. Signal Transduct. Target. Ther. 2021, 6, 183. [Google Scholar] [CrossRef]
- Dulyaninova, N.G.; House, R.P.; Betapudi, V.; Bresnick, A.R. Myosin-IIA Heavy-Chain Phosphorylation Regulates the Motility of MDA-MB-231 Carcinoma Cells. Mol. Biol. Cell 2007, 18, 3144–3155. [Google Scholar] [CrossRef] [PubMed]
- Beach, J.R.; Hussey, G.S.; Miller, T.E.; Chaudhury, A.; Patel, P.; Monslow, J.; Zheng, Q.; Keri, R.A.; Reizes, O.; Bresnick, A.R.; et al. Myosin II isoform switching mediates invasiveness after TGF-β–induced epithelial–mesenchymal transition. Proc. Natl. Acad. Sci. USA 2011, 108, 17991–17996. [Google Scholar] [CrossRef] [PubMed]
- Raab, M.; Swift, J.; Dingal, P.C.; Shah, P.; Shin, J.W.; Discher, D.E. Crawling from soft to stiff matrix polarizes the cytoskeleton and phosphoregulates myosin-II heavy chain. J. Cell Biol. 2012, 199, 669–683. [Google Scholar] [CrossRef]
- Rai, V.; Thomas, D.G.; Beach, J.R.; Egelhoff, T.T. Myosin IIA Heavy Chain Phosphorylation Mediates Adhesion Maturation and Protrusion in Three Dimensions. J. Biol. Chem. 2017, 292, 3099–3111. [Google Scholar] [CrossRef]
- Kollewe, A.; Chubanov, V.; Tseung, F.T.; Correia, L.; Schmidt, E.; Rossig, A.; Zierler, S.; Haupt, A.; Muller, C.S.; Bildl, W.; et al. The molecular appearance of native TRPM7 channel complexes identified by high-resolution proteomics. elife 2021, 10, e68544. [Google Scholar] [CrossRef] [PubMed]
- Fei, F.; Qu, J.; Li, C.; Wang, X.; Li, Y.; Zhang, S. Role of metastasis-induced protein S100A4 in human non-tumor pathophysiologies. Cell Biosci. 2017, 7, 64. [Google Scholar] [CrossRef]
- Kiss, B.; Duelli, A.; Radnai, L.; Kékesi, K.A.; Katona, G.; Nyitray, L. Crystal structure of the S100A4–nonmuscle myosin IIA tail fragment complex reveals an asymmetric target binding mechanism. Proc. Natl. Acad. Sci. USA 2012, 109, 6048–6053. [Google Scholar] [CrossRef]
- Ramagopal, U.A.; Dulyaninova, N.G.; Varney, K.M.; Wilder, P.T.; Nallamsetty, S.; Brenowitz, M.; Weber, D.J.; Almo, S.C.; Bresnick, A.R. Structure of the S100A4/myosin-IIA complex. BMC Struct. Biol. 2013, 13, 31. [Google Scholar] [CrossRef]
- Li, Z.-H.; Dulyaninova, N.G.; House, R.P.; Almo, S.C.; Bresnick, A.R. S100A4 Regulates Macrophage Chemotaxis. Mol. Biol. Cell 2010, 21, 2598–2610. [Google Scholar] [CrossRef] [PubMed]
- Dulyaninova, N.G.; Ruiz, P.D.; Gamble, M.J.; Backer, J.M.; Bresnick, A.R. S100A4 regulates macrophage invasion by distinct myosin-dependent and myosin-independent mechanisms. Mol. Biol. Cell 2018, 29, 632–642. [Google Scholar] [CrossRef]
- Davies, B.R.; O’Donnell, M.; Durkan, G.C.; Rudland, P.S.; Barraclough, R.; Neal, D.E.; Mellon, J.K. Expression of S100A4 protein is associated with metastasis and reduced survival in human bladder cancer. J. Pathol. 2002, 196, 292–299. [Google Scholar] [CrossRef] [PubMed]
- Du, M.; Wang, G.; Ismail, T.M.; Gross, S.; Fernig, D.G.; Barraclough, R.; Rudland, P.S. S100P Dissociates Myosin IIA Filaments and Focal Adhesion Sites to Reduce Cell Adhesion and Enhance Cell Migration. J. Biol. Chem. 2012, 287, 15330–15344. [Google Scholar] [CrossRef] [PubMed]
- Cao, F.; Miao, Y.; Xu, K.; Liu, P. Lethal (2) Giant Larvae: An Indispensable Regulator of Cell Polarity and Cancer Development. Int. J. Biol. Sci. 2015, 11, 380–389. [Google Scholar] [CrossRef]
- Dahan, I.; Petrov, D.; Cohen-Kfir, E.; Ravid, S. The tumor suppressor Lgl1 forms discrete complexes with NMII-A, and Par6α–aPKCζ that are affected by Lgl1 phosphorylation. J. Cell Sci. 2013, 127, 295–304. [Google Scholar] [CrossRef]
- Ravid, S. The tumor suppressor Lgl1 regulates front-rear polarity of migrating cells. Cell Adhes. Migr. 2014, 8, 378–383. [Google Scholar] [CrossRef]
- Lin, Y.-H.; Zhen, Y.-Y.; Chien, K.-Y.; Lee, I.C.; Lin, W.-C.; Chen, M.-Y.; Pai, L.-M. LIMCH1 regulates nonmuscle myosin-II activity and suppresses cell migration. Mol. Biol. Cell 2017, 28, 1054–1065. [Google Scholar] [CrossRef]
- Hosono, Y.; Usukura, J.; Yamaguchi, T.; Yanagisawa, K.; Suzuki, M.; Takahashi, T. MYBPH inhibits NM IIA assembly via direct interaction with NMHC IIA and reduces cell motility. Biochem. Biophys. Res. Commun. 2012, 428, 173–178. [Google Scholar] [CrossRef]
- Zhu, T.; He, Y.; Yang, J.; Fu, W.; Xu, X.; Si, Y. MYBPH inhibits vascular smooth muscle cell migration and attenuates neointimal hyperplasia in a rat carotid balloon-injury model. Exp. Cell Res. 2017, 359, 154–162. [Google Scholar] [CrossRef]
- Hundt, N.; Steffen, W.; Pathan-Chhatbar, S.; Taft, M.H.; Manstein, D.J. Load-dependent modulation of non-muscle myosin-2A function by tropomyosin 4.2. Sci. Rep. 2016, 6, 20554. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Jana, S.S.; Anne Conti, M.; Kawamoto, S.; Claycomb, W.C.; Adelstein, R.S. Ablation of Nonmuscle Myosin II-B and II-C Reveals a Role for Nonmuscle Myosin II in Cardiac Myocyte Karyokinesis. Mol. Biol. Cell 2010, 21, 3952–3962. [Google Scholar] [CrossRef]
- Beach, J.R.; Hammer, J.A. Myosin II isoform co-assembly and differential regulation in mammalian systems. Exp. Cell Res. 2015, 334, 2–9. [Google Scholar] [CrossRef] [PubMed]
- Beach, J.R.; Shao, L.; Remmert, K.; Li, D.; Betzig, E.; Hammer, J.A. Nonmuscle Myosin II Isoforms Coassemble in Living Cells. Curr. Biol. 2014, 24, 1160–1166. [Google Scholar] [CrossRef]
- Shutova, M.S.; Spessott, W.A.; Giraudo, C.G.; Svitkina, T. Endogenous Species of Mammalian Nonmuscle Myosin IIA and IIB Include Activated Monomers and Heteropolymers. Curr. Biol. 2014, 24, 1958–1968. [Google Scholar] [CrossRef]
- Shutova, M.S.; Asokan, S.B.; Talwar, S.; Assoian, R.K.; Bear, J.E.; Svitkina, T.M. Self-sorting of nonmuscle myosins IIA and IIB polarizes the cytoskeleton and modulates cell motility. J. Cell Biol. 2017, 216, 2877–2889. [Google Scholar] [CrossRef] [PubMed]
- Billington, N.; Beach, J.R.; Heissler, S.M.; Remmert, K.; Guzik-Lendrum, S.; Nagy, A.; Takagi, Y.; Shao, L.; Li, D.; Yang, Y.; et al. Myosin 18A Coassembles with Nonmuscle Myosin 2 to Form Mixed Bipolar Filaments. Curr. Biol. 2015, 25, 942–948. [Google Scholar] [CrossRef]
- Ma, X.; Bao, J.; Adelstein, R.S. Loss of Cell Adhesion Causes Hydrocephalus in Nonmuscle Myosin II-B–ablated and Mutated Mice. Mol. Biol. Cell 2007, 18, 2305–2312. [Google Scholar] [CrossRef]
- Conti, M.A.; Even-Ram, S.; Liu, C.; Yamada, K.M.; Adelstein, R.S. Defects in cell adhesion and the visceral endoderm following ablation of nonmuscle myosin heavy chain II-A in mice. J. Biol. Chem. 2004, 279, 41263–41266. [Google Scholar] [CrossRef]
- Guo, S.; Kemphues, K.J. A non-muscle myosin required for embryonic polarity in Caenorhabditis elegans. Nature 1996, 382, 455–458. [Google Scholar] [CrossRef]
- Osorio, D.S.; Chan, F.Y.; Saramago, J.; Leite, J.; Silva, A.M.; Sobral, A.F.; Gassmann, R.; Carvalho, A.X. Crosslinking activity of non-muscle myosin II is not sufficient for embryonic cytokinesis in C. elegans. Development 2019, 146, dev179150. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Conti, M.A.; Malide, D.; Dong, F.; Wang, A.; Shmist, Y.A.; Liu, C.; Zerfas, P.; Daniels, M.P.; Chan, C.-C.; et al. Mouse models of MYH9-related disease: Mutations in nonmuscle myosin II-A. Blood 2012, 119, 238–250. [Google Scholar] [CrossRef]
- Lienkamp, S.S.; Liu, K.; Karner, C.M.; Carroll, T.J.; Ronneberger, O.; Wallingford, J.B.; Walz, G. Vertebrate kidney tubules elongate using a planar cell polarity–dependent, rosette-based mechanism of convergent extension. Nat. Genet. 2012, 44, 1382–1387. [Google Scholar] [CrossRef] [PubMed]
- Miura, K.; Kurihara, H.; Horita, S.; Chikamoto, H.; Hattori, M.; Harita, Y.; Tsurumi, H.; Kajiho, Y.; Sawada, Y.; Sasaki, S.; et al. Podocyte expression of nonmuscle myosin heavy chain-IIA decreases in idiopathic nephrotic syndrome, especially in focal segmental glomerulosclerosis. Nephrol. Dial. Transplant. 2013, 28, 2993–3003. [Google Scholar] [CrossRef]
- Lee, J.; Andreeva, A.; Sipe, C.W.; Liu, L.; Cheng, A.; Lu, X. PTK7 Regulates Myosin II Activity to Orient Planar Polarity in the Mammalian Auditory Epithelium. Curr. Biol. 2012, 22, 956–966. [Google Scholar] [CrossRef]
- Yamamoto, N.; Okano, T.; Ma, X.; Adelstein, R.S.; Kelley, M.W. Myosin II regulates extension, growth and patterning in the mammalian cochlear duct. Development 2009, 136, 1977–1986. [Google Scholar] [CrossRef]
- Straight, A.F.; Cheung, A.; Limouze, J.; Chen, I.; Westwood, N.J.; Sellers, J.R.; Mitchison, T.J. Dissecting temporal and spatial control of cytokinesis with a myosin II Inhibitor. Science 2003, 299, 1743–1747. [Google Scholar] [CrossRef] [PubMed]
- De Lozanne, A.; Spudich, J.A. Disruption of the Dictyostelium myosin heavy chain gene by homologous recombination. Science 1987, 236, 1086–1091. [Google Scholar] [CrossRef]
- Mabuchi, I.; Okuno, M. The effect of myosin antibody on the division of starfish blastomeres. J. Cell Biol. 1977, 74, 251–263. [Google Scholar] [CrossRef]
- Fujiwara, K.; Pollard, T.D. Fluorescent antibody localization of myosin in the cytoplasm, cleavage furrow, and mitotic spindle of human cells. J. Cell Biol. 1976, 71, 848–875. [Google Scholar] [CrossRef]
- Green, R.A.; Paluch, E.; Oegema, K. Cytokinesis in animal cells. Annu. Rev. Cell Dev. Biol. 2012, 28, 29–58. [Google Scholar] [CrossRef] [PubMed]
- Descovich, C.P.; Cortes, D.B.; Ryan, S.; Nash, J.; Zhang, L.; Maddox, P.S.; Nedelec, F.; Maddox, A.S. Cross-linkers both drive and brake cytoskeletal remodeling and furrowing in cytokinesis. Mol. Biol. Cell 2018, 29, 622–631. [Google Scholar] [CrossRef]
- Reymann, A.C.; Staniscia, F.; Erzberger, A.; Salbreux, G.; Grill, S.W. Cortical flow aligns actin filaments to form a furrow. elife 2016, 5, e17807. [Google Scholar] [CrossRef] [PubMed]
- Davies, T.; Jordan, S.N.; Chand, V.; Sees, J.A.; Laband, K.; Carvalho, A.X.; Shirasu-Hiza, M.; Kovar, D.R.; Dumont, J.; Canman, J.C. High-Resolution Temporal Analysis Reveals a Functional Timeline for the Molecular Regulation of Cytokinesis. Dev. Cell 2014, 30, 209–223. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Adelstein, R.S. The role of vertebrate nonmuscle Myosin II in development and human disease. BioArchitecture 2014, 4, 88–102. [Google Scholar] [CrossRef] [PubMed]
- Tsankova, A.; Pham, T.T.; Garcia, D.S.; Otte, F.; Cabernard, C. Cell Polarity Regulates Biased Myosin Activity and Dynamics during Asymmetric Cell Division via Drosophila Rho Kinase and Protein Kinase N. Dev. Cell 2017, 42, 143–155.e145. [Google Scholar] [CrossRef]
- Sugioka, K.; Bowerman, B. Combinatorial Contact Cues Specify Cell Division Orientation by Directing Cortical Myosin Flows. Dev. Cell 2018, 46, 257–270.e255. [Google Scholar] [CrossRef]
- Sun, Z.; Guo, S.S.; Fässler, R. Integrin-mediated mechanotransduction. J. Cell Biol. 2016, 215, 445–456. [Google Scholar] [CrossRef]
- Kim, K.Y.; Kovacs, M.; Kawamoto, S.; Sellers, J.R.; Adelstein, R.S. Disease-associated mutations and alternative splicing alter the enzymatic and motile activity of nonmuscle myosins II-B and II-C. J. Biol. Chem. 2005, 280, 22769–22775. [Google Scholar] [CrossRef]
- Thomas, D.G.; Yenepalli, A.; Denais, C.M.; Rape, A.; Beach, J.R.; Wang, Y.-L.; Schiemann, W.P.; Baskaran, H.; Lammerding, J.; Egelhoff, T.T. Non-muscle myosin IIB is critical for nuclear translocation during 3D invasion. J. Cell Biol. 2015, 210, 583–594. [Google Scholar] [CrossRef]
- Cai, Y.; Biais, N.; Giannone, G.; Tanase, M.; Jiang, G.; Hofman, J.M.; Wiggins, C.H.; Silberzan, P.; Buguin, A.; Ladoux, B.; et al. Nonmuscle Myosin IIA-Dependent Force Inhibits Cell Spreading and Drives F-Actin Flow. Biophys. J. 2006, 91, 3907–3920. [Google Scholar] [CrossRef]
- Hartsock, A.; Nelson, W.J. Adherens and tight junctions: Structure, function and connections to the actin cytoskeleton. Biochim. Et Biophys. Acta (BBA) Biomembr. 2008, 1778, 660–669. [Google Scholar] [CrossRef] [PubMed]
- Mui, K.L.; Chen, C.S.; Assoian, R.K. The mechanical regulation of integrin–cadherin crosstalk organizes cells, signaling and forces. J. Cell Sci. 2016, 129, 1093–1100. [Google Scholar] [CrossRef] [PubMed]
- Pasapera, A.M.; Schneider, I.C.; Rericha, E.; Schlaepfer, D.D.; Waterman, C.M. Myosin II activity regulates vinculin recruitment to focal adhesions through FAK-mediated paxillin phosphorylation. J. Cell Biol. 2010, 188, 877–890. [Google Scholar] [CrossRef] [PubMed]
- Curran, S.; Strandkvist, C.; Bathmann, J.; de Gennes, M.; Kabla, A.; Salbreux, G.; Baum, B. Myosin II Controls Junction Fluctuations to Guide Epithelial Tissue Ordering. Dev. Cell 2017, 43, 480–492.e486. [Google Scholar] [CrossRef] [PubMed]
- Ladoux, B.; Mège, R.-M. Mechanobiology of collective cell behaviours. Nat. Rev. Mol. Cell Biol. 2017, 18, 743–757. [Google Scholar] [CrossRef]
- Smutny, M.; Cox, H.L.; Leerberg, J.M.; Kovacs, E.M.; Conti, M.A.; Ferguson, C.; Hamilton, N.A.; Parton, R.G.; Adelstein, R.S.; Yap, A.S. Myosin II isoforms identify distinct functional modules that support integrity of the epithelial zonula adherens. Nat. Cell Biol. 2010, 12, 696–702. [Google Scholar] [CrossRef]
- Vicente-Manzanares, M.; Ma, X.; Adelstein, R.S.; Horwitz, A.R. Non-muscle myosin II takes centre stage in cell adhesion and migration. Nat. Rev. Mol. Cell Biol. 2009, 10, 778–790. [Google Scholar] [CrossRef]
- Vicente-Manzanares, M.; Zareno, J.; Whitmore, L.; Choi, C.K.; Horwitz, A.F. Regulation of protrusion, adhesion dynamics, and polarity by myosins IIA and IIB in migrating cells. J. Cell Biol. 2007, 176, 573–580. [Google Scholar] [CrossRef]
- Heuze, M.L.; Sankara Narayana, G.H.N.; D’Alessandro, J.; Cellerin, V.; Dang, T.; Williams, D.S.; Van Hest, J.C.; Marcq, P.; Mege, R.M.; Ladoux, B. Myosin II isoforms play distinct roles in adherens junction biogenesis. elife 2019, 8, e46599. [Google Scholar] [CrossRef]
- Ivanov, A.I.; Bachar, M.; Babbin, B.A.; Adelstein, R.S.; Nusrat, A.; Parkos, C.A. A Unique Role for Nonmuscle Myosin Heavy Chain IIA in Regulation of Epithelial Apical Junctions. PLoS ONE 2007, 2, e658. [Google Scholar] [CrossRef] [PubMed]
- Ruprecht, V.; Wieser, S.; Callan-Jones, A.; Smutny, M.; Morita, H.; Sako, K.; Barone, V.; Ritsch-Marte, M.; Sixt, M.; Voituriez, R.; et al. Cortical Contractility Triggers a Stochastic Switch to Fast Amoeboid Cell Motility. Cell 2015, 160, 673–685. [Google Scholar] [CrossRef]
- Shutova, M.S.; Svitkina, T.M. Common and Specific Functions of Nonmuscle Myosin II Paralogs in Cells. Biochemistry 2018, 83, 1459–1468. [Google Scholar] [CrossRef] [PubMed]
- Barbier, L.; Sáez, P.J.; Attia, R.; Lennon-Duménil, A.-M.; Lavi, I.; Piel, M.; Vargas, P. Myosin II Activity Is Selectively Needed for Migration in Highly Confined Microenvironments in Mature Dendritic Cells. Front. Immunol. 2019, 10, 747. [Google Scholar] [CrossRef] [PubMed]
- Schaub, S.; Bohnet, S.; Laurent, V.M.; Meister, J.-J.; Verkhovsky, A.B. Comparative Maps of Motion and Assembly of Filamentous Actin and Myosin II in Migrating Cells. Mol. Biol. Cell 2007, 18, 3723–3732. [Google Scholar] [CrossRef]
- Wolf, K.; Te Lindert, M.; Krause, M.; Alexander, S.; Te Riet, J.; Willis, A.L.; Hoffman, R.M.; Figdor, C.G.; Weiss, S.J.; Friedl, P. Physical limits of cell migration: Control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J. Cell Biol. 2013, 201, 1069–1084. [Google Scholar] [CrossRef]
- Gardel, M.L.; Sabass, B.; Ji, L.; Danuser, G.; Schwarz, U.S.; Waterman, C.M. Traction stress in focal adhesions correlates biphasically with actin retrograde flow speed. J. Cell Biol. 2008, 183, 999–1005. [Google Scholar] [CrossRef]
- Alexandrova, A.Y.; Arnold, K.; Schaub, S.; Vasiliev, J.M.; Meister, J.-J.; Bershadsky, A.D.; Verkhovsky, A.B. Comparative Dynamics of Retrograde Actin Flow and Focal Adhesions: Formation of Nascent Adhesions Triggers Transition from Fast to Slow Flow. PLoS ONE 2008, 3, e3234. [Google Scholar] [CrossRef]
- Lo, C.-M.; Buxton, D.B.; Chua, G.C.H.; Dembo, M.; Adelstein, R.S.; Wang, Y.-L. Nonmuscle Myosin IIB Is Involved in the Guidance of Fibroblast Migration. Mol. Biol. Cell 2004, 15, 982–989. [Google Scholar] [CrossRef]
- Betapudi, V.; Licate, L.S.; Egelhoff, T.T. Distinct Roles of Nonmuscle Myosin II Isoforms in the Regulation of MDA-MB-231 Breast Cancer Cell Spreading and Migration. Cancer Res. 2006, 66, 4725–4733. [Google Scholar] [CrossRef]
- Liu, Z.; Ho, C.-H.; Grinnell, F. The different roles of myosin IIA and myosin IIB in contraction of 3D collagen matrices by human fibroblasts. Exp. Cell Res. 2014, 326, 295–306. [Google Scholar] [CrossRef]
- Even-Ram, S.; Doyle, A.D.; Conti, M.A.; Matsumoto, K.; Adelstein, R.S.; Yamada, K.M. Myosin IIA regulates cell motility and actomyosin–microtubule crosstalk. Nat. Cell Biol. 2007, 9, 299–309. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.-J.; Le Berre, M.; Lautenschlaeger, F.; Maiuri, P.; Callan-Jones, A.; Heuzé, M.; Takaki, T.; Voituriez, R.; Piel, M. Confinement and Low Adhesion Induce Fast Amoeboid Migration of Slow Mesenchymal Cells. Cell 2015, 160, 659–672. [Google Scholar] [CrossRef] [PubMed]
- Svitkina, T.M.; Verkhovsky, A.B.; McQuade, K.M.; Borisy, G.G. Analysis of the Actin–Myosin II System in Fish Epidermal Keratocytes: Mechanism of Cell Body Translocation. J. Cell Biol. 1997, 139, 397–415. [Google Scholar] [CrossRef]
- Agarwal, P.; Zaidel-Bar, R. Diverse roles of non-muscle myosin II contractility in 3D cell migration. Essays Biochem. 2019, 63, 497–508. [Google Scholar] [CrossRef] [PubMed]
- Nagy, S.; Ricca, B.L.; Norstrom, M.F.; Courson, D.S.; Brawley, C.M.; Smithback, P.A.; Rock, R.S. A myosin motor that selects bundled actin for motility. Proc. Natl. Acad. Sci. USA 2008, 105, 9616–9620. [Google Scholar] [CrossRef]
- Hind, L.E.; Vincent, W.J.; Huttenlocher, A. Leading from the Back: The Role of the Uropod in Neutrophil Polarization and Migration. Dev. Cell 2016, 38, 161–169. [Google Scholar] [CrossRef]
- Yoshida, K.; Soldati, T. Dissection of amoeboid movement into two mechanically distinct modes. J. Cell Sci. 2006, 119, 3833–3844. [Google Scholar] [CrossRef]
- Charras, G.; Paluch, E. Blebs lead the way: How to migrate without lamellipodia. Nat. Rev. Mol. Cell Biol. 2008, 9, 730–736. [Google Scholar] [CrossRef]
- Sánchez-Madrid, F.; Serrador, J.M. Bringing up the rear: Defining the roles of the uropod. Nat. Rev. Mol. Cell Biol. 2009, 10, 353–359. [Google Scholar] [CrossRef]
- Conrad, G.W.; Davis, S.E. Polar lobe formation and cytokinesis in fertilized eggs of Ilyanassa obsoleta. III. Large bleb formation caused by Sr2+, ionophores X537A and A23187, and compound 48/80. Dev. Biol. 1980, 74, 152–172. [Google Scholar] [CrossRef] [PubMed]
- Charras, G.T. A short history of blebbing. J. Microsc. 2008, 231, 466–478. [Google Scholar] [CrossRef] [PubMed]
- Fackler, O.T.; Grosse, R. Cell motility through plasma membrane blebbing. J. Cell Biol. 2008, 181, 879–884. [Google Scholar] [CrossRef] [PubMed]
- Rottner, K.; Schaks, M. Assembling actin filaments for protrusion. Curr. Opin. Cell Biol. 2019, 56, 53–63. [Google Scholar] [CrossRef]
- de Lucas, B.; Bernal, A.; Pérez, L.M.; Martín, N.S.; Gálvez, B.G. Membrane Blebbing Is Required for Mesenchymal Precursor Migration. PLoS ONE 2016, 11, e0150004. [Google Scholar] [CrossRef]
- Charras, G.T.; Yarrow, J.C.; Horton, M.A.; Mahadevan, L.; Mitchison, T.J. Non-equilibration of hydrostatic pressure in blebbing cells. Nature 2005, 435, 365–369. [Google Scholar] [CrossRef]
- Ikenouchi, J.; Aoki, K. Membrane bleb: A seesaw game of two small GTPases. Small GTPases 2017, 8, 85–89. [Google Scholar] [CrossRef]
- Tinevez, J.-Y.; Schulze, U.; Salbreux, G.; Roensch, J.; Joanny, J.-F.; Paluch, E. Role of cortical tension in bleb growth. Proc. Natl. Acad. Sci. USA 2009, 106, 18581–18586. [Google Scholar] [CrossRef]
- Ridley, A. Blebs on the move. Dev. Cell 2011, 20, e1. [Google Scholar] [CrossRef]
- Gong, X.; Didan, Y.; Lock, J.G.; Stromblad, S. KIF13A-regulated RhoB plasma membrane localization governs membrane blebbing and blebby amoeboid cell migration. EMBO J. 2018, 37, e98994. [Google Scholar] [CrossRef]
- Norman, L.; Sengupta, K.; Aranda-Espinoza, H. Blebbing dynamics during endothelial cell spreading. Eur. J. Cell Biol. 2011, 90, 37–48. [Google Scholar] [CrossRef] [PubMed]
- Charras, G.T.; Coughlin, M.; Mitchison, T.J.; Mahadevan, L. Life and Times of a Cellular Bleb. Biophys. J. 2008, 94, 1836–1853. [Google Scholar] [CrossRef] [PubMed]
- Charras, G.T.; Hu, C.-K.; Coughlin, M.; Mitchison, T.J. Reassembly of contractile actin cortex in cell blebs. J. Cell Biol. 2006, 175, 477–490. [Google Scholar] [CrossRef]
- Sheetz, M.P.; Sable, J.E.; Dobereiner, H.G. Continuous membrane-cytoskeleton adhesion requires continuous accommodation to lipid and cytoskeleton dynamics. Annu. Rev. Biophys. Biomol. Struct. 2006, 35, 417–434. [Google Scholar] [CrossRef]
- Taneja, N.; Burnette, D.T. Myosin IIA drives membrane bleb retraction. Mol. Biol. Cell 2019, 30, 1051–1059. [Google Scholar] [CrossRef]
- Brito, C.; Mesquita, F.S.; Bleck, C.K.E.; Sellers, J.R.; Cabanes, D.; Sousa, S. Perfringolysin O-Induced Plasma Membrane Pores Trigger Actomyosin Remodeling and Endoplasmic Reticulum Redistribution. Toxins 2019, 11, 419. [Google Scholar] [CrossRef]
- Arous, C.; Rondas, D.; Halban, P.A. Non-muscle myosin IIA is involved in focal adhesion and actin remodelling controlling glucose-stimulated insulin secretion. Diabetologia 2013, 56, 792–802. [Google Scholar] [CrossRef] [PubMed]
- Saito, T.; Okada, S.; Shimoda, Y.; Tagaya, Y.; Osaki, A.; Yamada, E.; Shibusawa, R.; Nakajima, Y.; Ozawa, A.; Satoh, T.; et al. APPL1 promotes glucose uptake in response to mechanical stretch via the PKCzeta-non-muscle myosin IIa pathway in C2C12 myotubes. Cell Signal 2016, 28, 1694–1702. [Google Scholar] [CrossRef]
- Singh, K.; Kim, A.B.; Morgan, K.G. Non-muscle myosin II regulates aortic stiffness through effects on specific focal adhesion proteins and the non-muscle cortical cytoskeleton. J. Cell. Mol. Med. 2021, 25, 2471–2483. [Google Scholar] [CrossRef]
- Fenix, A.M.; Taneja, N.; Buttler, C.A.; Lewis, J.; Van Engelenburg, S.B.; Ohi, R.; Burnette, D.T. Expansion and concatenation of nonmuscle myosin IIA filaments drive cellular contractile system formation during interphase and mitosis. Mol. Biol. Cell 2016, 27, 1465–1478. [Google Scholar] [CrossRef]
- Nangia-Makker, P.; Shekhar, M.P.V.; Hogan, V.; Balan, V.; Raz, A. MYH9 binds to dNTPs via deoxyribose moiety and plays an important role in DNA synthesis. Oncotarget 2022, 13, 534–550. [Google Scholar] [CrossRef] [PubMed]
- Bondzie, P.A.; Chen, H.A.; Cao, M.Z.; Tomolonis, J.A.; He, F.; Pollak, M.R.; Henderson, J.M. Non-muscle myosin-IIA is critical for podocyte f-actin organization, contractility, and attenuation of cell motility. Cytoskeleton 2016, 73, 377–395. [Google Scholar] [CrossRef]
- Moura, P.L.; Hawley, B.R.; Mankelow, T.J.; Griffiths, R.E.; Dobbe, J.G.G.; Streekstra, G.J.; Anstee, D.J.; Satchwell, T.J.; Toye, A.M. Non-muscle myosin II drives vesicle loss during human reticulocyte maturation. Haematologica 2018, 103, 1997–2007. [Google Scholar] [CrossRef] [PubMed]
- Javier-Torrent, M.; Marco, S.; Rocandio, D.; Pons-Vizcarra, M.; Janes, P.W.; Lackmann, M.; Egea, J.; Saura, C.A. Presenilin/gamma-secretase-dependent EphA3 processing mediates axon elongation through non-muscle myosin IIA. elife 2019, 8, e43646. [Google Scholar] [CrossRef]
- Müller, T.; Rumpel, E.; Hradetzky, S.; Bollig, F.; Wegner, H.; Blumenthal, A.; Greinacher, A.; Endlich, K.; Endlich, N. Non-muscle myosin IIA is required for the development of the zebrafish glomerulus. Kidney Int. 2011, 80, 1055–1063. [Google Scholar] [CrossRef]
- Gutzman, J.H.; Sahu, S.U.; Kwas, C. Non-muscle myosin IIA and IIB differentially regulate cell shape changes during zebrafish brain morphogenesis. Dev. Biol. 2015, 397, 103–115. [Google Scholar] [CrossRef] [PubMed]
- Arora, P.D.; Wang, Y.; Janmey, P.A.; Bresnick, A.; Yin, H.L.; McCulloch, C.A. Gelsolin and non-muscle myosin IIA interact to mediate calcium-regulated collagen phagocytosis. J. Biol. Chem. 2011, 286, 34184–34198. [Google Scholar] [CrossRef]
- You, G.-R.; Chang, J.T.; Li, Y.-L.; Huang, C.-W.; Tsai, Y.-L.; Fan, K.-H.; Kang, C.-J.; Huang, S.-F.; Chang, P.-H.; Cheng, A.-J. MYH9 Facilitates Cell Invasion and Radioresistance in Head and Neck Cancer via Modulation of Cellular ROS Levels by Activating the MAPK-Nrf2-GCLC Pathway. Cells 2022, 11, 2855. [Google Scholar] [CrossRef]
- Coaxum, S.D.; Tiedeken, J.; Garrett-Mayer, E.; Myers, J.; Rosenzweig, S.A.; Neskey, D.M. The tumor suppressor capability of p53 is dependent on non-muscle myosin IIA function in head and neck cancer. Oncotarget 2017, 8, 22991–23007. [Google Scholar] [CrossRef]
- Schramek, D.; Sendoel, A.; Segal, J.P.; Beronja, S.; Heller, E.; Oristian, D.; Reva, B.; Fuchs, E. Direct in Vivo RNAi Screen Unveils Myosin IIa as a Tumor Suppressor of Squamous Cell Carcinomas. Science 2014, 343, 309–313. [Google Scholar] [CrossRef]
- Anne Conti, M.; Saleh, A.D.; Brinster, L.R.; Cheng, H.; Chen, Z.; Cornelius, S.; Liu, C.; Ma, X.; Van Waes, C.; Adelstein, R.S. Conditional deletion of nonmuscle myosin II-A in mouse tongue epithelium results in squamous cell carcinoma. Sci. Rep. 2015, 5, 14068. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.K.; Sinha, S.; Padhan, J.; Jangde, N.; Ray, R.; Rai, V. MYH9 suppresses melanoma tumorigenesis, metastasis and regulates tumor microenvironment. Med. Oncol. 2020, 37, 88. [Google Scholar] [CrossRef] [PubMed]
- Yao, H.; Liu, J.; Zhang, C.; Shao, Y.; Li, X.; Yu, Z.; Huang, Y. Apatinib inhibits glioma cell malignancy in patient-derived orthotopic xenograft mouse model by targeting thrombospondin 1/myosin heavy chain 9 axis. Cell Death Dis. 2021, 12, 927. [Google Scholar] [CrossRef] [PubMed]
- Inukai, M.; Yokoi, A.; Ishizuka, Y.; Hashimura, M.; Matsumoto, T.; Oguri, Y.; Nakagawa, M.; Ishibashi, Y.; Ito, T.; Kumabe, T.; et al. A functional role of S100A4/non-muscle myosin IIA axis for pro-tumorigenic vascular functions in glioblastoma. Cell Commun. Signal. 2022, 20, 46. [Google Scholar] [CrossRef]
- Liu, Y.; Jiang, Q.; Liu, X.; Lin, X.; Tang, Z.; Liu, C.; Zhou, J.; Zhao, M.; Li, X.; Cheng, Z.; et al. Cinobufotalin powerfully reversed EBV-miR-BART22-induced cisplatin resistance via stimulating MAP2K4 to antagonize non-muscle myosin heavy chain IIA/glycogen synthase 3β/β-catenin signaling pathway. EBioMedicine 2019, 48, 386–404. [Google Scholar] [CrossRef]
- Lu, H.; Huang, H. FOXO1: A Potential Target for Human Diseases. Curr. Drug Targets 2011, 12, 1235–1244. [Google Scholar] [CrossRef]
- Li, Y.; Liu, X.; Lin, X.; Zhao, M.; Xiao, Y.; Liu, C.; Liang, Z.; Lin, Z.; Yi, R.; Tang, Z.; et al. Chemical compound cinobufotalin potently induces FOXO1-stimulated cisplatin sensitivity by antagonizing its binding partner MYH9. Signal Transduct. Target. Ther. 2019, 4, 48. [Google Scholar] [CrossRef]
- Zhao, M.; Luo, R.; Liu, Y.; Gao, L.; Fu, Z.; Fu, Q.; Luo, X.; Chen, Y.; Deng, X.; Liang, Z.; et al. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR–p-PI3K/AKT-c-JUN. Nat. Commun. 2016, 7, 11309. [Google Scholar] [CrossRef]
- Liang, Z.; Liu, Z.; Cheng, C.; Wang, H.; Deng, X.; Liu, J.; Liu, C.; Li, Y.; Fang, W. VPS33B interacts with NESG1 to modulate EGFR/PI3K/AKT/c-Myc/P53/miR-133a-3p signaling and induce 5-fluorouracil sensitivity in nasopharyngeal carcinoma. Cell Death Dis. 2019, 10, 305. [Google Scholar] [CrossRef]
- Katono, K.; Sato, Y.; Jiang, S.-X.; Kobayashi, M.; Nagashio, R.; Ryuge, S.; Fukuda, E.; Goshima, N.; Satoh, Y.; Saegusa, M.; et al. Prognostic Significance of MYH9 Expression in Resected Non-Small Cell Lung Cancer. PLoS ONE 2015, 10, e0121460. [Google Scholar] [CrossRef]
- Chen, M.; Sun, L.-X.; Yu, L.; Liu, J.; Sun, L.-C.; Yang, Z.-H.; Shu, X.; Ran, Y.-L. MYH9 is crucial for stem cell-like properties in non-small cell lung cancer by activating mTOR signaling. Cell Death Discov. 2021, 7, 282. [Google Scholar] [CrossRef] [PubMed]
- Chiu, H.-C.; Chang, T.-Y.; Huang, C.-T.; Chao, Y.-S.; Hsu, J.T.A. EGFR and myosin II inhibitors cooperate to suppress EGFR-T790M-mutant NSCLC cells. Mol. Oncol. 2012, 6, 299–310. [Google Scholar] [CrossRef]
- Zhou, W.; Liu, Y.; Gao, Y.; Cheng, Y.; Chang, R.; Li, X.; Zhou, Y.; Wang, S.; Liang, L.; Duan, C.; et al. MICAL2 is a novel nucleocytoplasmic shuttling protein promoting cancer invasion and growth of lung adenocarcinoma. Cancer Lett. 2020, 483, 75–86. [Google Scholar] [CrossRef] [PubMed]
- Lin, X.; Li, A.-M.; Li, Y.-H.; Luo, R.-C.; Zou, Y.-J.; Liu, Y.-Y.; Liu, C.; Xie, Y.-Y.; Zuo, S.; Liu, Z.; et al. Silencing MYH9 blocks HBx-induced GSK3β ubiquitination and degradation to inhibit tumor stemness in hepatocellular carcinoma. Signal Transduct. Target. Ther. 2020, 5, 13. [Google Scholar] [CrossRef] [PubMed]
- Hou, R.; Li, Y.; Luo, X.; Zhang, W.; Yang, H.; Zhang, Y.; Liu, J.; Liu, S.; Han, S.; Liu, C.; et al. ENKUR expression induced by chemically synthesized cinobufotalin suppresses malignant activities of hepatocellular carcinoma by modulating β-catenin/c-Jun/MYH9/USP7/c-Myc axis. Int. J. Biol. Sci. 2022, 18, 2553–2567. [Google Scholar] [CrossRef]
- Xia, Z.K.; Yuan, Y.C.; Yin, N.; Yin, B.L.; Tan, Z.P.; Hu, Y.R. Nonmuscle myosin IIA is associated with poor prognosis of esophageal squamous cancer. Dis. Esophagus 2012, 25, 427–436. [Google Scholar] [CrossRef]
- Yang, B.; Liu, H.; Bi, Y.; Cheng, C.; Li, G.; Kong, P.; Zhang, L.; Shi, R.; Zhang, Y.; Zhang, R.; et al. MYH9 promotes cell metastasis via inducing Angiogenesis and Epithelial Mesenchymal Transition in Esophageal Squamous Cell Carcinoma. Int. J. Med. Sci. 2020, 17, 2013–2023. [Google Scholar] [CrossRef]
- Pan, B.Q.; Xie, Z.H.; Hao, J.J.; Zhang, Y.; Xu, X.; Cai, Y.; Wang, M.R. PTP1B up-regulates EGFR expression by dephosphorylating MYH9 at Y1408 to promote cell migration and invasion in esophageal squamous cell carcinoma. Biochem. Biophys. Res. Commun. 2020, 522, 53–60. [Google Scholar] [CrossRef] [PubMed]
- Zhou, P.; Li, Y.; Li, B.; Zhang, M.; Liu, Y.; Yao, Y.; Li, D. NMIIA promotes tumor growth and metastasis by activating the Wnt/β-catenin signaling pathway and EMT in pancreatic cancer. Oncogene 2019, 38, 5500–5515. [Google Scholar] [CrossRef]
- Ye, G.; Yang, Q.; Lei, X.; Zhu, X.; Li, F.; He, J.; Chen, H.; Ling, R.; Zhang, H.; Lin, T.; et al. Nuclear MYH9-induced CTNNB1 transcription, targeted by staurosporin, promotes gastric cancer cell anoikis resistance and metastasis. Theranostics 2020, 10, 7545–7560. [Google Scholar] [CrossRef]
- Liu, T.; Ye, Y.; Zhang, X.; Zhu, A.; Yang, Z.; Fu, Y.; Wei, C.; Liu, Q.; Zhao, C.; Wang, G. Downregulation of non-muscle myosin IIA expression inhibits migration and invasion of gastric cancer cells via the c-Jun N-terminal kinase signaling pathway. Mol. Med. Rep. 2016, 13, 1639–1644. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Qi, X.; Liu, J.; Zhou, R.; Lin, C.; Shangguan, J.; Zhang, Z.; Zhao, L.; Li, G. MYH9 Promotes Growth and Metastasis via Activation of MAPK/AKT Signaling in Colorectal Cancer. J. Cancer 2019, 10, 874–884. [Google Scholar] [CrossRef] [PubMed]
- Zhong, Y.; Long, T.; Gu, C.-S.; Tang, J.-Y.; Gao, L.-F.; Zhu, J.-X.; Hu, Z.-Y.; Wang, X.; Ma, Y.-D.; Ding, Y.-Q.; et al. MYH9-dependent polarization of ATG9B promotes colorectal cancer metastasis by accelerating focal adhesion assembly. Cell Death Differ. 2021, 28, 3251–3269. [Google Scholar] [CrossRef] [PubMed]
- Liao, Q.; Li, R.; Zhou, R.; Pan, Z.; Xu, L.; Ding, Y.; Zhao, L. LIM kinase 1 interacts with myosin-9 and alpha-actinin-4 and promotes colorectal cancer progression. Br. J. Cancer 2017, 117, 563–571. [Google Scholar] [CrossRef] [PubMed]
- Choi, C.; Kwon, J.; Lim, S.; Helfman, D.M. Integrin β1, myosin light chain kinase and myosin IIA are required for activation of PI3K-AKT signaling following MEK inhibition in metastatic triple negative breast cancer. Oncotarget 2016, 7, 63466–63487. [Google Scholar] [CrossRef] [PubMed]
- Hindman, B.; Goeckeler, Z.; Sierros, K.; Wysolmerski, R. Non-Muscle Myosin II Isoforms Have Different Functions in Matrix Rearrangement by MDA-MB-231 Cells. PLoS ONE 2015, 10, e0131920. [Google Scholar] [CrossRef]
- Gao, Y.; Khan, G.J.; Wei, X.; Zhai, K.F.; Sun, L.; Yuan, S. DT-13 inhibits breast cancer cell migration via non-muscle myosin II-A regulation in tumor microenvironment synchronized adaptations. Clin. Transl. Oncol. 2020, 22, 1591–1602. [Google Scholar] [CrossRef]
- Xu, Z.; Li, P.; Wei, D.; Wang, Z.; Bao, Y.; Sun, J.; Qu, L.; Wang, L. NMMHC-IIA-dependent nuclear location of CXCR4 promotes migration and invasion in renal cell carcinoma. Oncol. Rep. 2016, 36, 2681–2688. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA A Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
- Gao, S.; Wang, S.; Zhao, Z.; Zhang, C.; Liu, Z.; Ye, P.; Xu, Z.; Yi, B.; Jiao, K.; Naik, G.A.; et al. TUBB4A interacts with MYH9 to protect the nucleus during cell migration and promotes prostate cancer via GSK3β/β-catenin signalling. Nat. Commun. 2022, 13, 2792. [Google Scholar] [CrossRef]
- Zhang, H.; Liu, S.; Tang, L.; Ge, J.; Lu, X. Long non-coding RNA (LncRNA) MRPL23-AS1 promotes tumor progression and carcinogenesis in osteosarcoma by activating Wnt/β-catenin signaling via inhibiting microRNA miR-30b and upregulating myosin heavy chain 9 (MYH9). Bioengineered 2021, 12, 162–171. [Google Scholar] [CrossRef] [PubMed]
- Yu, S.-T.; Sun, B.-H.; Ge, J.-N.; Shi, J.-L.; Zhu, M.-S.; Wei, Z.-G.; Li, T.-T.; Zhang, Z.-C.; Chen, W.-S.; Lei, S.-T. CRLF1–MYH9 Interaction Regulates Proliferation and Metastasis of Papillary Thyroid Carcinoma Through the ERK/ETV4 Axis. Front. Endocrinol. 2020, 11, 535. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; He, H.; Li, W.; Phay, J.; Shen, R.; Yu, L.; Hancioglu, B.; de La Chapelle, A. MYH9 binds to lncRNA gene PTCSC2 and regulates FOXE1 in the 9q22 thyroid cancer risk locus. Proc. Natl. Acad. Sci. USA 2017, 114, 474–479. [Google Scholar] [CrossRef]
- Chang, F.; Kong, S.J.; Wang, L.; Choi, B.K.; Lee, H.; Kim, C.; Kim, J.M.; Park, K. Targeting Actomyosin Contractility Suppresses Malignant Phenotypes of Acute Myeloid Leukemia Cells. Int. J. Mol. Sci. 2020, 21, 3460. [Google Scholar] [CrossRef]
- Minegishi, M.; Tachibana, K.; Sato, T.; Iwata, S.; Nojima, Y.; Morimoto, C. Structure and function of Cas-L, a 105-kD Crk-associated substrate-related protein that is involved in beta 1 integrin-mediated signaling in lymphocytes. J. Exp. Med. 1996, 184, 1365–1375. [Google Scholar] [CrossRef]
- Shagisultanova, E.; Gaponova, A.V.; Gabbasov, R.; Nicolas, E.; Golemis, E.A. Preclinical and clinical studies of the NEDD9 scaffold protein in cancer and other diseases. Gene 2015, 567, 1–11. [Google Scholar] [CrossRef]
- Manié, S.N.; Beck, A.R.P.; Astier, A.; Law, S.F.; Canty, T.; Hirai, H.; Druker, B.J.; Avraham, H.; Haghayeghi, N.; Sattler, M.; et al. Involvement of p130Cas and p105HEF1, a Novel Cas-like Docking Protein, in a Cytoskeleton-dependent Signaling Pathway Initiated by Ligation of Integrin or Antigen Receptor on Human B Cells. J. Biol. Chem. 1997, 272, 4230–4236. [Google Scholar] [CrossRef]
- Browne, C.D.; Hoefer, M.M.; Chintalapati, S.K.; Cato, M.H.; Wallez, Y.; Ostertag, D.V.; Pasquale, E.B.; Rickert, R.C. SHEP1 partners with CasL to promote marginal zone B-cell maturation. Proc. Natl. Acad. Sci. USA 2010, 107, 18944–18949. [Google Scholar] [CrossRef] [PubMed]
- Semelakova, M.; Grauzam, S.; Betadthunga, P.; Tiedeken, J.; Coaxum, S.; Neskey, D.M.; Rosenzweig, S.A. Vimentin and Non-Muscle Myosin IIA are Members of the Neural Precursor Cell Expressed Developmentally Down-Regulated 9 (NEDD9) Interactome in Head and Neck Squamous Cell Carcinoma Cells. Transl. Oncol. 2019, 12, 49–61. [Google Scholar] [CrossRef]
- Que, T.; Zheng, H.; Zeng, Y.; Liu, X.; Qi, G.; La, Q.; Liang, T.; Li, Z.; Yi, G.; Zhang, S.; et al. HMGA1 stimulates MYH9-dependent ubiquitination of GSK-3β via PI3K/Akt/c-Jun signaling to promote malignant progression and chemoresistance in gliomas. Cell Death Dis. 2021, 12, 1147. [Google Scholar] [CrossRef]
- Picariello, H.S.; Kenchappa, R.S.; Rai, V.; Crish, J.F.; Dovas, A.; Pogoda, K.; McMahon, M.; Bell, E.S.; Chandrasekharan, U.; Luu, A.; et al. Myosin IIA suppresses glioblastoma development in a mechanically sensitive manner. Proc. Natl. Acad. Sci. USA 2019, 116, 15550–15559. [Google Scholar] [CrossRef] [PubMed]
- Kwon, H.; Yun, M.; Kwon, T.-H.; Bang, M.; Lee, J.; Lee, Y.S.; Ko, H.Y.; Chong, K. Fibronectin Type III Domain Containing 3B as a Potential Prognostic and Therapeutic Biomarker for Glioblastoma. Biomedicines 2023, 11, 3168. [Google Scholar] [CrossRef]
- Li, Y.Q.; Chen, Y.; Xu, Y.F.; He, Q.M.; Yang, X.J.; Li, Y.Q.; Hong, X.H.; Huang, S.Y.; Tang, L.L.; Liu, N. FNDC3B 3′-UTR shortening escapes from microRNA-mediated gene repression and promotes nasopharyngeal carcinoma progression. Cancer Sci. 2020, 111, 1991–2003. [Google Scholar] [CrossRef]
- Suda, K.; Onozato, R.; Yatabe, Y.; Mitsudomi, T. EGFR T790M Mutation: A Double Role in Lung Cancer Cell Survival? J. Thorac. Oncol. 2009, 4, 1–4. [Google Scholar] [CrossRef]
- Zhao, R.; Sun, L.; Lin, S.; Bai, X.; Yu, B.; Yuan, S.; Zhang, L. The saponin monomer of dwarf lilyturf tuber, DT-13, inhibits angiogenesis under hypoxia and normoxia via multi-targeting activity. Oncol. Rep. 2013, 29, 1379–1386. [Google Scholar] [CrossRef]
- Wei, X.-H.; Lin, S.-S.; Liu, Y.; Zhao, R.-P.; Khan, G.J.; Du, H.-Z.; Mao, T.-T.; Yu, B.-Y.; Li, R.-M.; Yuan, S.-T.; et al. DT-13 attenuates human lung cancer metastasis via regulating NMIIA activity under hypoxia condition. Oncol. Rep. 2016, 36, 991–999. [Google Scholar] [CrossRef]
- Ping, Q.; Yan, R.; Cheng, X.; Wang, W.; Zhong, Y.; Hou, Z.; Shi, Y.; Wang, C.; Li, R. Cancer-associated fibroblasts: Overview, progress, challenges, and directions. Cancer Gene Ther. 2021, 28, 984–999. [Google Scholar] [CrossRef]
- Du, H.; Huang, Y.; Hou, X.; Yu, X.; Lin, S.; Wei, X.; Li, R.; Khan, G.J.; Yuan, S.; Sun, L. DT-13 inhibits cancer cell migration by regulating NMIIA indirectly in the tumor microenvironment. Oncol. Rep. 2016, 36, 721–728. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Sun, B.; Xu, A.; Tang, J.; Zhang, H.; Gao, J.; Wang, L. MICAL2 implies immunosuppressive features and acts as an independent and adverse prognostic biomarker in pancreatic cancer. Sci. Rep. 2024, 14, 3177. [Google Scholar] [CrossRef]
- Ioannou, G.N.; Splan, M.F.; Weiss, N.S.; McDonald, G.B.; Beretta, L.; Lee, S.P. Incidence and Predictors of Hepatocellular Carcinoma in Patients With Cirrhosis. Clin. Gastroenterol. Hepatol. 2007, 5, 938–945.e934. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q.; Lu, Y.; Lin, J.; Gu, Y. ENKUR acts as a tumor suppressor in lung adenocarcinoma cells through PI3K/Akt and MAPK/ERK signaling pathways. J. Cancer 2019, 10, 3975–3984. [Google Scholar] [CrossRef]
- Ma, Q.; Lu, Y.; Gu, Y. ENKUR Is Involved in the Regulation of Cellular Biology in Colorectal Cancer Cells via PI3K/Akt Signaling Pathway. Technol. Cancer Res. Treat. 2019, 18, 153303381984143. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.H.; Yang, H.L.; Deng, S.T.; Hu, Z.; Chen, W.F.; Yan, W.W.; Hou, R.T.; Li, Y.H.; Xian, R.T.; Xie, Y.Y.; et al. The small molecule chemical compound cinobufotalin attenuates resistance to DDP by inducing ENKUR expression to suppress MYH9-mediated c-Myc deubiquitination in lung adenocarcinoma. Acta Pharmacol. Sin. 2022, 43, 2687–2695. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Yu, Y.; Sun, S.; Wang, Z.; Liu, P.; Liu, S.; Jiang, J. Bradykinin promotes migration and invasion of hepatocellular carcinoma cells through TRPM7 and MMP2. Exp. Cell Res. 2016, 349, 68–76. [Google Scholar] [CrossRef] [PubMed]
- Clark, K.; Langeslag, M.; Van Leeuwen, B.; Ran, L.; Ryazanov, A.G.; Figdor, C.G.; Moolenaar, W.H.; Jalink, K.; Van Leeuwen, F.N. TRPM7, a novel regulator of actomyosin contractility and cell adhesion. EMBO J. 2006, 25, 290–301. [Google Scholar] [CrossRef]
- Zhao, R.; Ge, Y.; Gong, Y.; Li, B.; Xiao, B.; Zuo, S. NAP1L5 targeting combined with MYH9 Inhibit HCC progression through PI3K/AKT/mTOR signaling pathway. Aging 2022, 14, 9000–9019. [Google Scholar] [CrossRef]
- Calimano-Ramirez, L.F.; Daoud, T.; Gopireddy, D.R.; Morani, A.C.; Waters, R.; Gumus, K.; Klekers, A.R.; Bhosale, P.R.; Virarkar, M.K. Pancreatic acinar cell carcinoma: A comprehensive review. World J. Gastroenterol. 2022, 28, 5827–5844. [Google Scholar] [CrossRef]
- Balouchi-Anaraki, S.; Ahmadvand, S.; Safaei, A.; Ghaderi, A. 4H12, a Murine Monoclonal Antibody Directed against Myosin Heavy Chain-9 Expressed on Acinar Cell Carcinoma of Pancreas with Potential Therapeutic Application. Iran. Biomed. J. 2021, 25, 310–322. [Google Scholar] [CrossRef]
- Liu, J.; Liu, Z.; Yan, W.; Yang, H.; Fang, S.; Deng, S.; Wen, Y.; Shen, P.; Li, Y.; Hou, R.; et al. ENKUR recruits FBXW7 to ubiquitinate and degrade MYH9 and further suppress MYH9-induced deubiquitination of β-catenin to block gastric cancer metastasis. MedComm 2022, 3, e185. [Google Scholar] [CrossRef]
- Liang, S.; He, L.; Zhao, X.; Miao, Y.; Gu, Y.; Guo, C.; Xue, Z.; Dou, W.; Hu, F.; Wu, K.; et al. MicroRNA Let-7f Inhibits Tumor Invasion and Metastasis by Targeting MYH9 in Human Gastric Cancer. PLoS ONE 2011, 6, e18409. [Google Scholar] [CrossRef]
- Liu, T.; Liu, Y.; Wei, C.; Yang, Z.; Chang, W.; Zhang, X. LncRNA HULC promotes the progression of gastric cancer by regulating miR-9-5p/MYH9 axis. Biomed. Pharmacother. 2020, 121, 109607. [Google Scholar] [CrossRef] [PubMed]
- Fang, X.; Bai, Y.; Zhang, L.; Ding, S. Silencing circSLAMF6 represses cell glycolysis, migration, and invasion by regulating the miR-204-5p/MYH9 axis in gastric cancer under hypoxia. Biosci. Rep. 2020, 40, BSR20201275. [Google Scholar] [CrossRef] [PubMed]
- Ye, G.; Huang, K.; Yu, J.; Zhao, L.; Zhu, X.; Yang, Q.; Li, W.; Jiang, Y.; Zhuang, B.; Liu, H.; et al. MicroRNA-647 Targets SRF-MYH9 Axis to Suppress Invasion and Metastasis of Gastric Cancer. Theranostics 2017, 7, 3338–3353. [Google Scholar] [CrossRef]
- Li, F.; Shi, J.; Xu, Z.; Yao, X.; Mou, T.; Yu, J.; Liu, H.; Li, G. S100A4-MYH9 Axis Promote Migration and Invasion of Gastric Cancer Cells by Inducing TGF-β-Mediated Epithelial-Mesenchymal Transition. J. Cancer 2018, 9, 3839–3849. [Google Scholar] [CrossRef]
- Wang, Z.; Zhu, Z.; Li, C.; Zhang, Y.; Li, Z.; Sun, S. NMIIA promotes tumorigenesis and prevents chemosensitivity in colorectal cancer by activating AMPK/mTOR pathway. Exp. Cell Res. 2021, 398, 112387. [Google Scholar] [CrossRef] [PubMed]
- Cao, M.; Wang, Y.; Xiao, Y.; Zheng, D.; Zhi, C.; Xia, X.; Yuan, X. Activation of the clock gene TIMELESS by H3k27 acetylation promotes colorectal cancer tumorigenesis by binding to Myosin-9. J. Exp. Clin. Cancer Res. 2021, 40, 162. [Google Scholar] [CrossRef]
- Zhou, Z.; Wu, L.; Liu, Z.; Zhang, X.; Han, S.; Zhao, N.; Bao, H.; Yuan, W.; Chen, J.; Ji, J.; et al. MicroRNA-214-3p targets the PLAGL2-MYH9 axis to suppress tumor proliferation and metastasis in human colorectal cancer. Aging 2020, 12, 9633–9657. [Google Scholar] [CrossRef]
- Liu, X.; Liu, Y.; Liu, Z.; Lin, C.; Meng, F.; Xu, L.; Zhang, X.; Zhang, C.; Zhang, P.; Gong, S.; et al. CircMYH9 drives colorectal cancer growth by regulating serine metabolism and redox homeostasis in a p53-dependent manner. Mol. Cancer 2021, 20, 114. [Google Scholar] [CrossRef]
- Wang, G.; Zhang, S.; Fernig, D.G.; Martin-Fernandez, M.; Rudland, P.S.; Barraclough, R. Mutually antagonistic actions of S100A4 and S100A1 on normal and metastatic phenotypes. Oncogene 2005, 24, 1445–1454. [Google Scholar] [CrossRef]
- Li, Z.H.; Spektor, A.; Varlamova, O.; Bresnick, A.R. Mts1 regulates the assembly of nonmuscle myosin-IIA. Biochemistry 2003, 42, 14258–14266. [Google Scholar] [CrossRef]
- Parkkila, S.; Pan, P.-W.; Ward, A.; Gibadulinova, A.; Oveckova, I.; Pastorekova, S.; Pastorek, J.; Martinez, A.R.; Helin, H.O.; Isola, J. The calcium-binding protein S100P in normal and malignant human tissues. BMC Clin. Pathol. 2008, 8, 2. [Google Scholar] [CrossRef]
- Wang, G.; Zhang, S.; Fernig, D.G.; Spiller, D.; Martin-Fernandez, M.; Zhang, H.; Ding, Y.; Rao, Z.; Rudland, P.S.; Barraclough, R. Heterodimeric interaction and interfaces of S100A1 and S100P. Biochem. J. 2004, 382, 375–383. [Google Scholar] [CrossRef] [PubMed]
- Du, M.; Wang, G.; Barsukov, I.L.; Gross, S.R.; Smith, R.; Rudland, P.S. Direct interaction of metastasis-inducing S100P protein with tubulin causes enhanced cell migration without changes in cell adhesion. Biochem. J. 2020, 477, 1159–1178. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Wang, Z.; Su, P.; Liang, Y.; Li, Z.; Zhang, H.; Song, X.; Han, D.; Wang, X.; Liu, Y.; et al. circ-EIF6 encodes EIF6-224aa to promote TNBC progression via stabilizing MYH9 and activating the Wnt/beta-catenin pathway. Mol. Ther. 2022, 30, 415–430. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; Huang, W.; Li, W.; Chen, S.; Chen, W.; Zhou, Y.; Peng, P.; Gu, W. TFPI-2 suppresses breast cancer cell proliferation and invasion through regulation of ERK signaling and interaction with actinin-4 and myosin-9. Sci. Rep. 2018, 8, 14402. [Google Scholar] [CrossRef]
- Derycke, L.; Stove, C.; Vercoutter-Edouart, A.-S.; De Wever, O.; Dollé, L.; Colpaert, N.; Depypere, H.; Michalski, J.-C.; Bracke, M. The role of non-muscle myosin IIA in aggregation and invasion of human MCF-7 breast cancer cells. Int. J. Dev. Biol. 2011, 55, 835–840. [Google Scholar] [CrossRef]
- Cruz-Ramos, E.; Macias-Silva, M.; Sandoval-Hernandez, A.; Tecalco-Cruz, A.C. Non-muscle myosin IIA is post-translationally modified by interferon-stimulated gene 15 in breast cancer cells. Int. J. Biochem. Cell Biol. 2019, 107, 14–26. [Google Scholar] [CrossRef]
- Yoneda, A.; Minomi, K.; Tamura, Y. HSP47 promotes metastasis of breast cancer by interacting with myosin IIA via the unfolded protein response transducer IRE1α. Oncogene 2020, 39, 4519–4537. [Google Scholar] [CrossRef]
- Zhang, L.; Zhou, X.; Liu, B.; Shi, X.; Li, X.; Xu, F.; Fu, X.; Wang, X.; Ye, K.; Jin, T.; et al. HBXIP blocks myosin-IIA assembly by phosphorylating and interacting with NMHC-IIA in breast cancer metastasis. Acta Pharm. Sin. B 2023, 13, 1053–1070. [Google Scholar] [CrossRef]
- Alanazi, S.M.; Feroz, W.; Mishra, R.; Kilroy, M.K.; Patel, H.; Yuan, L.; Storr, S.J.; Garrett, J.T. HER2 inhibition increases non-muscle myosin IIA to promote tumorigenesis in HER2+ breast cancers. PLoS ONE 2023, 18, e0285251. [Google Scholar] [CrossRef]
- Xu, Z.; Liu, M.; Wang, J.; Liu, K.; Xu, L.; Fan, D.; Zhang, H.; Hu, W.; Wei, D.; Wang, J. Single-cell RNA-sequencing analysis reveals MYH9 promotes renal cell carcinoma development and sunitinib resistance via AKT signaling pathway. Cell Death Discov. 2022, 8, 125. [Google Scholar] [CrossRef]
- Chen, F.; Yin, S.; Feng, Z.; Liu, C.; Lv, J.; Chen, Y.; Shen, R.; Wang, J.; Deng, Z. Knockdown of circ_NEK6 Decreased (131)I Resistance of Differentiated Thyroid Carcinoma via Regulating miR-370-3p/MYH9 Axis. Technol. Cancer Res. Treat. 2021, 20, 15330338211004950. [Google Scholar] [CrossRef]
- Hu, S.; Ren, S.; Cai, Y.; Liu, J.; Han, Y.; Zhao, Y.; Yang, J.; Zhou, X.; Wang, X. Glycoprotein PTGDS promotes tumorigenesis of diffuse large B-cell lymphoma by MYH9-mediated regulation of Wnt–β-catenin–STAT3 signaling. Cell Death Differ. 2022, 29, 642–656. [Google Scholar] [CrossRef] [PubMed]
- Nguyen-Ngoc, K.-V.; Silvestri, V.L.; Georgess, D.; Fairchild, A.N.; Ewald, A.J. Mosaic loss of non-muscle myosin IIA and IIB is sufficient to induce mammary epithelial proliferation. J. Cell Sci. 2017, 130, 3213–3221. [Google Scholar] [CrossRef]
- Newell-Litwa, K.A.; Horwitz, R.; Lamers, M.L. Non-muscle myosin II in disease: Mechanisms and therapeutic opportunities. Dis. Models Mech. 2015, 8, 1495–1515. [Google Scholar] [CrossRef]
- Kunishima, S.; Matsushita, T.; Kojima, T.; Amemiya, N.; Choi, Y.M.; Hosaka, N.; Inoue, M.; Jung, Y.; Mamiya, S.; Matsumoto, K.; et al. Identification of six novel MYH9 mutations and genotype–Phenotype relationships in autosomal dominant macrothrombocytopenia with leukocyte inclusions. J. Hum. Genet. 2001, 46, 722–729. [Google Scholar] [CrossRef] [PubMed]
- Syndrome Consortium, T.M.-H.F. Mutations in MYH9 result in the May-Hegglin anomaly, and Fechtner and Sebastian syndromes. Nat. Genet. 2000, 26, 103–105. [Google Scholar] [CrossRef]
- Lalwani, A.K.; Goldstein, J.A.; Kelley, M.J.; Luxford, W.; Castelein, C.M.; Mhatre, A.N. Human Nonsyndromic Hereditary Deafness DFNA17 Is Due to a Mutation in Nonmuscle Myosin MYH9. Am. J. Hum. Genet. 2000, 67, 1121–1128. [Google Scholar] [CrossRef] [PubMed]
- Kelley, M.J.; Jawien, W.; Ortel, T.L.; Korczak, J.F. Mutation of MYH9, encoding non-muscle myosin heavy chain A, in May-Hegglin anomaly. Nat. Genet. 2000, 26, 106–108. [Google Scholar] [CrossRef]
- Althaus, K.; Greinacher, A. MYH9-Related Platelet Disorders. Semin. Thromb. Hemost. 2009, 35, 189–203. [Google Scholar] [CrossRef]
- Asensio-Juarez, G.; Llorente-Gonzalez, C.; Vicente-Manzanares, M. Linking the Landscape of MYH9-Related Diseases to the Molecular Mechanisms that Control Non-Muscle Myosin II-A Function in Cells. Cells 2020, 9, 1458. [Google Scholar] [CrossRef] [PubMed]
- Pecci, A.; Panza, E.; Pujol-Moix, N.; Klersy, C.; Di Bari, F.; Bozzi, V.; Gresele, P.; Lethagen, S.; Fabris, F.; Dufour, C.; et al. Position of nonmuscle myosin heavy chain IIA (NMMHC-IIA) mutations predicts the natural history ofMYH9-related disease. Hum. Mutat. 2008, 29, 409–417. [Google Scholar] [CrossRef] [PubMed]
- Kunishima, S. May-Hegglin anomaly--from genome research to clinical laboratory. Rinsho Byori 2003, 51, 898–904. [Google Scholar] [PubMed]
- Kunishima, S.; Matsushita, T.; Kojima, T.; Sako, M.; Kimura, F.; Jo, E.-K.; Inoue, C.; Kamiya, T.; Saito, H. Immunofluorescence Analysis of Neutrophil Nonmuscle Myosin Heavy Chain-A in MYH9 Disorders: Association of Subcellular Localization with MYH9 Mutations. Lab. Investig. 2003, 83, 115–122. [Google Scholar] [CrossRef]
- Saposnik, B.; Binard, S.; Fenneteau, O.; Nurden, A.; Nurden, P.; Hurtaud-Roux, M.F.; Schlegel, N. Mutation spectrum and genotype-phenotype correlations in a large French cohort of MYH 9-Related Disorders. Mol. Genet. Amp; Genom. Med. 2014, 2, 297–312. [Google Scholar] [CrossRef]
- Pecci, A.; Verver, E.J.; Schlegel, N.; Canzi, P.; Boccio, C.M.; Platokouki, H.; Krause, E.; Benazzo, M.; Topsakal, V.; Greinacher, A. Cochlear implantation is safe and effective in patients with MYH9-related disease. Orphanet J. Rare Dis. 2014, 9, 100. [Google Scholar] [CrossRef]
- Pal, K.; Nowak, R.; Billington, N.; Liu, R.; Ghosh, A.; Sellers, J.R.; Fowler, V.M. Megakaryocyte migration defects due to nonmuscle myosin IIA mutations underlie thrombocytopenia in MYH9-related disease. Blood 2020, 135, 1887–1898. [Google Scholar] [CrossRef]
- Pecci, A.; Panza, E.; De Rocco, D.; Pujol-Moix, N.; Girotto, G.; Podda, L.; Paparo, C.; Bozzi, V.; Pastore, A.; Balduini, C.L.; et al. MYH9 related disease: Four novel mutations of the tail domain of myosin-9 correlating with a mild clinical phenotype. Eur. J. Haematol. 2010, 84, 291–297. [Google Scholar] [CrossRef]
- Malara, A.; Badalucco, S.; Bozzi, V.; Torti, M.; Balduini, C.L.; Balduini, A.; Pecci, A. Megakaryocytes of patients with MYH9-related thrombocytopenia present an altered proplatelet formation. Thromb. Haemost. 2009, 102, 90–96. [Google Scholar] [CrossRef]
- Heynen, M.J.; Blockmans, D.; Verwilghen, R.L.; Vermylen, J. Congenital macrothrombocytopenia, leucocyte inclusions, deafness and proteinuria: Functional and electron microscopic observations on platelets and megakaryocytes. Br. J. Haematol. 1988, 70, 441–448. [Google Scholar] [CrossRef]
- Chen, Z.; Naveiras, O.; Balduini, A.; Mammoto, A.; Conti, M.A.; Adelstein, R.S.; Ingber, D.; Daley, G.Q.; Shivdasani, R.A. The May-Hegglin anomaly gene MYH9 is a negative regulator of platelet biogenesis modulated by the Rho-ROCK pathway. Blood 2007, 110, 171–179. [Google Scholar] [CrossRef]
- Canobbio, I.; Noris, P.; Pecci, A.; Balduini, A.; Balduini, C.L.; Torti, M. Altered cytoskeleton organization in platelets from patients with MYH9-related disease. J. Thromb. Haemost. 2005, 3, 1026–1035. [Google Scholar] [CrossRef] [PubMed]
- Di Pumpo, M.; Noris, P.; Pecci, A.; Savoia, A.; Seri, M.; Ceresa, I.F.; Balduini, C.L. Defective expression of GPIb/IX/V complex in platelets from patients with May-Hegglin anomaly and Sebastian syndrome. Haematologica 2002, 87, 943–947. [Google Scholar]
- Heath, K.E.; Campos-Barros, A.; Toren, A.; Rozenfeld-Granot, G.; Carlsson, L.E.; Savige, J.; Denison, J.C.; Gregory, M.C.; White, J.G.; Barker, D.F.; et al. Nonmuscle Myosin Heavy Chain IIA Mutations Define a Spectrum of Autosomal Dominant Macrothrombocytopenias: May-Hegglin Anomaly and Fechtner, Sebastian, Epstein, and Alport-Like Syndromes. Am. J. Hum. Genet. 2001, 69, 1033–1045. [Google Scholar] [CrossRef]
- Seri, M.; Pecci, A.; Di Bari, F.; Cusano, R.; Savino, M.; Panza, E.; Nigro, A.; Noris, P.; Gangarossa, S.; Rocca, B.; et al. MYH9-related disease: May-Hegglin anomaly, Sebastian syndrome, Fechtner syndrome, and Epstein syndrome are not distinct entities but represent a variable expression of a single illness. Medicine 2003, 82, 203–215. [Google Scholar] [CrossRef]
- Galeano, D.; Zanoli, L.; L’Imperio, V.; Fatuzzo, P.; Granata, A. Renal diseases related to MYH9 disorders. G Ital. Nefrol. 2017, 34, 40–57. [Google Scholar] [PubMed]
- Furlano, M.; Arlandis, R.; Venegas, M.D.P.; Novelli, S.; Crespi, J.; Bullich, G.; Ayasreh, N.; Remacha, A.; Ruiz, P.; Lorente, L.; et al. MYH9 Associated nephropathy. Nefrología 2019, 39, 133–140. [Google Scholar] [CrossRef] [PubMed]
- Pecci, A.; Biino, G.; Fierro, T.; Bozzi, V.; Mezzasoma, A.; Noris, P.; Ramenghi, U.; Loffredo, G.; Fabris, F.; Momi, S.; et al. Alteration of Liver Enzymes Is a Feature of the Myh9-Related Disease Syndrome. PLoS ONE 2012, 7, e35986. [Google Scholar] [CrossRef]
- Li, J.; Cai, T.; Jiang, Y.; Chen, H.; He, X.; Chen, C.; Li, X.; Shao, Q.; Ran, X.; Li, Z.; et al. Genes with de novo mutations are shared by four neuropsychiatric disorders discovered from NPdenovo database. Mol. Psychiatry 2016, 21, 290–297. [Google Scholar] [CrossRef]
- Weber, M.; Kim, S.; Patterson, N.; Rooney, K.; Searles, C.D. MiRNA-155 targets myosin light chain kinase and modulates actin cytoskeleton organization in endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H1192–H1203. [Google Scholar] [CrossRef]
- Moore, C.S.; Rao, V.T.S.; Durafourt, B.A.; Bedell, B.J.; Ludwin, S.K.; Bar-Or, A.; Antel, J.P. miR-155 as a multiple sclerosis–relevant regulator of myeloid cell polarization. Ann. Neurol. 2013, 74, 709–720. [Google Scholar] [CrossRef]
- Parisi, C.; Arisi, I.; D’Ambrosi, N.; Storti, A.E.; Brandi, R.; D’Onofrio, M.; Volonté, C. Dysregulated microRNAs in amyotrophic lateral sclerosis microglia modulate genes linked to neuroinflammation. Cell Death Dis. 2013, 4, e959. [Google Scholar] [CrossRef]
- Tan, L.; Yuan, X.; Liu, Y.; Cai, X.; Guo, S.; Wang, A. Non-muscle Myosin II: Role in Microbial Infection and Its Potential as a Therapeutic Target. Front. Microbiol. 2019, 10, 401. [Google Scholar] [CrossRef]
- Almeida, M.T.; Mesquita, F.S.; Cruz, R.; Osório, H.; Custódio, R.; Brito, C.; Vingadassalom, D.; Martins, M.; Leong, J.M.; Holden, D.W.; et al. Src-dependent Tyrosine Phosphorylation of Non-muscle Myosin Heavy Chain-IIA Restricts Listeria monocytogenes Cellular Infection. J. Biol. Chem. 2015, 290, 8383–8395. [Google Scholar] [CrossRef] [PubMed]
- Veettil, M.V.; Sadagopan, S.; Kerur, N.; Chakraborty, S.; Chandran, B. Interaction of c-Cbl with Myosin IIA Regulates Bleb Associated Macropinocytosis of Kaposi’s Sarcoma-Associated Herpesvirus. PLoS Pathog. 2010, 6, e1001238. [Google Scholar] [CrossRef] [PubMed]
- Mesquita, F.S.; Brito, C.; Cabanes, D.; Sousa, S. Control of cytoskeletal dynamics during cellular responses to pore forming toxins. Commun. Integr. Biol. 2017, 10, e1349582. [Google Scholar] [CrossRef]
- Mesquita, F.S.; Brito, C.; Moya, M.J.M.; Pinheiro, J.C.; Mostowy, S.; Cabanes, D.; Sousa, S. Endoplasmic reticulum chaperone Gp96 controls actomyosin dynamics and protects against pore-forming toxins. EMBO Rep. 2017, 18, 303–318. [Google Scholar] [CrossRef] [PubMed]
- Allingham, J.S.; Smith, R.; Rayment, I. The structural basis of blebbistatin inhibition and specificity for myosin II. Nat. Struct. Amp; Mol. Biol. 2005, 12, 378–379. [Google Scholar] [CrossRef]
- Limouze, J.; Straight, A.F.; Mitchison, T.; Sellers, J.R. Specificity of blebbistatin, an inhibitor of myosin II. J. Muscle Res. Cell Motil. 2004, 25, 337–341. [Google Scholar] [CrossRef]
- Rauscher, A.A.; Gyimesi, M.; Kovacs, M.; Malnasi-Csizmadia, A. Targeting Myosin by Blebbistatin Derivatives: Optimization and Pharmacological Potential. Trends Biochem. Sci. 2018, 43, 700–713. [Google Scholar] [CrossRef]
- Robinson, T.; Pai, M.; Liu, J.; Vizeacoumar, F.; Sun, T.; Egan, S.; Datti, A.; Huang, J.; Zacksenhaus, E. High-throughput screen identifies disulfiram as a potential therapeutic for triple-negative breast cancer cells: Interaction with IQ motif-containing factors. Cell Cycle 2013, 12, 3013–3024. [Google Scholar] [CrossRef]
- Zhou, W.; Huo, J.; Yang, Y.; Zhang, X.; Li, S.; Zhao, C.; Ma, H.; Liu, Y.; Liu, J.; Li, J.; et al. Aminated Fullerene Abrogates Cancer Cell Migration by Directly Targeting Myosin Heavy Chain 9. ACS Appl. Mater. Interfaces 2020, 12, 56862–56873. [Google Scholar] [CrossRef] [PubMed]
- Qian, Y.; Zhao, M.; Han, Q.; Wang, J.; Liao, L.; Yang, H.; Liu, D.; Tu, P.; Liang, H.; Zeng, K. Pharmacologically targeting molecular motor promotes mitochondrial fission for anti-cancer. Acta Pharm. Sin. B 2021, 11, 1853–1866. [Google Scholar] [CrossRef]
- Zhang, T.; Shen, S.; Zhu, Z.; Lu, S.; Yin, X.; Zheng, J.; Jin, J. Homoharringtonine binds to and increases myosin-9 in myeloid leukaemia. Br. J. Pharmacol. 2016, 173, 212–221. [Google Scholar] [CrossRef]
- Feng, Y.; LoGrasso, P.V.; Defert, O.; Li, R. Rho Kinase (ROCK) Inhibitors and Their Therapeutic Potential. J. Med. Chem. 2016, 59, 2269–2300. [Google Scholar] [CrossRef]
- Garnock-Jones, K.P. Ripasudil: First Global Approval. Drugs 2014, 74, 2211–2215. [Google Scholar] [CrossRef]
- Mehran, N.A.; Sinha, S.; Razeghinejad, R. New glaucoma medications: Latanoprostene bunod, netarsudil, and fixed combination netarsudil-latanoprost. Eye 2020, 34, 72–88. [Google Scholar] [CrossRef] [PubMed]
- Cutler, C.; Lee, S.J.; Arai, S.; Rotta, M.; Zoghi, B.; Lazaryan, A.; Ramakrishnan, A.; Defilipp, Z.; Salhotra, A.; Chai-Ho, W.; et al. Belumosudil for chronic graft-versus-host disease after 2 or more prior lines of therapy: The ROCKstar Study. Blood 2021, 138, 2278–2289. [Google Scholar] [CrossRef]
- Jagasia, M.; Lazaryan, A.; Bachier, C.R.; Salhotra, A.; Weisdorf, D.J.; Zoghi, B.; Essell, J.; Green, L.; Schueller, O.; Patel, J.; et al. ROCK2 Inhibition With Belumosudil (KD025) for the Treatment of Chronic Graft-Versus-Host Disease. J. Clin. Oncol. 2021, 39, 1888–1898. [Google Scholar] [CrossRef]
- Anastassiadis, T.; Deacon, S.W.; Devarajan, K.; Ma, H.; Peterson, J.R. Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat. Biotechnol. 2011, 29, 1039–1045. [Google Scholar] [CrossRef] [PubMed]
- Smit, M.A.; Maddalo, G.; Greig, K.; Raaijmakers, L.M.; Possik, P.A.; Van Breukelen, B.; Cappadona, S.; Heck, A., Jr.; Altelaar, A.M.; Peeper, D.S. ROCK 1 is a potential combinatorial drug target for BRAF mutant melanoma. Mol. Syst. Biol. 2014, 10, 772. [Google Scholar] [CrossRef] [PubMed]
- McLeod, R.; Kumar, R.; Papadatos-Pastos, D.; Mateo, J.; Brown, J.S.; Garces, A.H.I.; Ruddle, R.; Decordova, S.; Jueliger, S.; Ferraldeschi, R.; et al. First-in-Human Study of AT13148, a Dual ROCK-AKT Inhibitor in Patients with Solid Tumors. Clin. Cancer Res. 2020, 26, 4777–4784. [Google Scholar] [CrossRef]
- Unbekandt, M.; Croft, D.R.; Crighton, D.; Mezna, M.; McArthur, D.; McConnell, P.; Schüttelkopf, A.W.; Belshaw, S.; Pannifer, A.; Sime, M.; et al. A novel small-molecule MRCK inhibitor blocks cancer cell invasion. Cell Commun. Signal. 2014, 12, 54. [Google Scholar] [CrossRef] [PubMed]
- Castoreno, A.B.; Smurnyy, Y.; Torres, A.D.; Vokes, M.S.; Jones, T.R.; Carpenter, A.E.; Eggert, U.S. Small molecules discovered in a pathway screen target the Rho pathway in cytokinesis. Nat. Chem. Biol. 2010, 6, 457–463. [Google Scholar] [CrossRef] [PubMed]
- Patel, R.A.; Forinash, K.D.; Pireddu, R.; Sun, Y.; Sun, N.; Martin, M.P.; Schönbrunn, E.; Lawrence, N.J.; Sebti, S.M. RKI-1447 Is a Potent Inhibitor of the Rho-Associated ROCK Kinases with Anti-Invasive and Antitumor Activities in Breast Cancer. Cancer Res. 2012, 72, 5025–5034. [Google Scholar] [CrossRef]
- Patel, R.A.; Liu, Y.; Wang, B.; Li, R.; Sebti, S.M. Identification of novel ROCK inhibitors with anti-migratory and anti-invasive activities. Oncogene 2014, 33, 550–555. [Google Scholar] [CrossRef]
- Sun, J.; Zhang, D.; Zheng, Y.; Zhao, Q.; Zheng, M.; Kovacevic, Z.; Richardson, D.R. Targeting the Metastasis Suppressor, NDRG1, Using Novel Iron Chelators: Regulation of Stress Fiber-Mediated Tumor Cell Migration via Modulation of the ROCK1/pMLC2 Signaling Pathway. Mol. Pharmacol. 2013, 83, 454–469. [Google Scholar] [CrossRef]
- Pollock, J.K.; Verma, N.K.; O’Boyle, N.M.; Carr, M.; Meegan, M.J.; Zisterer, D.M. Combretastatin (CA)-4 and its novel analogue CA-432 impair T-cell migration through the Rho/ROCK signalling pathway. Biochem. Pharmacol. 2014, 92, 544–557. [Google Scholar] [CrossRef]
- Hu, Y.; Yang, C.; Amorim, T.; Maqbool, M.; Lin, J.; Li, C.; Fang, C.; Xue, L.; Kwart, A.; Fang, H.; et al. Cisplatin-Mediated Upregulation of APE2 Binding to MYH9 Provokes Mitochondrial Fragmentation and Acute Kidney Injury. Cancer Res. 2021, 81, 713–723. [Google Scholar] [CrossRef]
- Crosas-Molist, E.; Samain, R.; Kohlhammer, L.; Orgaz, J.L.; George, S.L.; Maiques, O.; Barcelo, J.; Sanz-Moreno, V. Rho GTPase signaling in cancer progression and dissemination. Physiol. Rev. 2022, 102, 455–510. [Google Scholar] [CrossRef]
- Rath, N.; Olson, M.F. Rho-associated kinases in tumorigenesis: Re-considering ROCK inhibition for cancer therapy. EMBO Rep. 2012, 13, 900–908. [Google Scholar] [CrossRef] [PubMed]
- Truebestein, L.; Elsner, D.J.; Fuchs, E.; Leonard, T.A. A molecular ruler regulates cytoskeletal remodelling by the Rho kinases. Nat. Commun. 2015, 6, 10029. [Google Scholar] [CrossRef]
- Knipe, R.S.; Tager, A.M.; Liao, J.K. The Rho Kinases: Critical Mediators of Multiple Profibrotic Processes and Rational Targets for New Therapies for Pulmonary Fibrosis. Pharmacol. Rev. 2015, 67, 103–117. [Google Scholar] [CrossRef]
- Barcelo, J.; Samain, R.; Sanz-Moreno, V. Preclinical to clinical utility of ROCK inhibitors in cancer. Trends Cancer 2023, 9, 250–263. [Google Scholar] [CrossRef] [PubMed]
- Pandya, P.; Orgaz, J.L.; Sanz-Moreno, V. Modes of invasion during tumour dissemination. Mol. Oncol. 2017, 11, 5–27. [Google Scholar] [CrossRef]
- Pandya, P.; Orgaz, J.L.; Sanz-Moreno, V. Actomyosin contractility and collective migration: May the force be with you. Curr. Opin. Cell Biol. 2017, 48, 87–96. [Google Scholar] [CrossRef]
- Orgaz, J.L.; Crosas-Molist, E.; Sadok, A.; Perdrix-Rosell, A.; Maiques, O.; Rodriguez-Hernandez, I.; Monger, J.; Mele, S.; Georgouli, M.; Bridgeman, V.; et al. Myosin II Reactivation and Cytoskeletal Remodeling as a Hallmark and a Vulnerability in Melanoma Therapy Resistance. Cancer Cell 2020, 37, 85–103.e109. [Google Scholar] [CrossRef]
- Georgouli, M.; Herraiz, C.; Crosas-Molist, E.; Fanshawe, B.; Maiques, O.; Perdrix, A.; Pandya, P.; Rodriguez-Hernandez, I.; Ilieva, K.M.; Cantelli, G.; et al. Regional Activation of Myosin II in Cancer Cells Drives Tumor Progression via a Secretory Cross-Talk with the Immune Microenvironment. Cell 2019, 176, 757–774.e723. [Google Scholar] [CrossRef] [PubMed]
- Mizuno, K. Signaling mechanisms and functional roles of cofilin phosphorylation and dephosphorylation. Cell Signal 2013, 25, 457–469. [Google Scholar] [CrossRef]
- Amano, M.; Nakayama, M.; Kaibuchi, K. Rho-kinase/ROCK: A key regulator of the cytoskeleton and cell polarity. Cytoskeleton 2010, 67, 545–554. [Google Scholar] [CrossRef]
- Kumper, S.; Mardakheh, F.K.; McCarthy, A.; Yeo, M.; Stamp, G.W.; Paul, A.; Worboys, J.; Sadok, A.; Jorgensen, C.; Guichard, S.; et al. Rho-associated kinase (ROCK) function is essential for cell cycle progression, senescence and tumorigenesis. elife 2016, 5, e12994. [Google Scholar] [CrossRef] [PubMed]
- Mali, R.S.; Ramdas, B.; Ma, P.; Shi, J.; Munugalavadla, V.; Sims, E.; Wei, L.; Vemula, S.; Nabinger, S.C.; Goodwin, C.B.; et al. Rho Kinase Regulates the Survival and Transformation of Cells Bearing Oncogenic Forms of KIT, FLT3, and BCR-ABL. Cancer Cell 2011, 20, 357–369. [Google Scholar] [CrossRef] [PubMed]
- Graziani, V.; Rodriguez-Hernandez, I.; Maiques, O.; Sanz-Moreno, V. The amoeboid state as part of the epithelial-to-mesenchymal transition programme. Trends Cell Biol. 2022, 32, 228–242. [Google Scholar] [CrossRef]
- Orgaz, J.L.; Pandya, P.; Dalmeida, R.; Karagiannis, P.; Sanchez-Laorden, B.; Viros, A.; Albrengues, J.; Nestle, F.O.; Ridley, A.J.; Gaggioli, C.; et al. Diverse matrix metalloproteinase functions regulate cancer amoeboid migration. Nat. Commun. 2014, 5, 4255. [Google Scholar] [CrossRef]
- Samuel, M.S.; Lopez, J.I.; McGhee, E.J.; Croft, D.R.; Strachan, D.; Timpson, P.; Munro, J.; Schröder, E.; Zhou, J.; Brunton, V.G.; et al. Actomyosin-Mediated Cellular Tension Drives Increased Tissue Stiffness and β-Catenin Activation to Induce Epidermal Hyperplasia and Tumor Growth. Cancer Cell 2011, 19, 776–791. [Google Scholar] [CrossRef]
- Gaggioli, C.; Hooper, S.; Hidalgo-Carcedo, C.; Grosse, R.; Marshall, J.F.; Harrington, K.; Sahai, E. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 2007, 9, 1392–1400. [Google Scholar] [CrossRef] [PubMed]
- Wong, P.-P.; Muñoz-Félix, J.M.; Hijazi, M.; Kim, H.; Robinson, S.D.; De Luxán-Delgado, B.; Rodríguez-Hernández, I.; Maiques, O.; Meng, Y.-M.; Meng, Q.; et al. Cancer Burden Is Controlled by Mural Cell-β3-Integrin Regulated Crosstalk with Tumor Cells. Cell 2020, 181, 1346–1363.e1321. [Google Scholar] [CrossRef]
- Xie, Y.; Yue, L.; Shi, Y.; Su, X.; Gan, C.; Liu, H.; Xue, T.; Ye, T. Application and Study of ROCK Inhibitors in Pulmonary Fibrosis: Recent Developments and Future Perspectives. J. Med. Chem. 2023, 66, 4342–4360. [Google Scholar] [CrossRef]
- Chong, C.M.; Ai, N.; Lee, S.M. ROCK in CNS: Different Roles of Isoforms and Therapeutic Target for Neurodegenerative Disorders. Curr. Drug Targets 2017, 18, 455–462. [Google Scholar] [CrossRef]
- Chen, W.; Nyuydzefe, M.S.; Weiss, J.M.; Zhang, J.; Waksal, S.D.; Zanin-Zhorov, A. ROCK2, but not ROCK1 interacts with phosphorylated STAT3 and co-occupies TH17/TFH gene promoters in TH17-activated human T cells. Sci. Rep. 2018, 8, 16636. [Google Scholar] [CrossRef]
- Song, J.; Xi, J.Y.; Yu, W.B.; Yan, C.; Luo, S.S.; Zhou, L.; Zhu, W.H.; Lu, J.H.; Dong, Q.; Xiao, B.G.; et al. Inhibition of ROCK activity regulates the balance of Th1, Th17 and Treg cells in myasthenia gravis. Clin. Immunol. 2019, 203, 142–153. [Google Scholar] [CrossRef]
- Gao, Y.; Yan, Y.; Fang, Q.; Zhang, N.; Kumar, G.; Zhang, J.; Song, L.-J.; Yu, J.; Zhao, L.; Zhang, H.-T.; et al. The Rho kinase inhibitor fasudil attenuates Aβ1–42-induced apoptosis via the ASK1/JNK signal pathway in primary cultures of hippocampal neurons. Metab. Brain Dis. 2019, 34, 1787–1801. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Liu, B. ROCK inhibitor Y-27632 protects rats against cerebral ischemia/reperfusion-induced behavioral deficits and hippocampal damage. Mol. Med. Rep. 2019, 20, 3395–3405. [Google Scholar] [CrossRef] [PubMed]
- Wei, W.; Wang, Y.; Zhang, J.; Gu, Q.; Liu, X.; Song, L.; Chai, Z.; Guo, M.; Yu, J.; Ma, C. Fasudil ameliorates cognitive deficits, oxidative stress and neuronal apoptosis via inhibiting ROCK/MAPK and activating Nrf2 signalling pathways in APP/PS1 mice. Folia Neuropathol. 2021, 59, 32–49. [Google Scholar] [CrossRef]
- Whatcott, C.J.; Ng, S.; Barrett, M.T.; Hostetter, G.; Von Hoff, D.D.; Han, H. Inhibition of ROCK1 kinase modulates both tumor cells and stromal fibroblasts in pancreatic cancer. PLoS ONE 2017, 12, e0183871. [Google Scholar] [CrossRef] [PubMed]
- Vennin, C.; Chin, V.T.; Warren, S.C.; Lucas, M.C.; Herrmann, D.; Magenau, A.; Melenec, P.; Walters, S.N.; Del Monte-Nieto, G.; Conway, J.R.W.; et al. Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis. Sci. Transl. Med. 2017, 9, eaai8504. [Google Scholar] [CrossRef]
- Al-Hilal, T.A.; Hossain, M.A.; Alobaida, A.; Alam, F.; Keshavarz, A.; Nozik-Grayck, E.; Stenmark, K.R.; German, N.A.; Ahsan, F. Design, synthesis and biological evaluations of a long-acting, hypoxia-activated prodrug of fasudil, a ROCK inhibitor, to reduce its systemic side-effects. J. Control. Release 2021, 334, 237–247. [Google Scholar] [CrossRef]
- Kim, S.; Kim, S.A.; Nam, G.-H.; Hong, Y.; Kim, G.B.; Choi, Y.; Lee, S.; Cho, Y.; Kwon, M.; Jeong, C.; et al. In situ immunogenic clearance induced by a combination of photodynamic therapy and rho-kinase inhibition sensitizes immune checkpoint blockade response to elicit systemic antitumor immunity against intraocular melanoma and its metastasis. J. ImmunoTherapy. Cancer 2021, 9, e001481. [Google Scholar] [CrossRef]
- Nam, G.-H.; Lee, E.J.; Kim, Y.K.; Hong, Y.; Choi, Y.; Ryu, M.-J.; Woo, J.; Cho, Y.; Ahn, D.J.; Yang, Y.; et al. Combined Rho-kinase inhibition and immunogenic cell death triggers and propagates immunity against cancer. Nat. Commun. 2018, 9, 2165. [Google Scholar] [CrossRef]
- Federico, C.; Alhallak, K.; Sun, J.; Duncan, K.; Azab, F.; Sudlow, G.P.; De La Puente, P.; Muz, B.; Kapoor, V.; Zhang, L.; et al. Tumor microenvironment-targeted nanoparticles loaded with bortezomib and ROCK inhibitor improve efficacy in multiple myeloma. Nat. Commun. 2020, 11, 6037. [Google Scholar] [CrossRef]
- Misek, S.A.; Appleton, K.M.; Dexheimer, T.S.; Lisabeth, E.M.; Lo, R.S.; Larsen, S.D.; Gallo, K.A.; Neubig, R.R. Rho-mediated signaling promotes BRAF inhibitor resistance in de-differentiated melanoma cells. Oncogene 2020, 39, 1466–1483. [Google Scholar] [CrossRef] [PubMed]
- Meng, F.; Su, Y.; Xu, B. Rho-associated protein kinase-dependent moesin phosphorylation is required for PD-L1 stabilization in breast cancer. Mol. Oncol. 2020, 14, 2701–2712. [Google Scholar] [CrossRef]
- Vogel, C.J.; Smit, M.A.; Maddalo, G.; Possik, P.A.; Sparidans, R.W.; Van Der Burg, S.H.; Verdegaal, E.M.; Heck, A.J.R.; Samatar, A.A.; Beijnen, J.H.; et al. Cooperative induction of apoptosis in <scp>NRAS</scp> mutant melanoma by inhibition of MEK and ROCK. Pigment Cell Melanoma Res. 2015, 28, 307–317. [Google Scholar] [CrossRef] [PubMed]
- Iskit, S.; Lieftink, C.; Halonen, P.; Shahrabi, A.; Possik, P.A.; Beijersbergen, R.L.; Peeper, D.S. Integrated in vivo genetic and pharmacologic screening identifies co-inhibition of EGRF and ROCK as a potential treatment regimen for triple-negative breast cancer. Oncotarget 2016, 7, 42859–42872. [Google Scholar] [CrossRef] [PubMed]
- Rontogianni, S.; Iskit, S.; van Doorn, S.; Peeper, D.S.; Altelaar, M. Combined EGFR and ROCK Inhibition in Triple-negative Breast Cancer Leads to Cell Death Via Impaired Autophagic Flux. Mol. Cell. Proteom. 2020, 19, 261–277. [Google Scholar] [CrossRef] [PubMed]
Interaction | Interaction Type | Components Involved | Specific Regions | Significance |
---|---|---|---|---|
BF1 | Head–Head | Blocked Head (BH) ↔ Free Head (FH) | BH: Loop I365–N381, Helix T382–L390 ↔ FH: Helix E727–Y734, Loop E735–D748 | Stabilizes the myosin heads in the pre-power stroke state, preventing ATP hydrolysis and actin binding. |
BF2 | Head–Head | BH RLC ↔ FH RLC | N terminal lobes: BH: Helix A, A–B linker ↔ FH: Helix D, A–B linker | Strengthens head–head interaction, crucial for regulating muscle activity in the 10S state. |
TB1 | Head–Tail | Blocked Head (BH) ↔ Tail (Seg3) | Seg3: L1604–E1612 ↔ BH: Loop L450–F460 | Provides weak electrostatic interaction that contributes to maintaining the 10S state. |
TB2 | Head–Tail | Blocked Head (BH) ↔ Tail (Seg2) | Seg2: L1431–D1436 ↔ BH: Helix K72–D74 (SH3 domain) | Physically blocks BH converter domain movement, preventing ATP turnover. |
TB3 | Head–Tail | Blocked Head (BH) ↔ Tail (Seg2) | Seg2: Q1445–L1452 ↔ BH: R718, L766 (near converter domain) | Inhibits necessary movements in the BH, trapping ATP hydrolysis products. |
TB4 | Head–Tail | Blocked Head (BH) ↔ Tail | Tail: L1494–L1498 ↔ BH: Helix D (ELC N-lobe) | Stabilizes 10S conformation by reinforcing the interaction between the tail and BH. |
TB5 | Head–Tail | Blocked Head (BH) ↔ Tail (Seg3) | Seg3: A1577–R1584 ↔ BH: Helix E (RLC) | Further stabilizes the 10S structure by anchoring Seg3 to the BH regulatory domain. |
TB6 | Head–Tail | Blocked Head (BH) RLC ↔ Tail (Seg3) | BH RLC N terminal extension ↔ Seg3: Residues 1560–1572 | Crucial for maintaining the 10S state by strengthening the head–tail interaction, involving the phosphorylation domain (PD). |
TF1 | Head–Tail | Free Head (FH) ↔ Tail (Seg1) | FH: CM Loop T404–K420 ↔ Seg1: M925–A941 | Inhibits FH actin binding, keeping the myosin in an inactive state. |
TF2 | Head–Tail | Free Head (FH) ↔ Tail (Seg1) | FH: Loop 2 K626–T658 ↔ Seg1: M925–A941 | Further inhibits actin binding by the FH, reinforcing the 10S state. |
TF3 | Head–Tail | Free Head (FH) RLC ↔ Tail (Seg1) | FH RLC Helix A ↔ Seg1: L850–Q856 | Influences regulatory movements of the FH RLC, contributing to the stability of the 10S conformation. |
TT1 | Tail–Tail | Segment 1 (Seg1) ↔ Segment 3 (Seg3) | Weak electrostatic interactions across several regions | Supports the compact folding of the tail, crucial for maintaining the 10S state. |
TT2 | Tail–Tail | Segment 1 (Seg1) ↔ Segment 3 (Seg3) | Seg1: R910–M925 ↔ Seg3: L1628–E1647 | Maintains close alignment of the tail segments, reinforcing the 10S conformation. |
Tumor Type | Mechanism | References |
---|---|---|
Squamous cell carcinoma |
| [200,201] |
Tumor suppressor: | ||
| [202] | |
| [202] | |
| [203] | |
Melanoma |
| [204] |
Tumor promoter: | ||
Gliomas |
| [205] |
| [206] | |
Nasopharyngeal carcinoma |
| [207,208,209,210,211] |
Lung carcinoma |
| [212,213] |
| [214] | |
| [215] | |
Hepatocellular carcinoma |
| [216] |
| [217] | |
Esophageal squamous cell carcinoma |
| [218,219,220] |
Pancreatic cancer |
| [221] |
Gastric cancer |
| [222,223] |
Colorectal cancer |
| [224,225,226] |
Breast cancer |
| [95] |
| [227] | |
| [228] | |
Renal cancer |
| [229] |
| [230] | |
Prostate cancer |
| [231] |
Osteosarcoma |
| [232] |
Thyroid cancer |
| [233] |
| [234] | |
Acute myeloid leukemia |
| [235] |
Diffuse large B cell lymphoma |
| [236] |
Drugs | Mode of Action | References |
---|---|---|
Blebbistatin |
| [320] |
Cinobufotalin |
| [217,255] |
Apatinib |
| [205] |
Disulfiram |
| [323] |
C70-EDA |
| [324] |
J13 |
| [325] |
Staurosporine |
| [222] |
Homoharringtonine |
| [326] |
Fasudil |
| [327] |
Ripasudil and Netarsudil |
| [328,329] |
Belumosudil |
| [330,331] |
Y27632 |
| [332] |
GSK269962A |
| [333] |
AT13148 |
| [334] |
BDP5290 |
| [335] |
Rhodblock 6 |
| [336] |
RKI-1447 and RKI-18 |
| [337,338] |
Thiosemicarbazone iron chelators |
| [339] |
Combretastatin (CA-4) |
| [340] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Feroz, W.; Park, B.S.; Siripurapu, M.; Ntim, N.; Kilroy, M.K.; Sheikh, A.M.A.; Mishra, R.; Garrett, J.T. Non-Muscle Myosin II A: Friend or Foe in Cancer? Int. J. Mol. Sci. 2024, 25, 9435. https://doi.org/10.3390/ijms25179435
Feroz W, Park BS, Siripurapu M, Ntim N, Kilroy MK, Sheikh AMA, Mishra R, Garrett JT. Non-Muscle Myosin II A: Friend or Foe in Cancer? International Journal of Molecular Sciences. 2024; 25(17):9435. https://doi.org/10.3390/ijms25179435
Chicago/Turabian StyleFeroz, Wasim, Briley SoYoung Park, Meghna Siripurapu, Nicole Ntim, Mary Kate Kilroy, Arwah Mohammad Ali Sheikh, Rosalin Mishra, and Joan T. Garrett. 2024. "Non-Muscle Myosin II A: Friend or Foe in Cancer?" International Journal of Molecular Sciences 25, no. 17: 9435. https://doi.org/10.3390/ijms25179435