Next Article in Journal
Targeting CD36-Mediated Lipid Metabolism by Selective Inhibitor-Augmented Antitumor Immune Responses in Oral Cancer
Previous Article in Journal
Molecular Characterization of Odorant-Binding Protein Genes Associated with Host-Seeking Behavior in Oides leucomelaena
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Muscle Myosin II A: Friend or Foe in Cancer?

1
Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA
2
Cancer Research Scholars Program, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH 45267, USA
3
Division of Endocrinology, University of Cincinnati, Cincinnati, OH 45267, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(17), 9435; https://doi.org/10.3390/ijms25179435
Submission received: 23 July 2024 / Revised: 26 August 2024 / Accepted: 28 August 2024 / Published: 30 August 2024
(This article belongs to the Section Molecular Oncology)

Abstract

:
Non-muscle myosin IIA (NM IIA) is a motor protein that belongs to the myosin II family. The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of NM IIA. NM IIA is a hexamer and contains three pairs of peptides, which include the dimer of heavy chains, essential light chains, and regulatory light chains. NM IIA is a part of the actomyosin complex that generates mechanical force and tension to carry out essential cellular functions, including adhesion, cytokinesis, migration, and the maintenance of cell shape and polarity. These functions are regulated via light and heavy chain phosphorylation at different amino acid residues. Apart from physiological functions, NM IIA is also linked to the development of cancer and genetic and neurological disorders. MYH9 gene mutations result in the development of several autosomal dominant disorders, such as May-Hegglin anomaly (MHA) and Epstein syndrome (EPS). Multiple studies have reported NM IIA as a tumor suppressor in melanoma and head and neck squamous cell carcinoma; however, studies also indicate that NM IIA is a critical player in promoting tumorigenesis, chemoradiotherapy resistance, and stemness. The ROCK-NM IIA pathway regulates cellular movement and shape via the control of cytoskeletal dynamics. In addition, the ROCK-NM IIA pathway is dysregulated in various solid tumors and leukemia. Currently, there are very few compounds targeting NM IIA, and most of these compounds are still being studied in preclinical models. This review provides comprehensive evidence highlighting the dual role of NM IIA in multiple cancer types and summarizes the signaling networks involved in tumorigenesis. Furthermore, we also discuss the role of NM IIA as a potential therapeutic target with a focus on the ROCK-NM IIA pathway.

1. Introduction

Cytoskeletal proteins, such as actin filaments (7–8 nm), intermediate filaments (10 nm), and microtubules (24 nm), are important for cell motility, contraction, polarity, the maintenance of cell morphology, and mechanical support during cytokinesis [1]. Actin filaments and microtubules maintain the directionality of intracellular transport and serve as paths for motor proteins, including myosin, dyneins, and kinesins [1,2]. Intermediate filaments absorb mechanical stress and provide integrity for the cytoskeleton [3]. Myosin motor proteins are molecular machines that perform various motor functions, which require mechanical force and movement. There are more than 30 unique classes of proteins in the myosin superfamily with different distribution patterns and functions [4,5,6]. Non-muscle myosin IIA (NM IIA) is a hexamer composed of three pairs of peptides: a dimer of heavy chains (230 kDa), essential light chains (ELCs, 17 kDa), and regulatory light chains (RLCs, 20 kDa).
In smooth and striated muscles, myosin regulates muscle contraction, whereas non-muscle myosin (NMII) regulates cell migration, adhesion, cytokinesis, and polarity. NMIIs are also important for the growth and differentiation of muscle cells [7]. Additionally, they have a distinct function in the maintenance of smooth muscle tension [8,9]. Myosin binds to actin filaments and induces the magnesium-dependent hydrolysis of ATP to generate mechanical force and tension that propels the sliding of actin filaments. This mechanical force is used for cell motility and division, the intracellular transport of molecules, the maintenance of cell polarity, and signal transduction [10,11]. The cellular functions of myosin proteins vary greatly due to differences in their enzymatic activity and regulation. A total of 38 genes encode various myosin proteins and are divided into 12 classes based on their sequence similarities [11].
Vertebrates have three different isoforms of non-muscle myosin II heavy chains: NMHC IIA, NMHC IIB, and NMHC IIC [12]. The genomes of Saccharomyces cerevisiae (yeast), Drosophila melanogaster (fruit fly), and Dictyostelium discoideum (amoeba) carry a single myosin heavy chain (MHC) gene encoding the NMHC II isoform [13,14,15]. A structural analysis revealed 60–80% identity in the primary sequence of NMHC IIs with the highest similarity in the head domains [16,17]. NMHC IIs combined with myosin light chains (ELC plus RLC) are denoted as NM IIA, NM IIB, and NM IIC, respectively. Although NMII isoforms share sequence and structural similarity, they differ significantly in their motor head ATPase activity, the rate of assembly of myosin filaments, and in the time spent attaching to actin filaments [18,19,20]. Furthermore, each isoform exhibits different patterns of expression in tissues with distinct intracellular distributions [21,22,23,24,25,26]. However, it is important to note that they also share overlapping functions; for example, all NMII isoforms efficiently contribute to cytokinesis [27].
NM IIA is involved in embryogenesis, organogenesis, and immunity; however, its critical role in cancer, autosomal dominant disorders, cataracts, infections, kidney and vascular diseases, and neuronal disorders is gaining importance. This study provides an in-depth review of the structure, function, and regulation of NM IIA. We focus on understanding the role and significance of NM IIA in tumorigenesis as some studies have reported it to be a tumor suppressor, whereas others have shown it to be a tumor promoter. Furthermore, we address the pathological consequences of MYH9 mutations. Figure 1 shows the frequency of MYH9 mRNA expression and alterations across 32 pancancer studies obtained from cBioPortal (www.cbioportal.org). Finally, we address the potential role of NM IIA as a therapeutic target, with a particular focus on targeting the ROCK-NM IIA pathway.

2. Structure of NM IIA

The human MYH9 (myosin heavy chain 9) gene is located on chromosome 22q12-13 and is composed of 41 exons. It is a widely expressed housekeeping gene with a high GC content and no TATA boxes. MYH9 in mice encodes a protein that is 97% identical to human NMHC IIA [28]. For this review, we used the following nomenclature: MYH9—a gene; NMHC IIA—a protein produced by the MYH9 gene; and NM IIA—a fully functional hexamer composed of two homodimers of NMHC IIA heavy chains, two ECLs (MYL6 gene), and two RLCs (MYL 12A/B genes) that stabilize the heavy chain structure [28] (Figure 2). All myosin II complexes, including the isoforms, have both ELC and RLC, which are essential for its function and regulation. Myosin complex assembly involves complicated machinery. For example, in the Golgi apparatus, UNC-45/CRO1/She4 (UCS) chaperones regulate the folding of heavy chain peptides of the complex. Furthermore, the UCS chaperones are evolutionarily conserved and promote myosin-related functions including cytokinesis, muscle development, endocytosis, and RNA transport. In Caenorhabditis elegans, UCS chaperones regulate myosin II complex function to ensure proper filament assembly [29].
The heavy chain of NM IIA is divided into two regions: the N and C termini. The N terminus comprises a globular motor head domain, the functional engine of myosin II, which can bind to actin and generate force via MgATPase activity (with Mg2+ serving as an essential cofactor) [30,31]. The neck region, which follows the head region, binds to both myosin light chains (ELC and RLC) [32]. The tail domain or the C terminus is important for homodimerization with other myosin in a helical fashion [33]. Despite showing significant sequence similarity, these regions exhibit different binding affinities for actin filaments. Due to this differential actin binding, different isoforms carry out mechanical work in the cell with varying energy efficiencies [34]. The motor head is composed of four subdomains: the N terminal SH3-like motif, upper subdomain, lower subdomain, and the converter region [34].
Figure 2. This figure illustrates the two assembly states of non-muscle myosin II (NMII): the 10S assembly-incompetent state and the 6S assembly-competent state. In the 10S assembly-incompetent state (left), the myosin molecule is folded, with the globular head and heavy chain regions interacting through various tail-binding sites, leading to a compact structure. Many intramolecular interactions keep the 10S state in an inactive stable form (Table 1). The interactions involve the Blocked Head (BH), which is the myosin head prevented from binding to actin, and the Free Head (FH), another myosin head that is inhibited but not directly involved in actin binding in the 10S state. The transition to the 6S assembly-competent state (right) occurs upon the phosphorylation of the regulatory light chain (RLC), resulting in an extended, active conformation where the heavy chain regions are aligned, allowing for actin binding and ATPase activity, which are essential for NMII’s role in cell contractility and motility. ELC: Essential Light Chain; RLC: Regulatory Light Chain; FH: Free Head; BH: Blocked Head; TF: Tail–Free Head Interaction; TB: Tail–Blocked Head Interaction; TT: Tail–Tail Interaction.
Figure 2. This figure illustrates the two assembly states of non-muscle myosin II (NMII): the 10S assembly-incompetent state and the 6S assembly-competent state. In the 10S assembly-incompetent state (left), the myosin molecule is folded, with the globular head and heavy chain regions interacting through various tail-binding sites, leading to a compact structure. Many intramolecular interactions keep the 10S state in an inactive stable form (Table 1). The interactions involve the Blocked Head (BH), which is the myosin head prevented from binding to actin, and the Free Head (FH), another myosin head that is inhibited but not directly involved in actin binding in the 10S state. The transition to the 6S assembly-competent state (right) occurs upon the phosphorylation of the regulatory light chain (RLC), resulting in an extended, active conformation where the heavy chain regions are aligned, allowing for actin binding and ATPase activity, which are essential for NMII’s role in cell contractility and motility. ELC: Essential Light Chain; RLC: Regulatory Light Chain; FH: Free Head; BH: Blocked Head; TF: Tail–Free Head Interaction; TB: Tail–Blocked Head Interaction; TT: Tail–Tail Interaction.
Ijms 25 09435 g002
The neck region is like a lever arm that transforms the force produced by the rotation of the motor head into mechanical work. Furthermore, an extended helix with two conserved IQ motifs (IQxxxRGxxxR) is present in the neck region. However, different classes of myosin possess different numbers of IQ motifs [35]. IQ motifs form a hydrophobic seven-turn α-helix chain that can bind to light chains (RLC or ELC) in a Ca2+-independent manner. ELC binds the first IQ motif that is located closer to the N-terminus, providing stability to the non-muscle heavy chain (NMHC). RLC binds to the adjacent IQ-like motif that is closer to the coiled coil region, providing both stability and functional regulation to NMHC. The phosphorylation of serine 19 (Ser 19) on the RLC increases the motor head ATPase activity and contributes to filament assembly [16]. The length of the neck region directly impacts myosin II motor function [36].
Various genes encode myosin light chains (MLCs) in humans. For example, MYL9, MYL12A, and MYL12B encode RLCs, whereas MYL6 and MYL6B encode ELCs [37]. Alternative splicing is crucial for regulating gene expression and enhancing protein diversity in eukaryotes. Both MYL9 and MYL6 undergo alternative splicing to diversify NMII multimeric complexes. Studies have shown that ELC MYL6 might interact specifically with NMHC IIC to regulate it both spatially and temporally. Thus, a specific function is assigned to NMHC IIC in addition to its traditional mechanical and kinetic functions [18,38]. The coiled coil region of C-terminus homodimerizes to form a single rod-like structure. These rod-like structures can interact with other myosin proteins to assemble as bipolar filaments, which re nucleation sites for the formation of larger filaments measuring approximately 300 nm in length [39,40]
The proline kink in the helix located at the junction of the head and rod domains allows the myosin II complex to fold into a compact shape. When the RLC is unphosphorylated or inactive, the head and tail domains interact to create a compact conformation of myosin II complex [41,42]. The inactive 10S compact structure of myosin II sediments at 10 S (Svedberg) and does not possess any actin-activated ATPase activity [43,44]. The serine 19 (Ser19) phosphorylation of RLC by the calcium–calmodulin–myosin light chain kinase (MLCK) pathway [45] results in the unfolding of the 10S structure and subsequent formation of the assembly-competent 6S. The 6S form sediments at 6 Svedberg. In their active state, the tail domain assembles in an anti-parallel fashion into ~300 nm long bipolar thick filaments. These bipolar filaments crosslink actin filaments to create different meshes of actomyosin bundles. In the absence of RLC, the tail domains form disorganized filaments. Therefore, the RLC is important to maintain a non-aggregated myosin state that can form filaments and does not directly contribute to ATP hydrolysis [46].
All of the paralogs of NMII can hydrolyze ATP and participate in different cellular processes; however, the rate of hydrolysis differs. For example, compared with muscle myosin II, NM IIA hydrolyzes ATP at a slower rate [47]. Furthermore, the binding time to actin filaments is also low, which might be the reason for the low stability of NM IIA mini-filaments [48]. Vincente et al. reported the continuous assembly and disassembly of NM IIA filaments during membrane protrusion [49]. In contrast, NM IIB and NM IIC exhibit fast kinetics of ATPase hydrolysis, which makes them ideal for force generation in non-muscle cells [50,51]. Therefore, the turnover and assembly rates of NM IIB and NMIIC are slower than those of NM IIA [52].

3. Regulation of NM IIA

The specific group of proteins that regulate myosin activity in skeletal and cardiac muscles includes the troponin–tropomyosin complex, myosin-binding protein C (MyBP-C in cardiac muscle), myosin-binding protein H (MyBP-H in skeletal muscle), myosin light chain kinase (MLCK), protein kinase A (PKA) and protein kinase C (PKC), and calcium ions (Ca2+) [53] (Figure 3). The three paralogs, NM IIA, NMIIB, and NM IIC, display 60–80% of amino acid sequence similarity and share a common quaternary structure. However, in a homeostatic state, these three paralogs differ in their regulatory mechanisms [54].

3.1. Role of RLC Phosphorylation in Regulating NM IIA Activity

As discussed earlier, NM IIA can assume two different conformations: the 10S form in which NM IIA is compact and inactive, and the 6S form, which is the unfolded active form. In the 10S inactive folded form, the important biological properties of NMII, including ATPase hydrolysis activity, bipolar filament formation, and actin filament sliding, are inhibited [55,56,57]. RLCs undergo reversible phosphorylation at various amino acid residues, including serine1/2/19 and threonine 9/18, to regulate myosin II activity. RLC phosphorylation controls the activation and inactivation of myosin II, which was discovered in rabbit skeletal muscle [58].
The phosphorylation of RLC (serine 19 and/or threonine 18), which activates NM IIA, can be achieved by several kinds of kinases. Among the various phosphorylation sites on RLC, Ser19 controls the activation of NM IIA and the subsequent formation of bipolar filaments [59]. Phosphorylation introduces a negative charge that breaks the head–tail interaction of the inactive form [56,60,61]. Ser19 phosphorylation increases both the ATPase activity and actin-binding capacity of NM IIA [62,63]. Ser19 phosphorylation is mediated by several kinases, including MLCK, Rho-associated coiled coil-containing kinase (ROCK), and myotonic dystrophy kinase–related Cdc42-binding kinase (MRCK). ROCK enzymes target myosin phosphatase target subunit 1 (MYPT1) to increase the phosphorylation of Ser19 [64]. A conformational change from 10S to 6S via Ser19 phosphorylation is observed in all of the paralogs of NMII; however, the reverse change in conformation from 6S to 10S is paralog-dependent. Therefore, paralog dependency during the reverse conformation step is essential for controlling isoform-specific functions [65].
Apart from Ser19 phosphorylation, additional phosphorylation on Thr18 can modulate NM IIA activity. Thr18 phosphorylation stabilizes NM IIA in the 6S conformation, making the filaments more organized [66]. Therefore, upon Ser19 and Thr18 phosphorylation, the cells can exhibit three different RLC statuses: non-phosphorylated RLC, pSer19, and pSer19 + pThr18 [67]. Biphosphorylation can influence the strength of filaments; for example, filaments with pThr18/pSer19-RLC residues are present at the trailing edge [65], or in the mitotic spindle midzone, which is subjected to greater stress [68,69]. Rho kinase and protein phosphatase 1 can regulate muscle contraction and cell motility by controlling the phosphorylation state of myosin light chains (MLCs). Rho kinase phosphorylates and inhibits myosin light chain phosphatase (MLCP), a complex that includes protein phosphatase 1 as its catalytic subunit. This inhibition reduces protein phosphatase 1 activity, leading to the decreased dephosphorylation of MLC (MLC remains phosphorylated) and the sustained activation of NM IIA [28].
In addition to the phosphorylation of Ser19 and Thr18, other phosphorylation events may reduce NM IIA activity. For example, protein kinase C (PKC)-induced phosphorylation at the Ser1/2 and Thr9 positions renders NMII a poor substrate for MLCK to decrease its activity [70,71]. However, these phosphorylation events, unlike those at Ser19/Thr18, appear to have a modulatory effect on in vivo NMII activity [72]. PKC-induced phosphorylation regulates actin filament reorganization induced by platelet-derived growth factor (PDGF) in platelets [73]. Asokan et al. reported that in mesenchymal cells, local myosin IIA inactivation via the Ser1/2 phosphorylation of RLC impaired mini-filament contraction downstream of conventional PKC [74]. Kinases involved in RLC phosphorylation events are present in specific locations inside the cell. For example, MLCK is localized next to the cell membrane and activates myosin II in response to stimulation via the Ca2+-calmodulin complex [75]. Abl tyrosine kinase and p21-activated kinase 1 (PAK1) are involved in regulating the activity and localization of MLCK inside the cell [76,77,78]. Similarly, RhoA, which activates both ROCK and citron kinase, is localized in the middle of the cytoplasm. During the developmental stage, neuroepithelial cells, which are a type of stem cell, display the RLC phosphorylation of NMII mediated by the intracellular localization of ROCK, which is directed by Shroom 3 [79,80]. DAPK3 is a member of the death-associated protein kinase (DAPK) family. In cells undergoing apoptosis, DAPK3 localizes to the nucleus, where it phosphorylates RLC in a Ca2+- calmodulin-independent manner [81].

3.2. Role of NMHC Phosphorylation in Regulating NM IIA Activity

The first evidence of NMHC phosphorylation was observed in phagocytes, where it is required for filament assembly and disassembly [82]. Some of the common phosphorylation sites include Thr1800, Ser1803, and Ser1919 in the coiled coil domain and Ser1943 in the non-helical tail piece (Figure 4). Several kinases are involved in heavy-chain phosphorylation, including casein kinase II (CK II), PKC, the alpha kinase family, and transient receptor potential melastatin 7 (TRPM7) [83,84,85,86,87].
PKC family enzymes phosphorylate the Ser1916 and Ser1937 residues in the coiled coil domain to inhibit NMII assembly [88,89]. Phorbol esters are organic compounds that are derived from the resin of the tropical plant family Euphorbiaceae. These compounds are tumor inducers and activate PKC family members. Phorbol esters promote the phosphorylation of NM IIA at the Ser1916 residue in thrombocytes, T-lymphocytes, and mast cells [90,91]. In mast cells, the PKC-dependent phosphorylation of the NMHC is correlated with mast cell secretion via exocytosis [92]. Apart from NM IIA, PKC also phosphorylates serine in NM IIB and threonine in NM IIC [83,87].
Casein kinase II (CK II) is one of the earliest kinases discovered and has contributed immensely to the development of the human phosphoproteome [93]. It is a serine/threonine kinase that is involved in tumorigenesis, where it regulates the activity of malignant hallmarks [94]. CK II phosphorylates the Ser1943 residue of NM IIA to inhibit assembly [95]. In addition, the phosphorylation of the Ser1943 residue by CK II also controls the organization of the NM IIA assembly [84]. Beach et al. reported that in murine mammary gland epithelial cells, the induction of EMT via TGF-β signaling increases the phosphorylation of NMHC IIA at the Ser1916 and Ser1943 residues. These molecular changes are associated with alterations in filament dynamics, indicating a potential role in the cellular transitions associated with EMT [96].
Using mesenchymal stem cells, Rabb et al. reported the significance of Ser1943 as a mechanosensor. When NM IIA is phosphorylated at the Ser1943 position, stem cells exhibit a random migratory pattern on a soft matrix. However, the dephosphorylation of Ser1943 was observed when the cells were plated on a stiffer matrix. Stem cells switched to NM IIB-dependent migration when plated on a stiffer matrix [97]. Dulyaninova et al. showed that EGF stimulation results in transient Ser1943 phosphorylation of NM IIA to inhibit filament assembly [95]. Beach et al. replaced the wild-type NM IIA with a mutant NM IIA in which the Ser1943 residue cannot be phosphorylated. They observed that cells with mutant NM IIA showed an increase in assembly at lamellar regions and a decrease in cell migration [72]. Similarly, Rai et al. mutated either Ser1916 or Ser1943 to alanine (serine to alanine) and observed a decrease in NM IIA filament formation at the leading edge. When studying 3D migration, they observed that knocking down NM IIA resulted in a defect in cell invasion [98]. Transient receptor potential melastatin-subfamily member 7 (TRPM7) is a membrane protein important for the regulation of ions, such as Zn2+, Mg2+, and Ca2+ [99]. TRPM7 kinase can phosphorylate the heavy chain of all three paralogs of NMII. For example, TRPM7 phosphorylates Ser1803/1808 and Thr1800 in NM IIA to decrease filament assembly in vitro and reduce actomyosin formation in vivo [85,86].

3.3. Role of Protein Interactions in Regulating NM IIA Activity

In this section, we will focus on various protein interactions that can regulate NM IIA activity. Several proteins, including Lethal (2) giant larvae (Lgl), S100A4, and myosin binding protein H-like (MYBPHL), can interact with NM IIA to modulate its localization and filament assembly. S100A4/MTS1 is a member of the S100 family that can promote tumor development and metastasis. The S100 family of proteins regulates cell cycle events and are localized in the cytoplasm and nucleus [100]. In response to a conformational change induced by Ca2+, S100A4 binds to the C terminus using a hydrophobic cleft to disassemble the NM IIA filaments. Kiss et al. and Ramagopal et al. elucidated the crystal structure of the S100A4-NM IIA complex [101,102]. Although Ser1943 is not directly involved, its phosphorylation alone can disrupt the S100A4-NM IIA complex formation [84]. Li et al. showed that phagocytes lacking S400A4 display an over-assembly of NM IIA filaments, which results in reduced chemotaxis [103]. S100A4 knockdown phagocytes also exhibit a decrease in podosome formation [104]. In contrast, the overexpression of S100A4 induces metastasis by increasing cytoskeletal plasticity and migration [105]. S100-P, another member of the S100 family, can regulate NM IIA filament assembly and migration [106].
Lgl proteins are present mainly in the apical and basolateral domains of the cell. Lgl proteins are important for regulating the apico-basal polarity of cells [107]. Lgl1, a member of the Lgl family, is a tumor suppressor that interacts with NM IIA in vitro to inhibit filament formation [108,109]. Atypical protein kinase C (aPKCζ) phosphorylates Lgl1 to disrupt its interaction with NM IIA. NMIIA and aPKCζ compete to interact with Lgl1 directly [108,109]. Therefore, it is possible that Lgl1 and NM IIA might be involved in determining front–rear polarity. LIM and calponin-homology domains 1 (LIMCH1) is an actin stress fiber-associated protein that regulates NMII activity. Lin et al. reported that the N terminal region of LIMCH1 specifically binds to the head motor domain of NM IIA [110]. The knockdown of LIMCH1 enhanced migration via a decrease in actin fibers and focal adhesions in HeLa cells [110]. In lung adenocarcinoma cells, MYBPHL interacts with Rho kinase 1 to inhibit RLC phosphorylation on NM IIA [111]. MYBPHL interacts with the rod domain of the NM IIA heavy chain to inhibit the assembly and reduce the motility of lung adenocarcinoma cells. In a rat model, Zhu et al. reported that the interaction of MYBPHL with Rho kinase 1 reduces hyperplasia after injury in the carotid artery [112]. Tropomyosin 4.2 (Tpm4.2) is a tropomyosin isoform of actin-binding protein. Tpm4.2 recruits NM IIA to actin filaments during the formation of stress fibers [113].

4. Physiological Functions of NM IIA

The expression of NMII isoforms significantly differs in cells. Thrombocytes and the spleen express only the NM IIA isoform, whereas phagocytes express both the NM IIA isoform and the NM IIB isoform. Similarly, mature cardiomyocytes express only the NM IIB isoform, whereas non-myocytes express both the NM IIA and NM IIB isoforms. Lung tissue expresses the same amounts of NM IIA and NM IIC (40% each) and approximately 20% of NM IIB [114]. Apart from the presence of multiple isoforms, all three paralogs of NMII can co-assemble to form heterotypic filaments inside the cell. This finding implies that NMII isoforms can perform specific functions alone and co-assemble with other paralogs to carry out redundant functions [115,116,117]. The co-polymerization of NM IIA and NM IIB paralogs, coupled with their distinct turnover rates, leads to the formation of a polarized actin–NMII cytoskeleton [118]. Myosin 18A is a non-muscle II myosin that lacks ATP hydrolysis activity and can co-assemble with different NMII filaments to widen the functional range of each subtype [119].
The presence of NM IIA is important for mouse embryonic development. For example, NM IIA knock mice show poor development of the visceral endoderm [120,121]. The absence of NM IIA disrupts cell junctions, resulting in the loss of cell polarization with a disorganized endoderm [121]. NM IIA knockout embryos exhibited multinucleated cells, indicating abnormalities in cell division. In C. elegans, Guo et al. deleted NMY-2, which is one of the two NMHCs, and observed impaired cytokinesis in the embryo [122]. Similar results were observed by Osorio et al. when NMY-2 ATPase activity was reduced, indicating the essential role of NM IIA motor activity in cell division [123]. Mouse studies revealed that the presence of low ATPase activity in mutant NM IIA-R702 leads to abnormalities in placental development, indicating the significance of fully active NM IIA in the development process [124]. NM IIA is crucial for tissue-level developmental processes. Lienkamp et al. showed that the loss or mutation of the MYH9 gene results in nephropathies [125]. Dysregulated NM IIA activity is linked to the pathology of glomerular diseases [126]. The regulation of NM IIA activity by Wnt signaling pathways is important for the polarization of the auditory epithelium [127]. Reduced NM IIA function may disrupt the localization of junction proteins such as vinculin [127,128]. Such disturbances may interfere with sound perception, potentially contributing to deafness, a condition linked with MYH9-related disorders.
Studies using various models, such as Dictyostelium and starfish [129,130,131], have demonstrated that NM IIA is necessary for mitosis [131,132]. Using both single-cell (amoebas) and multicellular organisms (fungi) and animals, studies have shown that effective cytokinesis relies on the formation of an actomyosin contractile ring enriched with NMII, which facilitates the physical separation of daughter cells [133]. Although NMII is crucial for cytokinesis, studies have shown that its motor function is not necessary in budding yeast. Some studies have reported that NMII motor activity is not a prerequisite for cell division [129,134,135,136]. However, any of the NM II isoforms can promote cell division [137]. NM IIA activity is significant for polarization and asymmetrical cell division [138,139].
Cell adhesion is dependent on integrins, which act as transmembrane linkers between the ECM and actin. Integrins, in conjunction with actin filaments and tension generated by NM IIA, facilitate cell migration [140]. Among the NMII isoforms, NM IIA shows a faster rate of ATP hydrolysis and actin filament sliding [141]. Therefore, NM IIA is well suited for rapid remodeling and actin filament assembly at the leading edge [118,142,143]. Many forms of cell polarity are marked by the asymmetric distribution of proteins, cell organelles, and cytoplasm. For example, the apical–basal polarity of epithelial cells (composed of apical and basolateral junctional complexes) is the most common form of cell polarity and is determined by intercellular adhesion contacts [144]. In polarized cells, the junction complex assembles and disassembles at the apical region where connection with actomyosin bundles maintains their integrity and stability [145].
Blebbistatin blocks the myosin head in a complex state, which has a low affinity for actin filaments, leading to a decrease in the size and density of adhesion complexes [146]. NM IIA contractile forces drive the contraction of adherent junctions, which are vital for establishing polarized epithelial tissues both in vitro and in vivo [147,148,149]. Contractile forces are essential for the formation and stability of adhesion complexes [150,151,152]. Ivanov et al. reported that the knockdown of NM IIA via small-interfering RNA (siRNA) in intestinal epithelial cells disrupts cell–cell adhesion and alters morphology [153]. Similarly, in kidney epithelial cells, the knockdown of NM IIA disrupts stable intercellular adhesions [152]. In 2D cell cultures, cells are grown on a flat surface, whereas in 3D cell culture, they grow in a three-dimensional environment, typically within a gelatinous matrix or within a solid scaffold. Cells in a 2D environment migrate by reorganizing their actomyosin cytoskeleton to form a distinct trialing and leading edge [154,155]. NMII isoforms promote 2D cell migration, and their inhibition impairs cell motility [156,157]. During migration, the leading or the front edge extends via actin polymerization, forming lamellipodia, while retrograde actin flow, driven by NM IIA, aids in cytoskeletal remodeling. The formation of focal adhesion at the leading edge is important for efficient migration [158,159,160,161,162] (Figure 5). NM IIA is primarily found at the leading or front edge, whereas NM IIB levels are high at the trailing or rear edge.
During retrograde actin flow, NM IIA levels are notable in newly formed filaments due to the faster disassembly rate from NM IIA/NM IIB bipolar filaments. Conversely, NM IIB levels are high at the cell’s rear end due to its association with older actin fibers [118]. Furthermore, while NM IIA contributes to cellular contraction and facilitates the directional migration of the cell, elevated levels of NM IIB hinder cell motility and could enhance cytoskeletal stability [118]. At the rear edge of the cell, NM IIA is essential for the movement and disassembly of focal adhesions. NM IIA promotes forward movement via the retraction of the trailing or rear edge [163,164,165,166]. The different expression levels of NMII isoforms influence the cell migration pattern.
Cells undergoing 3D migration predominantly exhibit mesenchymal and ameboid behaviors [167]. In 3D migration, cells exhibit elongated shapes with thinner protrusions. Mesenchymal migration typically involves an elongated cell morphology and relies on protease-mediated matrix remodeling for movement through narrow spaces. In contrast, ameboid migration is characterized by rounded cell shapes and a more fluid-like movement, often independent of proteolysis, and it is capable of squeezing through matrix gaps. Mesenchymal and ameboid cells exhibit variable adherence to the ECM and differ in contractile forces and the cellular localization of NM IIA [167]. Mesenchymal migration is marked by the presence of NM IIA and actin-rich protrusions at the leading edge [168]. In contrast, ameboid migration is characterized by the presence of NM IIA at the front or rear end, and ECM adhesion is not essential [169,170]. At the leading edge, there is an increase in actomyosin contractility which drives the cell forward, leading to the formation of blebs [171], whereas at the rear end, there are uropods (highly contractile structures) associated with leukocyte movement [169,172].
Plasma membrane blebbing, which is observed in both physiological and pathological states, is well documented during cell migration and apoptosis [173,174,175]. Blebs are dynamic protrusions of the cell membrane controlled by different cytoskeletal proteins [176,177]. Blebs are formed when the actomyosin cytoskeleton at the cell cortex contracts, generating high intracellular hydrostatic pressure [176,177,178]. High intracellular pressure disrupts the plasma membrane and the underlying cytoskeleton to form dynamic protrusions [178,179,180,181,182]. Bleb expansion, which lacks polymerized actin, occurs because of persistent hydrostatic pressure inside the cells [183,184]. The expanded blebs will retract gradually (Figure 6), and prior to retraction, various linker proteins, such as ezrin (which links the plasma membrane to actin), initiate actin polymerization [179]. Furthermore, bleb expansion and retraction are regulated via actomyosin cytoskeletal molecules [179]. Studies have shown that the NM IIA powers the bleb retraction process by further recruiting and accumulating on actin filaments [171,175,185,186].
Rho GTPases are molecular switches that regulate cytoskeleton dynamics. ROCK (a specific serine–threonine kinase) is a downstream molecule of Rho GTPase that can activate NM IIA to induce actomyosin contraction [175,181,184,185]. The ezrin is a cross-linker protein, and its recruitment to the bleb facilitates the activation of RhoA by MyoGEF (myosin-interacting guanine nucleotide exchange factor) [185]. Agents that inhibit the activities of ROCK, MLCK, and NMII (blebbistatin), along with actin-depolymerizing agents, suppress membrane blebbing [178]. Among the three NM II paralogs, NM IIA motor activity is required for bleb retraction [187]. The bleb retraction rate is correlated with NM IIA turnover on bipolar filaments [174,187]. Calcium is essential not only for NMII-mediated contraction but also for subsequent bleb formation and retraction, as shown by studies in which calcium chelators disrupted blebbing [175]. Furthermore, various calcium-dependent mechanisms, including those that activate NMII and facilitate cytoskeletal rearrangements, are known to trigger blebbing [188].
NM IIA is important for glucose-stimulated insulin secretion (GSIS) because it regulates actin and focal adhesion (FA) remodeling [189]. Arous et al. showed that the knockdown or inhibition of NM IIA decreases GSIS and alters actin stress fibers and FAs, impairing insulin secretion [189]. These findings highlight NM IIA’s central regulatory role in orchestrating the cellular processes underlying GSIS. The APPL1 adaptor protein regulates cell proliferation and mediates the adiponectin and insulin signaling pathways. Saito et al. reported that in response to mechanical stretching, APPL1 interacts with NM IIA to enhance glucose uptake [190]. NM IIA is implicated in regulating the biomechanical properties of the mouse aorta [191]. Blebbistatin or MYH9 knockdown suppresses the activity of focal adhesion proteins such as FAK and paxillin [191].
Using motile U2-OS (human osteosarcoma) cells, Fenix et al. showed that NM IIA drives cellular movement and cytokinesis by organizing non-muscle myosin II filaments (NMII-Fs) [192]. NM IIA promotes the formation of NM IIA-F stacks through expansion and concatenation mechanisms, which are regulated by motor activity and actin filament availability. These mechanisms are crucial for cellular dynamics in both the interphase and mitotic phase [192]. MYH9 is important in regulating DNA synthesis [193]. MYH9 interacts directly with dNTPs with varying efficiencies [193]. The stability of MYH9 stability is enhanced by dNTPs, and inhibition with blebbistatin or siRNA leads to decreased cell proliferation, indicating its involvement in DNA synthesis [193]. Podocytes are highly specialized cells of the glomerulus that act as barriers to prevent the filtration of plasma proteins in the urine. NM IIA activity is important for the structure and function of podocytes. The inhibition of NM IIA activity results in alterations in the podocyte structure and increased cell motility [194]. These findings suggest an association between MYH9 mutations and glomerular pathologies [194].
NM IIA is crucial for the maturation of reticulocytes into erythrocytes [195]. NM IIA mediates axon growth and guidance in developing neurons. It is phosphorylated by the intracellular domain of EphA3, which is generated by presenilin-1/γ-secretase cleavage, leading to cytoskeleton rearrangement and facilitating axon elongation. NM IIA activity is crucial for presenilin/EphA3-dependent axon elongation, as evidenced by its ability to reverse axon retraction in presenilin-deficient neurons. Overall, NM IIA acts downstream of EphA3 cleavage to regulate axon growth, highlighting its importance in neuronal development [196].
Studies in zebrafish have shown that NMHC IIA is crucial for the formation and function of the glomerulus. The knockdown of zNMHC IIA (zebrafish NMHC IIA) induces a decrease in mesangial cells and impairs glomerular filtration [197]. NM IIA is also important for regulating cell shape changes during midbrain–hindbrain boundary (MHB) morphogenesis in zebrafish. NM IIA is specifically crucial for shortening cells at the MHB constriction (MHBC). Its activity ensures the proper angle of the MHB tissue. Furthermore, NM IIA knockdown results in an abnormal distribution of actin within MHBC cells. These findings signify the function of NM IIA in coordinating morphological changes that are essential for MHB morphogenesis [198]. NM IIA, along with gelsolin (an actin-binding protein), is integral to collagen phagocytosis by fibroblasts. It is essential for collagen binding and internalization, and its filament assembly is crucial for actin remodeling in nascent phagosomes. This assembly depends on both MLC phosphorylation and elevated intracellular calcium levels, indicating its regulation by calcium signaling. The interaction between NM IIA and gelsolin suggests a coordinated mechanism at collagen adhesion sites, where these proteins enable NM IIA filament assembly and actin remodeling in response to collagen binding, ultimately facilitating the phagocytic process [199].

5. MYH9 as an Oncogene

The MYH9 gene was initially identified as a tumor suppressor. However, subsequent studies have shown that MYH9 is also involved in tumorigenesis and treatment resistance. In addition, it can also play a dual role as a promoter and suppressor based on cancer type (Table 2).

5.1. Head and Neck Squamous Cell Carcinoma

Head and neck squamous cell carcinomas (HNSCCs) originate from the squamous cells of the oropharynx and larynx. MYH9 has been implicated in the development of radiotherapy resistance in HNSCCs. You et al. showed that the overexpression of MYH9 enhanced cell motility via the activation of the MAPK signaling pathway, including the upregulation of ERK, P38, and JNK. MAPK pathway upregulation further activates the Nrf2 transcription factor that decreases the levels of reactive oxygen species (ROS) to promote invasion and radioresistance [200]. NEDD9 (neural precursor cell expressed, developmentally downregulated 9) belongs to the CAS (Crk-associated substrate) family of non-catalytic scaffolding proteins [237]. The activation of integrins and B cell antigen receptor (BCR) induces the hyperphosphorylation of the NEDD9 protein, which regulates tumor invasion and the chemokine response [237,238,239,240]. Although mutations in NEDD9 are rare in tumors, NEDD9 is frequently overexpressed and constitutively hyperphosphorylated, which is correlated with unfavorable clinical outcomes. NEDD9 interacts with NM IIA to increase the phosphorylation of RLC at Ser1943, contributing to matrix metalloproteinase (MMP) secretion and metastasis. The treatment of HNSCC cells with blebbistatin altered cell morphology and decreased the secretion of MMPs 2 and 9 [241].

5.2. Gliomas

Glioma is the most common central nervous system (CNS) tumor originating from glial cells. These tumors are highly invasive and infiltrate the surrounding brain tissue extensively. Among gliomas, glioblastoma represents the most aggressive form, while pilocytic astrocytomas are considered the least malignant type of brain tumor. Glioma cells overexpress MYH9 and high mobility group A1 (HMGA1, a chromatin structural protein). HMGA1 activates the PI3K/Akt/c-Jun signaling pathway to upregulate the transcription of MYH9. High levels of MYH9 ubiquitinate glycogen synthase kinase-3β (GSK-3β) to enhance the nuclear translocation of β-catenin that contributes to invasion and resistance to temozolomide treatment [242].
Thrombospondin 1 (THBS1) is a key player in mechanotransduction and a potential target of apatinib [205]. THBS1 interacts with MYH9 to increase the malignancy of glioma cells. Treatment with apatinib indirectly decreased the levels of MYH9 to inhibit the invasion and migration of glioma cells [205]. Members of the S100 family are calcium-binding proteins that are commonly upregulated in various cancers. Using 94 patients with GBM, Inukai et al. reported the significance of NM IIA in GBM tumorigenesis [206]. Immunohistochemistry data revealed an interaction between S100A4 (a member of the S100 family) and NM IIA that contributes to aggressive phenotypes in GBM. NM IIA inhibition affects the recruitment and migration of S100A4-positive GBM cells along pre-existing blood vessels. This process is mediated by the release of vascular endothelial growth factor A (VEGFA) from S100A4-positive/hypoxia-inducible factor-1α (HIF-1α)-positive tumor cells. NM IIA inhibition leads to tumor progression by enhancing pro-tumorigenic vascular functions [206].
Picariello et al. showed that NM IIA plays a critical role in inhibiting tumor invasion, and its deletion paradoxically increases tumor proliferation, a response that is influenced by the mechanical properties of the tumor environment. On softer surfaces, NM IIA deletion enhances ERK1/2 signaling, promoting proliferation, while on stiffer surfaces, it increases NF-κB activity, which also contributes to tumor progression. The findings suggest that although targeting NMIIA might be effective in preventing tumor invasion, it could inadvertently accelerate tumor growth by activating compensatory signaling pathways. Therefore, the study underscores the need for therapeutic approaches that can simultaneously inhibit both tumor invasion and proliferation, considering the mechanical context of the tumor environment to avoid unintended consequences [243].

5.3. Nasopharyngeal Carcinoma (NPC)

Nasopharyngeal carcinoma (NPC) is a malignant tumor linked with Epstein–Barr virus (EBV) infection. EBV-miR-BART22 is a microRNA that belongs to the EBV-encoded microRNA BART (BamHI A rightward transcript) family [207]. EBV-miR-BART22 increases MYH9 levels via the activation of the PI3K/AKT/c-Jun pathway to induce tumor stemness, metastasis, and resistance to cisplatin. Like in glioma cells, MYH9 promotes GSK3β protein degradation via ubiquitination to promote the nuclear translocation of β-catenin [207].
FOXO1 (a tumor suppressor), also referred to as FKHR (forkhead in rhabdomyosarcoma), belongs to the forkhead box O-class (FoxO) subfamily of transcription factors. FOXO1 regulates cell proliferation, survival, metabolism, and the oxidative stress response [208]. MYH9 is a key player in FOXO1-mediated NPC pathogenesis and chemosensitivity to cisplatin [209]. MYH9 interacts with FOXO1, resulting in decreased levels of MYH9 via the activation of the PI3K/AKT/miR-133a-3p pathway. Low levels of MYH9 lead to a reduced interaction with TNF receptor-associated factor 6 (TRAF6) and GSK3β to inhibit EMT and stemness [209,210,211]. FNDC3B, also referred to as FAD104, is a novel gene that regulates the differentiation of adipocytes and osteoblasts. The abnormal expression of FNDC3B is observed in hepatocellular carcinoma (HCC), acute myeloid leukemia (AML), and cervical cancer [244]. Studies have shown that FNDC3B binds and stabilizes MYH9 to activate the Wnt/β-catenin pathway. MYH9 is implicated in counteracting the suppressive effects of FNDC3B knockdown, suggesting its crucial role in mediating the oncogenic functions of FNDC3B [245].

5.4. Non-Small Cell Lung Cancer (NSCLC)

Non-small cell lung cancers encompass approximately 85–90% of all lung cancers. Katono et al. reported that high MYH9 levels in patients with NSCLC is associated with several adverse clinicopathological features and poor prognosis [212]. Chen et al. reported that MYH9 significantly enhances the stemness of lung cancer cells (LCCs) by regulating the expression of cancer stem cell (CSC) markers, including CD44, SOX2, Nanog, CD133, and OCT4. Furthermore, MYH9 also contributes to NSCLC stemness via the mTOR pathway [213].
The epidermal growth factor receptor (EGFR) gene is overexpressed in most NSCLC cases. Patients with EGFR point mutations respond well to EGFR tyrosine kinase inhibitors (EGFR-TKIs), such as Gefitinib and Erlotinib. Resistance to TKIs develops due to a secondary mutation (T790M) in the intracellular tyrosine kinase domain. T790M is a missense point mutation that substitutes threonine (T) with methionine (M) at position 790 [246]. Chiu et al. reported the crucial role of MYH9 in mediating the interaction between the drug-resistant EGFR-T790M mutation and β-actin. Disrupting the EGFR-T790M/MYH9/β-actin signaling axis enhances the therapeutic efficacy of CL-387,785 (a kinase inhibitor for EGFR-T790M) against drug-resistant NSCLC cells [214].
DT-13 is a bioactive saponin that regulates VEGF and PI3K/Akt signaling to inhibit angiogenesis and cell proliferation. It is a potent anti-metastatic compound because it inhibits the secretion of MMP-2/9 or tissue factor [247]. Under hypoxic conditions, NM IIA expression is inhibited, and its localization within lung cancer cells is altered, potentially contributing to enhanced metastatic behavior. However, treatment with DT-13 counteracts these effects by upregulating NM IIA expression and influencing its cellular distribution, thereby inhibiting metastasis [248].
Cancer-associated fibroblasts (CAFs) are activated fibroblasts that show significant heterogeneity and plasticity within the tumor microenvironment (TME) [249]. Du et al. employed a hypoxia-induced CAF model and an adipocyte model to study the impact of the TME on tumorigenesis. They observed that the conditioned medium obtained from the tumor microenvironment increased NM IIA expression and subsequent cancer cell migration [250]. Lung adenocarcinoma falls under the umbrella of NSCLC and usually originates from the mucosal glands. Microtubule-associated monooxygenase, calponin, and LIM domain containing 2 (MICAL2) is a monooxygenase that regulates cytoskeletal dynamics. Elevated levels of MICAL2 facilitate tumor development by stimulating cell proliferation and migration [251]. MYH9 transports molecules that interact with MICAL2 in the cytoplasm. As a transporter, MYH9 is important for MICAL2-mediated malignancy in patients with lung adenocarcinoma [215].

5.5. Hepatocellular Carcinoma (HCC)

Hepatocellular carcinoma (HCC) is strongly associated with viral hepatitis B/C and alcohol use [252]. MYH9 interacts with GSK3β, adenomatous polyposis coli (APC), and axis inhibition protein 1 (AXIN1) to induce the ubiquitination of β-catenin [216]. MYH9 forms a feedback loop with c-Jun, which transcriptionally stimulates MYH9 expression. Furthermore, hepatitis B virus X protein (HBX) can induce the expression of MYH9 via the modulation of GSK3β/β-catenin/c-Jun signaling [216]. Enkurin is an adaptor protein that localizes signal transduction machinery to calcium channels. In lung adenocarcinoma and colorectal cancer, Enkurin functions as a tumor suppressor [253,254]. In HCC, Enkurin interacts with MYH9 to inhibit the nuclear translocation of β-catenin. This interaction decreases the levels of both c-Jun and MYH9. Reduced levels of MYH9 inhibit cell proliferation and resistance to sorafenib [217].
In lung adenocarcinoma, MYH9 stabilizes c-Myc via deubiquitination with the help of the deubiquitinating enzyme ubiquitin-specific peptidase 7 (USP7) to promote cell cycle progression and EMT signaling [255]. NMHC IIA plays a critical role in HCC metastasis, where its phosphorylation at Ser1943 is dynamically regulated by bradykinin, leading to increased cell migration and invasion. NMHC IIA is integral to cytoskeletal dynamics and cellular contractility. The activation of TRPM7 by bradykinin results in the phosphorylation of the myosin IIA heavy chain and subsequent modulation of cell and focal adhesion dynamics [256,257]. Nucleosome assembly protein 1-like 5 (NAP1L5) is a histone chaperone that acts as a tumor suppressor. In HCC, NAP1L5 downregulates MYH9 which, in turn, inhibits PI3K/AKT/mTOR signaling, leading to its tumor-suppressive effects [258].

5.6. Pancreatic Cancer (PC)

Pancreatic cancer (PC) originates from pancreatic duct cells. Zhou et al. found that NM IIA expression is significantly high in PC tissue compared with normal tissue. NM II regulates β-catenin transcriptional activity to promote tumorigenesis in PC cells both in vitro and in vivo [221]. Pancreatic acinar cell carcinoma (PACC) is an uncommon malignant tumor characterized by cells resembling acinar cells and exhibiting exocrine enzyme production. It constitutes approximately 1–2% of all pancreatic neoplasms and is considered a high-grade malignancy [259]. 4H12 is a monoclonal antibody generated against Faraz-ICR, a PACC cell line, that specifically targets MYH9. Using immunohistochemical staining, the authors observed high MYH9 expression in acinar cell tumors, the source of Faraz-ICR cells. The 4H12 mAb suppressed Faraz-ICR cell proliferation in a dose-dependent manner [260].

5.7. Esophageal Cancer

Esophageal cancer is classified into squamous cell carcinoma (SCC) and adenocarcinoma (ADCA). NM IIA expression is significantly high in patients with esophageal squamous cell carcinoma (ESCC) and is associated with lymph node metastasis, an advanced tumor stage, and shorter overall survival. Low NM IIA expression in ESCC cells leads to increased cell–matrix adhesion and decreased cell migration [218]. Yang et al. used genomic sequencing data from 104 patients with ESCC and found that MYH9 levels were elevated in patients with ESCC with lymph node metastasis. A PCR array analysis revealed that MYH9 knockdown altered angiogenesis and EMT gene expression [219]. Protein tyrosine phosphatase 1B (PTP1B) dephosphorylates MYH9 at tyrosine 1408 (Y1408), leading to the upregulation of EGFR expression and subsequent proliferation [220].

5.8. Gastric Cancer (GC)

Gastric or stomach cancer (GC) originates in the stomach and is one of the most prevalent cancers worldwide. In GC, MYH9 plays a critical role in the context of peritoneal metastasis. Nuclear MYH9 promotes the transcription of the β-catenin gene (CTNNB1) to activate the Wnt/β-catenin pathway, which, in turn, confers resistance to the anoikis form of cell death. The inhibition of MYH9 phosphorylation, particularly at Ser1943, by compounds such as staurosporine, inhibits CTNNB1 gene transcription [222]. The downregulation of NM IIA using siRNA leads to the downregulation of the JNK signaling pathway, which promotes GC cell migration and invasion [223]. MYH9 plays an essential role as a downstream effector of the Enkurin pathway. Enkurin recruits the E3 ubiquitin ligase F-Box and WD Repeat Domain Containing 7 (FBXW7) enzyme to downregulate MYH9 levels [261]. Liang et al. reported that the miRNA let-7f (a member of the let-7 family of microRNAs) functions as a tumor suppressor by targeting MYH9. The overexpression of the miRNA let-7f downregulates MYH9 levels to decrease migration and invasion [262].
Hepatocellular carcinoma upregulated long non-coding RNA (HULC) is an oncogenic long non-coding RNA (lncRNA) that acts like a molecular sponge for miR-9-5p mRNA to regulate MYH9 expression. The dysregulation of the HULC/miR-9-5p/MYH9 axis contributes to GC progression. The knockdown of HULC inhibits tumor growth, which highlights the potential significance of targeting the HULC/miR-9-5p/MYH9 axis for GC treatment [263]. Similarly, circSLAMF6, a circular RNA, upregulates MYH9 expression by sponging miR-204-5p. The knockdown of MYH9 attenuates glycolysis and invasion, indicating its role in promoting cancer hallmarks under hypoxia. Moreover, in vivo experiments have shown that circSLAMF6 deficiency inhibits tumor growth [264].
miR-647, which is downregulated in GC, suppresses in vitro migration and invasion. miR-647 targets the serum response factor (SRF) mRNA, which then upregulates MYH9 transcription. Studies in orthotropic GC models have demonstrated that the overexpression of miR-647 inhibits metastasis, whereas increased SRF expression reverses this effect [265]. MYH9 is consistently overexpressed in the peritoneal metastasis of advanced GC. The siRNA-mediated knockdown of MYH9 alters the levels of EMT marker genes. Interference with S100A4, an oncogene implicated in metastasis, leads to MYH9 downregulation and inactivation of the Smad pathway, which participates in the EMT process. MYH9 is downstream of the S100A4 pathway and promotes EMT and metastasis [266].

5.9. Colorectal Cancer (CRC)

Colorectal cancer (CRC) ranks as the second most common cause of cancer-related deaths globally. Most CRC cases are adenocarcinomas, comprising more than 90% of cases, with less common subtypes including adenosquamous, spindle, squamous, and undifferentiated carcinomas. NM IIA activates the AMPK/mTOR pathway to promote tumorigenesis. NM IIA knockdown downregulates the expression of CD44 and CD133, indicating a role in regulating stemness. NM IIA protects CRC cells from 5-fluorouracil (5-FU)-induced apoptosis and prevents the inhibition of proliferation through the activation of the AMPK/mTOR pathway [267]. MYH9 promotes tumorigenesis in CRC by modulating MAPK/AKT signaling and influencing EMT, leading to aggressive cancer behavior [224].
Zhong et al. reported that MYH9 promotes CRC metastasis by interacting with autophagy-related protein 9B (ATG9B). MYH9 binds to ATG9B, enhancing its stability by preventing its ubiquitin-mediated degradation. The interaction between ATG9B and MYH9 stabilized both proteins by reducing their binding to the E3 ubiquitin ligase STIP1 Homology and U-Box Containing Protein 1 (STUB1), which would otherwise target both proteins for degradation. During cell invasion, MYH9 promotes the transport of ATG9B to the cell edge where it enhances focal adhesion and metastasis by activating integrin β1 [225]. LIM kinase 1 (LIMK1) upregulates both MYH9 and actinin-4 (ACTN4) to increase the aggressive phenotype associated with CRC [226]. TIMELESS (TIM), a circadian gene with evolutionary conservation, encodes a Timeless protein. It is important for facilitating effective DNA replication and maintaining genome integrity. TIMELESS interacts with MYH9 to promote its nuclear translocation and the subsequent activation of the β-catenin pathway, which promotes CRC tumorigenesis [268].
MYH9 binds to polymorphic adenoma-like protein 2 (PLAGL2), a downstream molecule regulated by miR-214-3p, to promote tumor development [269]. Circular MYH9 (circMYH9), an intron-derived circular RNA, plays a crucial role in CRC by promoting serine/glycine metabolism and reducing ROS generation to facilitate tumor growth [270]. In response to amino acid deprivation, circMYH9 expression is induced by hypoxia-inducible factor 1 subunit-α (HIF1α), which exacerbates CRC progression. Mouse studies have shown that circMYH9 overexpression promotes chemically induced carcinogenesis by suppressing p53 [270].

5.10. Breast Cancer

Breast cancer is among the most prevalent cancers in women, both in terms of newly diagnosed cases and mortality rates. S100A4 belongs to the S100 family and has been implicated in promoting metastasis in various tumors. High levels of S100A4 are correlated with a more aggressive cancer phenotype and poorer patient outcomes. S100A4 interacts with MYH9 to inhibit NM IIA filament formation and promote metastasis [271,272]. However, Wang et al. found that S100A1, another member of the S100 family, can inhibit the metastatic function of S100A4 [271]. S100P (member of S100 family) is upregulated in various cancer types [273]. S100P can form a heterodimer with S100A1 that weakens the binding affinity of S100P for MYH9 [274]. S100P interacts with NM IIA to promote cell migration. However, in cells that do not naturally express NM IIA or have low basal levels of NM IIA, S100P interacts with α, β-tubulin to promote cell migration [275].
Circular EIF6 (circ-EIF6) is an oncogenic circular RNA that encodes the novel peptide EIF6-224 [276]. EIF6-224 increases the level of MYH9 by inhibiting its degradation via the ubiquitin proteasome pathway. High MYH9 activates the Wnt/β-catenin signaling pathway to promote the metastasis of TNBC cells [276]. Furthermore, in TNBC cells, myosin IIA along with integrin β1 and MLCK contribute to adaptive resistance development against MEK inhibitors by mediating crosstalk between the Ras-ERK and PI3K-AKT signaling pathways [227]. In contrast, tissue factor pathway inhibitor 2 (TFPI-2), a tumor suppressor, binds to MYH9 and actinin-4 to inhibit the proliferation of breast cancer cells [277]. NM IIA modulates the biophysical characteristics of the ECM during cancer progression. In TNBC cells, the loss of NM IIA results in changes in the collagen stiffness of the constructs, which highlights its importance in efficient matrix remodeling during metastasis [228]. Derycke et al. highlighted the significance of NM IIA in breast cancer cell invasion into surrounding tissue in vitro [278].
Interferon-γ (IFN-γ) regulates the interaction between NM IIA and interferon-stimulated gene 15 (ISG15) in the cytoplasm of breast cancer cells [279]. Heat shock protein (HSP) 47 interacts with NM IIA via the unfolded protein response transducer IRE1α (encoded by the endoplasmic reticulum to nucleus signaling 1 (ERN1) gene) to enhance the metastatic potential of breast cancer cells [280]. Hepatitis B X-interacting protein (HBXIP) is an oncoprotein that interacts with the assembly-competent domain (ACD) of NMHC IIA. The phosphorylation of Ser1916 on the heavy chain of NM IIA by PKCβII further strengthens this interaction [281]. In HER2+ breast cancer cells, NM IIA levels increase upon treatment with the pan-HER inhibitor neratinib, suggesting its role in tumor growth. NM IIA modulation affects HER3 levels and downstream signaling, impacting cell growth and invasion [282]. DT-13 inhibits NM IIA and tumor microenvironment (TME)-induced breast cancer cell migration [229].

5.11. Renal Cell Carcinomas (RCCs)

Renal cell carcinomas (RCCs) are malignant tumors that originate from the renal cortex. Nuclear C-X-C motif chemokine receptor 4 (CXCR4) plays a key role in RCCs. NM IIA promotes RCC metastasis by facilitating the nuclear translocation of CXCR4 [230]. Clear cell renal cell carcinoma (ccRCC) is a predominant form of RCC, accounting for more than 90% of cases. MYH9 is upregulated in ccRCC and plays an important role in its development. Single-cell RNA sequencing (scRNA-seq) data show that MYH9 activates the AKT signaling pathway to promote ccRCC development. The MYH9/AKT signaling axis also confers resistance to sunitinib therapy in ccRCC [283].

5.12. Prostate Cancer

Prostate cancer is the second most common cause of death among men in the United States [231]. Samples from patients with prostate cancer show a high expression of human tubulin beta class IVa (TUBB4A), a member of the β-tubulin family. Cell migration, especially in narrow spaces, can induce genomic stress in prostate cancer cells. MYH9 interacts with TUBB4A to maintain genomic stability by protecting the nucleus against severe DNA damage [232].

5.13. Osteosarcoma

Osteosarcoma (OS) is a malignant bone tumor that originates from mesenchymal cells. MRPL23-AS1 is a lncRNA whose expression is increased in OS cell lines and tissues. Elevated levels of the lncRNA MRPL23-AS1 result in the increased expression of MYH9 via the competitive inhibition of miR-30b. Upregulated MYH9 promotes metastasis via the activation of β-catenin signaling [233].

5.14. Papillary Thyroid Carcinoma (PTC)

Papillary thyroid carcinoma (PTC) is an endocrine cancer that accounts for approximately 90% of all thyroid cancer cases. MYH9 enhances the stability of the cytokine receptor-like factor 1 (CRLF1) protein, which activates the ERK pathway. This activation leads to the upregulation of ETS variant transcription factor 4 (ETV4), which subsequently promotes the secretion of matrix metalloproteinase 1 (MMP1), facilitating PTC cell proliferation and metastasis [234].
In differentiated thyroid carcinoma (DTC), MYH9 contributes to radioresistance. Circular NIMA-related kinase 6 (NEK6) (circ_NEK6) promotes resistance to iodine 131(131I)-based radiotherapy by regulating MYH9 expression through the miR-370-3p/MYH9 axis. Upregulated MYH9 serves as a downstream effector of circ_NEK6 to promote proliferation and radioresistance [284]. MYH9 inhibits the activity of the bidirectional promoter shared by Forkhead Box E1 (FOXE1) and the lncRNA gene papillary thyroid carcinoma susceptibility candidate 2 (PTCSC2) [235].

5.15. Acute Myeloid Leukemia (AML)

Acute myeloid leukemia (AML) is characterized by impaired erythropoiesis and bone marrow dysfunction. AML cells show enhanced actomyosin contractility due to the increased expression of NM IIA and the hyperphosphorylation of the RLC [236].

5.16. Diffuse Large B Cell Lymphoma (DLBCL)

Diffuse large B cell lymphoma (DLBCL) is the most common form of high-grade non-Hodgkin’s lymphoma in the United States. Glycoprotein prostaglandin D2 synthase (PTGDS), a glycoprotein belonging to the lipocalin superfamily, has dual functions in prostaglandin metabolism and lipid transportation. PTGDS participates in multiple cellular processes, including the initiation of solid tumor formation. MYH9 interacts with PTGDS to promote the development of DLBCL via the activation of the Wnt-β-catenin-STAT3 pathway [285].

6. The Role of NM IIA as a Tumor Suppressor in Mice

6.1. Squamous Cell Carcinoma of the Skin

Squamous cell carcinoma is a common skin malignancy that has a precursor lesion and actinic keratosis. Schramek et al. were the first to report that suppressing MYH9 expression via knockdown or RNA interference led to the emergence of an aggressive form of HNSCC and skin squamous cell carcinoma in mice predisposed to tumors. In patients with squamous cell carcinoma, Schramek et al. found that the downregulation of the MYH9 gene is associated with poor survival. In human and mouse keratinocytes, MYH9 promotes the nuclear accumulation of p53 to enhance its post-transcriptional stability [202]. In mouse tongue squamous cell carcinoma, MYH9 is essential for maintaining mitotic stability during karyokinesis, and its deletion promotes carcinoma progression, indicating its role as a potential tumor suppressor. However, contrary to an earlier study by Schramek et al. who found that MYH9 stabilizes p53, this study suggests that the tumor suppressor activity of MYH9 is due to its role in maintaining mitotic stability during nuclear division [203].

6.2. Melanoma

Melanoma arises from the malignant transformation of melanocytes and accounts for approximately 4% of skin cancers. Despite its relatively low prevalence, melanoma is responsible for most skin cancer-related deaths, accounting for approximately 80% of mortality. MYH9 knockdown or silencing in melanoma cells promoted tumor growth and metastasis. The modulation of MYH9 altered the expression of oncogenes involved in EMT and the Erk signaling pathway. Therefore, in a melanoma mouse model, MYH9 may act as a potential tumor suppressor [204]. The deletion of both NM IIA and NM IIB isoforms in mammary epithelium cells results in increased proliferation in 3D culture, even under conditions that do not typically support tissue growth [286].

7. Dual Role of NM IIA in Humans

Coaxum et al. reported the tumor suppressor activity of MYH9 in human HNSCC. Low levels of MYH9 are associated with decreased survival in patients with HNSCC, particularly those harboring the low-risk mutant TP53 (mutp53). The inhibition of NM IIA increases the invasive ability of cells harboring wild-type p53 (wtp53), indicating its role in suppressing invasive behavior. However, in cells carrying mutant p53, MYH9 expression did not correlate with disease prognosis. NM II inhibition not only decreased the expression of p53 target genes but also the nuclear localization of wtp53. This may be due to the significance of MYH9 for the nuclear retention and function of wtp53 [201]. Coaxum and their colleagues focused on the interaction between MYH9 and p53 without diving into the functional significance of MYH9 in HNSCC [201]. In contrast to the results of Coaxum et al., You and colleagues reported the tumor-promoting role of MYH9 in HNSCC. MYH9 activates pan-MAPK signaling molecules to modulate the ECM and promote radioresistance [200]. The above studies indicate that MYH9 acts as a tumor suppressor in mouse HNSCC.

8. Role of NM IIA in Other Pathological Conditions

NM IIA-associated diseases can occur due to the presence of mutations, splicing errors, or the dysregulation of NM IIA function [28,287]. These mutations are commonly observed in the heavy chain of NM IIA and are referred to as MYH9-related diseases (MYH9-RD) (Figure 7). MYH9-RD (autosomal dominant) is characterized by the presence of congenital thrombocytopenia with giant thrombocytes and leucocyte inclusions. MYH9-RD results in a plethora of syndromes, such as May–Hegglin anomaly (MHA), Sebastian syndrome (SBS), Fechtner syndrome (FTNS), and Epstein syndrome (EPS) [288,289,290,291]. Additional symptoms may include hearing loss, early-onset cataracts, increased liver enzyme levels, and progressive kidney damage that may ultimately result in end-stage renal disease (ESRD) [292].
The molecular mechanism underlying MYH9-RD involves specific mutations in the MYH9 gene that disrupt filament assembly, alter ATPase activity, and impair the actin-binding ability of NM IIA [293]. Structural and functional defects in NM IIA lead to the development of macrothrombocytopenia, hearing loss, renal failure, and cataracts [293]. Pecci et al. analyzed 108 patients with MYH9-RD from 50 unrelated families and observed that patients with mutations in the heavy chain motor domain had severe thrombocytopenia, nephritis, and deafness [294].
Various genetic alterations, including nonsense and frameshift mutations of the heavy chain, duplications, and in-frame deletions, can lead to MYH9-RD [295,296,297,298]. NM IIA is the only paralog expressed in platelets. It is important for the differentiation and maturation of megakaryocytes, which are precursors of mature thrombocytes. The current model of platelet formation establishes that megakaryocytes terminally differentiate and release platelets from cytoplasmic extensions called proplatelets. Mutations such as R702C, D1424N, and E1841K disrupt proplatelet formation, leading to the formation of giant proplatelets. MYH9 mutations impair the migration of megakaryocytes within the bone marrow, preventing proper movement toward the vasculature for platelet release [299,300,301,302,303]. Mutations can also alter the cytoskeleton dynamics of platelets [304]. Platelet adhesion is impacted by a decrease in glycoprotein 1b/IX/V in large platelets in patients with MYH9-RD, which could contribute to their bleeding tendencies [305].
May–Hegglin anomaly (MHA), Sebastian syndrome (SBS), Fechtner syndrome (FTNS), and Epstein syndrome (EPS) are autosomal dominant diseases associated with macrothrombocytopenia, sensorineural hearing loss, cataracts, and nephritis. Heath et al. evaluated 27 patients and found MYH9 mutations in 74%. R702C and R702H mutations are associated with FTNS, EPS, and Alport syndrome with macrothrombocytopenia (APSM) [306]. Seri et al. evaluated 19 families and reported an abnormal NMHC IIA distribution in leukocytes. A clinical evaluation revealed high-tone hearing loss in 83% of patients and cataracts in 23% of patients initially diagnosed with MHA or SBS [307]. Several studies have reported extra hemorrhagic symptoms in patients with MYH9-RD. Lalwani et al. reported the presence of the guanine (G) > adenine (A) mutation at nucleotide 2114, leading to an R705H missense mutation, which is associated with autosomal dominant hearing impairment [290]. MYH9 mutations can lead to the development of nephropathy [308,309]. Elevated liver enzymes are frequently found in patients with MYH9-RD but do not appear to cause significant structural damage [310].
Data from 36 studies using whole-exome sequencing (WES) and whole-genome sequencing (WGS) revealed 17,104 de novo mutations across four psychiatric disorders: autism spectrum disorder, epileptic encephalopathy, intellectual disability, and schizophrenia. MYH9 mutations were observed in patients with schizophrenia, intellectual disability, and autism [311]. Brain tissues from patients with schizophrenia show elevated levels of phosphorylated RLCs. In amyotrophic lateral sclerosis (ALS), miR-155, which is involved in the regulation of NM II, is upregulated, contributing to disease progression. Similarly, in multiple sclerosis (MS), miR-155 promotes inflammatory responses [312,313,314].
Aberrant NM IIA expression or activity is associated with microbial infections. The genetic ablation of NM II using shRNA or pharmacological inhibition impairs the dissemination of microbes [315]. NM IIA is critical for the host response to bacterial infection, particularly Listeria monocytogenes infection. The phosphorylation of NM IIA at the tyrosine-158 residue limits the intracellular level of L. monocytogenes. This phosphorylation is mediated via the tyrosine kinase Src and occurs near the ATP-binding side of the motor head domain [316]. Kaposi’s sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus-8 (HHV-8), targets NM IIA to induce its phosphorylation, which results in blebbing and macropinocytosis during viral infection [317]. NM IIA is important in the defense against bacterial pore-forming toxins (PFTs), such as listeriolysin O (LLO). Following PFT-induced damage, NM IIA coordinates with the ER chaperone Gp96 to regulate cytoskeletal dynamics, membrane blebbing, and cell migration [318,319]. Similarly, perfringolysin O (PFO), which is a pore-forming toxin produced by Clostridium perfringens, induces a significant reorganization of the actomyosin cytoskeleton [188].

9. Targeting MYH9

The small molecule myosin inhibitor blebbistatin was identified via a screen targeting NM IIA. It acts as a non-competitive inhibitor and stabilizes the metastable state of myosin which precedes the force-generating step catalyzed by ATP hydrolysis [320]. Limouze et al. evaluated the specificity and potency of blebbistatin across various members of the myosin superfamily [321]. With an IC50 value between 0.5 and 5 μM, it showed a potent inhibition of various striated muscle myosin and vertebrates NM IIA and NM IIB. It showed weak inhibitory activity against smooth muscle myosin with an IC50 value of 80 μM [321]. Despite its broad inhibitory effect across myosin isoforms, it has several limitations, including fluorescence, poor water solubility, cytotoxicity, and sensitivity to photodegradation [322].
Several drugs that modulate MYH9 activity are listed in Table 3. Cinobufotalin, a natural compound isolated from toad venom, has cardiotonic, diuretic, and potential cytotoxic effects. In nasopharyngeal carcinoma, cinobufotalin increases the levels of MAP2K4 to reduce cancer stemness, EMT, and resistance to cisplatin [207]. Furthermore, in HCC and lung cancer, cinobufotalin exerts an anti-tumor action by decreasing the levels of MYH9 [217,255]. Apatinib is a small molecule TKI that inhibits angiogenesis by blocking the vascular endothelial growth factor (VEGF)-induced phosphorylation of the VEGF Receptor-2 (VEGFR2) pathway. In glioma cells, apatinib targets thrombospondin 1 (THBS1) to inhibit malignancy [205]. Disulfiram is an FDA-approved drug for treating alcoholism. Robinson et al. screened approximately 3185 compounds and identified disulfiram (IC50 value of 300 nM) as a potent inhibitor of growth in TNBC cells. Disulfiram directly targets MYH9 and the Ras GTPase-activating-like protein IQGAP1 to inhibit growth. The combination of disulfiram and doxorubicin enhanced the killing of cancer stem cells by inducing cellular senescence [323]. C70-EDA is an aminated fullerene derivative that binds to the C terminal tail domain and modulates the subcellular distribution of MYH9 to inhibit the NM IIA filament [324]. J13 is a small molecule that disrupts the interaction of MYH9 with actin to increase mitochondrial fission. This study also revealed that heat shock protein A9 (HSPA9), a key player affected by MYH9-actin dysfunction, is crucial for modulating mitochondrial fission [325].
In gastric cancer, staurosporine decreases the activity of casein kinase II, leading to the reduced phosphorylation of MYH9 at the Ser1943 residue. This results in the downregulation of CTNNB1 gene transcription with the decreased activation of β-catenin signaling [222]. Homoharringtonine (HHT) is a natural alkaloid that upregulates MYH9 expression in acute and chronic myeloid leukemia. High MYH9 levels enhance the sensitivity of leukemia cell lines to HHT-induced cell death [326]. MYH9 is also involved in cisplatin-induced kidney injury. MYH9 interacts with upregulated apurinic/apyrimidinic endonuclease 2 (APE2) in proximal tubule cells to induce mitochondrial damage and kidney injury [341].

Targeting the ROCK-Myosin II Signaling Pathway

Rho GTPases are molecular switches that regulate cytoskeleton dynamics [342]. Rho GTPase enzymes are normally inactive when bound to GDP; however, upon activation, they are bound to GTP to activate downstream signaling molecules such as Rho-associated protein kinases 1 and 2 (ROCK1 and ROCK2) [342,343]. ROCK enzymes are serine/threonine AGC kinases that can be activated by other mechanisms, including mechanical stress, plasma membrane binding [344], and proteolytic cleavage by caspases and granzyme B [343]. They have a molecular weight of approximately 160 kDa and differ in their expression levels across tissues [345]. These enzymes participate in multiple processes, including cell migration, differentiation, apoptosis, and immune responses [346].
Myosin light chain 2 (MLC2) is a key player in the myosin II pathway. ROCKs phosphorylate MLC2, which then binds with the heavy chain of myosin (MHC) to promote actomyosin contraction [347,348,349,350]. Myosin phosphatase (MYPT) dephosphorylates myosin II and reduces its motor activity. However, ROCK can phosphorylate MYPT to reduce its phosphatase activity, which ultimately enhances myosin II motor activity [343]. LIM kinases are actin-binding kinases that are important regulators of actin polymerization and microtubule assembly. ROCK can activate LIM kinases, which prevent the disassembly of actin filaments [351]. Other poorly characterized downstream effectors of ROCK enzymes include elongation factor-1α (EF-1α), adducin, and calponin [352].
Studies have shown that some tumors proliferate and undergo transformation via the Rho–ROCK–myosin II pathway [353,354]. However, other tumors may use the ROCK–myosin II pathway for migration and EMT [347,355]. The ROCK–myosin II pathway also contributes to treatment resistance [349], the modulation of immune responses [350], and ECM regulation [356,357]. In cancer-associated fibroblasts (CAFs), ROCK enzymes regulate actomyosin dynamics to promote cancer cell migration through the ECM [358]. Wong et al. reported that C-C motif chemokine ligand 2 (CCL2) secretion by pericytes increases tumor growth via the activation of the ROCK2–myosin II pathway [359]. The ROCK–myosin II signaling pathway enables ATPase-driven actomyosin contraction. ROCK inhibitors inhibit ROCK activity and modify the ROCK–myosin II pathway, which can be beneficial in treating diseases such as glaucoma, cancer, renal failure, and fibrosis. Since ROCK enzymes regulate axon regeneration, ROCK inhibitors may offer some advantages in treating neurological disorders [327,360].
Currently, four different ROCK inhibitors are clinically approved. Fasudil is approved to treat symptoms of cerebral ischemia [327]. The overactivation of the ROCK signaling pathway contributes to inflammation and neuronal damage in neurodegenerative diseases [361]. Fasudil was the first clinically used ROCK inhibitor for neurodegenerative diseases. Fasudil repressed neuroinflammation and decreased the secretion of inflammatory factors by inhibiting CD4+ T-cell differentiation and phagocytes, thus restoring the balance of immune cells in vitro [362,363]. Fasudil also has a neuroprotective effect by reducing neuronal apoptosis due to the overexpression of ROCK in primary mouse hippocampal neurons [364,365,366]. The combination of fasudil with gemcitabine increased the uptake and efficacy of gemcitabine in PDAC [367]. The early treatment of pancreatic cancer with fasudil remodeled the metastatic niche by increasing blood vessel formation and reducing collagen deposition [368]. Hypoxia, a hallmark of many tumors, is involved in metastatic transition. The fasudil prodrug, which remains inactive under normoxia but becomes active under hypoxic conditions, can inhibit ROCK activity without inducing hypotension [369]. However, the use of fasudil is limited due to its toxicity, non-specificity for other kinases, and poor oral bioavailability.
In Japan, ripasudil is approved for treating patients with open-angle glaucoma and ocular hypertension [328]. Ripasudil, when combined with a programmed death-1/programmed death ligand-1 (PD-1/PD-L1) checkpoint inhibitor, recruits CD8+ T cells to increase the immune response in patients with uveal melanoma [370]. Netarsudil is a pan-ROCK inhibitor approved in both the EU and the USA for patients with open-angle glaucoma with raised intraocular pressure [329]. Belumosudil is a specific ROCK2 inhibitor approved in the USA for patients with chronic graft versus host disease (GVHD) who fail to respond to first-line therapy [330,331].
Y27632 is an orally active ROCK inhibitor that targets other kinases, including PKA and PKC [332]. Y27632 increases the effectiveness of doxorubicin in vivo and reduces the tumor size by inducing an anti-tumor immune response [371]. In multiple myeloma, Federico et al. used Y27632 to improve bortezomib efficacy via the disruption of the bone marrow microenvironment [372]. In melanoma, tumors resistant to BRAF inhibitors show a strong activation of Rho/ROCK signaling, which is linked to increased dedifferentiation. A machine learning analysis using Y27632 or fasudil suggested that poorly differentiated tumors are more sensitive to ROCK inhibition. Furthermore, both Y27632 and fasudil were able to restore the efficacy of BRAF inhibitors in resistant melanoma cells [373]. ROCK stabilizes the PD-L1 protein via moesin (MSN) phosphorylation, which is linked to poor prognosis in patients with breast cancer [374].
Studies have shown that silencing ROCK1 via genetic modulation or inhibition with GSK269962A (a selective ROCK1 inhibitor) enhances the effectiveness of BRAF and MAPK inhibitors against BRAF and NRAS mutant melanoma cells [333]. Furthermore, in vivo studies using a combination of ROCK inhibitors (GSK269962A or fasudil) with MEK or ERK inhibitors suppressed NRAS mutant melanoma growth [375]. The combination of EGFR and ROCK inhibitors has a synergistic effect and significantly reduces the growth of TNBC cells via cell cycle arrest [376]. A mass spectrometry-based proteomic approach revealed the mechanisms involved in the co-inhibition of EGFR and ROCK. EGFR inhibition alone induces autophagy, but combining it with GSK269962A impairs autophagosome clearance [377]. AT13148 is a ROCK/AKT inhibitor that has shown positive results in preclinical cancer models but has failed in clinical trials. A poor pharmacokinetic profile combined with a narrow therapeutic index led to the termination of the trial [334].
BDP5290 is a potent inhibitor of MRCK (myotonic dystrophy kinase-related CDC42-binding kinase), which is important for actomyosin contractility and metastasis. BDP5290 binds to the kinase domain of MRCK and blocks MLC phosphorylation to inhibit the invasion of breast cancer cells [335]. Using an RNAi screen, Castoreno et al. identified Rhodblock 6 as a Rho kinase inhibitor. It disrupts the correct localization of phosphorylated MLC during cell division [336]. RKI-1447 binds to the ATP-binding site of the ROCK enzyme to inhibit the binding of MLC-2 and MYPT-1 without affecting other kinases, such as AKT, MEK, and S6 kinase. It suppresses ROCK-mediated cytoskeletal reorganization, specifically inhibiting actin stress fiber formation [337]. Similarly, RKI-18 is also a potent Rho-kinase inhibitor with significant anti-tumor properties. It inhibits the phosphorylation of ROCK and MLC2, leading to diminished lamellipodia and filopodium formation in breast cancer cells [338].
Novel thiosemicarbazone iron chelators, such as di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone, have shown potential in cancer therapy by inhibiting metastasis. These chelators work by inducing cellular iron depletion, which upregulates the expression of metastasis suppressor N-Myc downstream regulated 1 (NDRG1). NDRG1 subsequently inhibits the ROCK1/p-MLC2 pathway to reduce migration and stress fiber formation [339]. Combretastatin (CA-4) and its analog CA-432 are potent anti-tumor agents that activate the RhoA/ROCK signaling pathway to increase the phosphorylation of MLC. Furthermore, these agents inhibit T-cell migration and chemotaxis by destabilizing microtubules, decreasing tubulin acetylation and microtubule stability [340].

10. Conclusion and Perspectives

NM IIA is a vital component of the actomyosin cytoskeleton that controls cell division and migration. It is important in early embryonic development for the formation of functional visceral endoderm and the maintenance of cell polarity. NM IIA is expressed not only in the cytoplasm but also in the cell membrane and nucleus. NM IIA activity is regulated at the levels of protein folding, filament assembly, and Mg2+-ATPase motor activity. NM IIA acts as a tumor promoter or a suppressor based on the tumor type and molecular environment. For example, NM IIA is reported to be a tumor suppressor in melanoma and squamous cell carcinoma. However, the main drawback of these studies is the use of mouse models instead of human cell lines or patient data. In contrast, numerous studies using human cancer cells and clinical data have reported the tumor-promoting role of NM IIA.
NM IIA also plays a significant role in promoting cancer cell migration, invasion, and metastasis; maintaining stemness; and regulating metabolism. Furthermore, it also inhibits apoptosis and autophagy and contributes to the development of resistance to chemoradiotherapy and targeted therapy by regulating the MAPK, PI3K/Akt, and Wnt/β-catenin signaling pathways. Therefore, because of the dual nature (as a tumor promoter and suppressor) and complexity of NM IIA’s function, it is necessary to further evaluate its molecular interactions and regulatory mechanisms using new technologies. Apart from its role in cancer, NM IIA is also involved in other pathological conditions such as renal disease, severe thrombocytopenia, and various neuropsychiatric and neuroinflammatory disorders. In this review, we discussed the significant role of NM IIA in tumor development and the pathogenesis of other diseases, underscoring its importance as a key therapeutic target. However, there are currently very few drugs or chemical agents targeting NM IIA, and the majority of them are still being studied in preclinical models. For example, blebbistatin, which inhibits NM IIA motor activity, is poorly water soluble, cytotoxic, phototoxic, and unstable with limited specificity. The mechanism of action and efficacy of some drugs, such as disulfiram and C70 -EDA, are not well understood.
ROCK enzymes are important for the regulation and activation of NM IIA. Fasudil, ripasudil, netarsudil, and nelumosudil are currently approved for targeting the ROCK pathway. Many preclinical cancer studies have demonstrated the significance of ROCK inhibitors, but there is one clinical trial targeting the ROCK-AKT pathway in advanced solid tumors. There are many drawbacks of using ROCK inhibitors to target the ROCK–myosin II pathway: the activation of NM IIA via other mechanisms, the systemic toxicity of ROCK inhibitors, and the lack of isoform-specific ROCK inhibitors. Although the ROCK–myosin II pathway is important for tumorigenesis, researchers have not been able to translate these preclinical findings to clinical applications. Currently, many researchers are exploring the potential of NM IIA as a therapeutic target in tumors and other diseases.

Author Contributions

Conceptualization, W.F., J.T.G., B.S.P., and M.S.; data curation, W.F., B.S.P., and M.K.K.; writing— W.F., J.T.G., B.S.P., N.N., and M.S.; Figures— A.M.A.S., M.K.K., and R.M.; supervision, J.T.G.; project administration, J.T.G.; funding acquisition, J.T.G. All authors have read and approved the final version of the manuscript.

Funding

This work was supported by grant from the University of Cincinnati Cancer Center (UCCC) Pilot Project Award Program: F101484-6264102002-1-S32335.

Institutional Review Board Statement

This manuscript is a review article and does not involve human or animal experiments. Therefore, ethics committee approval or consent was not required to participate.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data for this review were collected through PubMed. The following search terms were used: non-muscle myosin IIA; NM IIA; MYH9; tumor; cancer; MYH9-RD; myosin II; MYH9 mutations; oncogene; tumor suppressor; and actomyosin. Only articles published in English were included in the literature review. All relevant data are incorporated into the manuscript.

Acknowledgments

We would like to thank the University of Cincinnati for providing the necessary facilities for this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

MYH9Myosin heavy chain 9
MYH10Myosin heavy chain 10
MYH14Myosin heavy chain 14
NM IIANon-muscle myosin IIA
NMHC IIANon-muscle myosin IIA heavy chain
NMHC IIBNon-muscle myosin IIB heavy chain
NMHC IICNon-muscle myosin IIC heavy chain
MHAMay–Hegglin anomaly
EPSEpstein syndrome
FTSFechtner syndrome
SPSSebastian platelet syndrome
ELCEssential light chain
RLCRegulatory light chain
MHCMyosin heavy chain
MLCMyosin light chain
MHC IIAMyosin heavy chain IIA
ROCKRho-associated protein kinase
ORFOpen reading frame
MYL6Myosin light chain 6
MYL6BMyosin light chain 6B
MYL 12AMyosin light chain 12A
MYL 12BMyosin light chain 12B
MLCKMyosin light chain kinase
MyBP-CMyosin-binding protein C
MyBP-HMyosin-binding protein H
PKAProtein kinase A
PKCProtein kinase C
PKCβProtein kinase Cβ
TRPM7Transient receptor potential melastatin 7
CK IICasein kinase II
MRCKMyotonic dystrophy kinase-related CDC42-binding kinase
MYPT1Myosin phosphatase target subunit 1
PDGFPlatelet-derived growth factor
PAK1p21-activated kinase 1
DAPKDeath-associated protein kinase
NHTNon-helical tail
EMTEpithelial-to-mesenchymal transition
TGF-βTransforming growth factor-β
EGFEpidermal growth factor
LglLethal (2) giant larvae
aPKCζAtypical protein kinase C
LIMCH1LIM and calponin-homology domains 1
MYBPHLMyosin-binding protein H-like
Tpm4.2Tropomyosin 4.2
NMY-2Non-muscle myosin II heavy chain in C. elegans
ECMExtracellular matrix
MyoGEFMyosin-interacting guanine nucleotide exchange factor
GSISGlucose-stimulated insulin secretion
APPL1Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1
PP2AProtein phosphatase 2A
FAKFocal adhesion kinase
MHBMidbrain–hindbrain boundary
HNSCCHead and neck squamous cell carcinoma
NEDD9Neural precursor cell expressed, developmentally downregulated 9
CASCrk-associated substrate
MMPsMatrix metalloproteinases
HMGA1High mobility group AT-jook 1
PDOXPatient-derived orthotopic xenograft
GBMGlioblastoma
THBS1Thrombospondin 1
VEGFAVascular endothelial growth factor A
HIF-1αHypoxia-inducible factor-1α
NPCNasopharyngeal carcinoma
EBVEpstein–Barr virus
BARTBamHI A rightward transcript
FKHRForkhead in rhabdomyosarcoma
FoxOForkhead box O-class
TRAF6TNF receptor-associated factor 6
GSK3βGlycogen synthase kinase-3β
FNDC3BFibronectin type III domain containing 3B
NSCLCNon-small cell lung cancer
CSCCancer stem cell
LCCsLung cancer cells
EGFREpidermal growth factor receptor
TKIsTyrosine kinase inhibitors
TMETumor microenvironment
CAFsCancer-associated fibroblasts
MICAL2Microtubule-associated monooxygenase, calponin, and LIM domain containing 2
HCCHepatocellular carcinoma
APCAdenomatous polyposis coli
AXIN1Axis inhibition protein 1
HBXHepatitis B virus X protein
TRPCsCanonical transient receptor potential channels
NAP1L5Nucleosome assembly protein 1-like 5
PCPancreatic cancer
GEOGene expression omnibus
PACCPancreatic acinar cell carcinoma
SCCSquamous cell carcinoma
ADCAAdenocarcinoma
NGSNext-generation sequencing
ESCCEsophageal squamous cell carcinoma
PTP1BProtein tyrosine phosphatase 1B
GCGastric cancer
FBXW7F-Box and WD repeat domain containing 7
USP2Ubiquitin-specific peptidase 2
HULCHepatocellular carcinoma upregulated long non-coding RNA
SLAMF6SLAM family member 6
SRFSerum response factor
S100A4S100 calcium binding protein A4
S100A1S100 calcium binding protein A1
S100PS100 calcium binding protein P
CRCColorectal cancer
AMPKAMP-activated protein kinase
mTORMechanistic target of rapamycin kinase
MAPKMitogen-activated protein kinase
AKTAKT serine/threonine kinase
ATG9BAutophagy-related protein 9B
STUB1STIP1 homology and U-Box containing protein 1
ACTN4Actinin-4
LIMK1LIM kinase 1
TIMELESSTimeless circadian regulator
CRYCryptochrome proteins
PERPeriod proteins
PLAGL2Polymorphic adenoma-like protein 2
HNRNPA2B1Heterogeneous nuclear ribonucleoprotein A2/B1
HIF1αHypoxia inducible factor 1 subunit-α
EIF6Eukaryotic translation initiation factor 6
TFPI-2Tissue factor pathway inhibitor 2
ISG15Interferon-stimulated gene 15
IFN-γInterferon γ
HSPHeat shock protein
ERN1Endoplasmic reticulum to nucleus signaling 1
HBXIPHepatitis B X-interacting protein
ACDAssembly-competent domain
PKCβIIProtein kinase C βII
HER2Human epidermal growth factor receptor 2
HER3Human epidermal growth factor receptor 3
RCCRenal cell carcinoma
CXCR4C-X-C motif chemokine receptor 4
ccRCCClear cell renal cell carcinoma
TUBB4ATubulin beta class IVA
OSOsteosarcoma
lncRNALong non-coding RNA
PTCPapillary thyroid carcinoma
CRLF1Cytokine receptor-like factor 1
ETV4ETS variant transcription factor 4
MMP1Matrix metalloproteinase 1
DTCDifferentiated thyroid carcinoma
NEK6NIMA-related kinase 6
FOXE1Forkhead Box E1
PTCSC2Papillary thyroid carcinoma susceptibility candidate 2
AMLAcute myeloid leukemia
CMLChronic myeloid leukemia
DLBCLDiffuse large B cell lymphoma
PTGDSGlycoprotein prostaglandin D2 synthase
ROSReactive oxygen species
MYH9-RDMYH9-related diseases
SBSSebastian syndrome
FTNSFechtner syndrome
ESRDEnd-stage renal disease
WESWhole-exome sequencing
ALSAmyotrophic lateral sclerosis
MSMultiple sclerosis
shRNAShort hairpin RNA
KSHVKaposi’s sarcoma-associated herpesvirus
HHV-8Human herpesvirus 8
PFTsPore-forming toxins
LLOListeriolysin O
PFOPerfringolysin O
VEGFVascular endothelial growth factor
VEGFR2Vascular endothelial growth factor receptor-2
IQGAP1IQ motif containing GTPase activating protein 1
TNBCTriple-negative breast cancer
DARTSDrug affinity responsive target stability
HSPA9Heat shock protein A9
HHTHomoharringtonine
APE2Apurinic/apyrimidinic endonuclease 2
MLC2Myosin light chain 2
EF-1αElongation factor 1 α
SLC9A1/NHE1Solute carrier family 9 member A1
TAMsTumor-associated macrophages
CCL2C-C motif chemokine ligand 2
PD-1/PD-L1Programmed death-1/programmed death ligand-1
GVHDGraft versus host disease
MSNMoesin
NDRG1N-Myc downstream regulated 1
CA-4Combretastatin

References

  1. Fletcher, D.A.; Mullins, R.D. Cell mechanics and the cytoskeleton. Nature 2010, 463, 485–492. [Google Scholar] [CrossRef]
  2. 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]
  3. 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]
  4. Richards, T.A.; Cavalier-Smith, T. Myosin domain evolution and the primary divergence of eukaryotes. Nature 2005, 436, 1113–1118. [Google Scholar] [CrossRef]
  5. 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]
  6. 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]
  7. 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]
  8. 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]
  9. 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]
  10. Heissler, S.M.; Sellers, J.R. Kinetic Adaptations of Myosins for Their Diverse Cellular Functions. Traffic 2016, 17, 839–859. [Google Scholar] [CrossRef]
  11. 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]
  12. 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]
  13. Brown, S.S. Myosins in yeast. Curr. Opin. Cell Biol. 1997, 9, 44–48. [Google Scholar] [CrossRef] [PubMed]
  14. 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]
  15. 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]
  16. 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]
  17. 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]
  18. 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]
  19. 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]
  20. 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]
  21. Li, Y.; Lalwani, A.K.; Mhatre, A.N. Alternative Splice Variants of MYH9. DNA Cell Biol. 2008, 27, 117–125. [Google Scholar] [CrossRef]
  22. 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]
  23. 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]
  24. 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]
  25. 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]
  26. 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]
  27. 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]
  28. 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]
  29. 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]
  30. 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]
  31. 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]
  32. 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]
  33. 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]
  34. Sellers, J.R. Myosins: A diverse superfamily. Biochim. Biophys. Acta 2000, 1496, 3–22. [Google Scholar] [CrossRef] [PubMed]
  35. Cheney, R.E.; Mooseker, M.S. Unconventional myosins. Curr. Opin. Cell Biol. 1992, 4, 27–35. [Google Scholar] [CrossRef]
  36. 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]
  37. Heissler, S.M.; Sellers, J.R. Myosin light chains: Teaching old dogs new tricks. BioArchitecture 2014, 4, 169–188. [Google Scholar] [CrossRef]
  38. 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]
  39. 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]
  40. Craig, R.; Woodhead, J.L. Structure and function of myosin filaments. Curr. Opin. Struct. Biol. 2006, 16, 204–212. [Google Scholar] [CrossRef]
  41. 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]
  42. 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]
  43. 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]
  44. 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]
  45. 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]
  46. 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]
  47. Coluccio, L.M. Myosins; Springer: New York, NY, USA, 2012; pp. 1177–1182. [Google Scholar]
  48. Heissler, S.M.; Manstein, D.J. Nonmuscle myosin-2: Mix and match. Cell. Mol. Life Sci. 2013, 70, 1–21. [Google Scholar] [CrossRef]
  49. 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]
  50. 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]
  51. 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]
  52. 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]
  53. 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]
  54. 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]
  55. 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]
  56. 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]
  57. 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]
  58. 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]
  59. 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]
  60. 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]
  61. 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]
  62. Adelstein, R.S.; Conti, M.A. Phosphorylation of platelet myosin increases actin-activated myosin ATPase activity. Nature 1975, 256, 597–598. [Google Scholar] [CrossRef]
  63. 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]
  64. 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]
  65. 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]
  66. 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]
  67. 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]
  68. 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]
  69. 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]
  70. 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]
  71. Ikebe, M.; Reardon, S. Phosphorylation of bovine platelet myosin by protein kinase C. Biochemistry 1990, 29, 2713–2720. [Google Scholar] [CrossRef]
  72. 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]
  73. 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]
  74. 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]
  75. 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]
  76. 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]
  77. 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]
  78. 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]
  79. 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]
  80. 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]
  81. 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]
  82. Dulyaninova, N.G.; Bresnick, A.R. The heavy chain has its day. BioArchitecture 2013, 3, 77–85. [Google Scholar] [CrossRef]
  83. 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]
  84. 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]
  85. 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]
  86. 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]
  87. 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]
  88. 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]
  89. 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]
  90. 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]
  91. 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]
  92. 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]
  93. 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]
  94. 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]
  95. 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]
  96. 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]
  97. 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]
  98. 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]
  99. 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]
  100. 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]
  101. 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]
  102. 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]
  103. 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]
  104. 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]
  105. 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]
  106. 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]
  107. 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]
  108. 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]
  109. Ravid, S. The tumor suppressor Lgl1 regulates front-rear polarity of migrating cells. Cell Adhes. Migr. 2014, 8, 378–383. [Google Scholar] [CrossRef]
  110. 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]
  111. 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]
  112. 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]
  113. 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]
  114. 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]
  115. 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]
  116. 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]
  117. 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]
  118. 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]
  119. 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]
  120. 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]
  121. 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]
  122. Guo, S.; Kemphues, K.J. A non-muscle myosin required for embryonic polarity in Caenorhabditis elegans. Nature 1996, 382, 455–458. [Google Scholar] [CrossRef]
  123. 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]
  124. 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]
  125. 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]
  126. 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]
  127. 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]
  128. 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]
  129. 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]
  130. 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]
  131. 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]
  132. 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]
  133. Green, R.A.; Paluch, E.; Oegema, K. Cytokinesis in animal cells. Annu. Rev. Cell Dev. Biol. 2012, 28, 29–58. [Google Scholar] [CrossRef] [PubMed]
  134. 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]
  135. 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]
  136. 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]
  137. 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]
  138. 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]
  139. 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]
  140. Sun, Z.; Guo, S.S.; Fässler, R. Integrin-mediated mechanotransduction. J. Cell Biol. 2016, 215, 445–456. [Google Scholar] [CrossRef]
  141. 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]
  142. 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]
  143. 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]
  144. 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]
  145. 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]
  146. 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]
  147. 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]
  148. Ladoux, B.; Mège, R.-M. Mechanobiology of collective cell behaviours. Nat. Rev. Mol. Cell Biol. 2017, 18, 743–757. [Google Scholar] [CrossRef]
  149. 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]
  150. 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]
  151. 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]
  152. 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]
  153. 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]
  154. 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]
  155. 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]
  156. 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]
  157. 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]
  158. 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]
  159. 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]
  160. 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]
  161. 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]
  162. 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]
  163. 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]
  164. 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]
  165. 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]
  166. 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]
  167. 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]
  168. 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]
  169. 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]
  170. Yoshida, K.; Soldati, T. Dissection of amoeboid movement into two mechanically distinct modes. J. Cell Sci. 2006, 119, 3833–3844. [Google Scholar] [CrossRef]
  171. 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]
  172. 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]
  173. 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]
  174. Charras, G.T. A short history of blebbing. J. Microsc. 2008, 231, 466–478. [Google Scholar] [CrossRef] [PubMed]
  175. Fackler, O.T.; Grosse, R. Cell motility through plasma membrane blebbing. J. Cell Biol. 2008, 181, 879–884. [Google Scholar] [CrossRef] [PubMed]
  176. Rottner, K.; Schaks, M. Assembling actin filaments for protrusion. Curr. Opin. Cell Biol. 2019, 56, 53–63. [Google Scholar] [CrossRef]
  177. 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]
  178. 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]
  179. Ikenouchi, J.; Aoki, K. Membrane bleb: A seesaw game of two small GTPases. Small GTPases 2017, 8, 85–89. [Google Scholar] [CrossRef]
  180. 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]
  181. Ridley, A. Blebs on the move. Dev. Cell 2011, 20, e1. [Google Scholar] [CrossRef]
  182. 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]
  183. 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]
  184. 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]
  185. 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]
  186. 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]
  187. Taneja, N.; Burnette, D.T. Myosin IIA drives membrane bleb retraction. Mol. Biol. Cell 2019, 30, 1051–1059. [Google Scholar] [CrossRef]
  188. 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]
  189. 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]
  190. 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]
  191. 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]
  192. 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]
  193. 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]
  194. 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]
  195. 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]
  196. 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]
  197. 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]
  198. 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]
  199. 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]
  200. 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]
  201. 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]
  202. 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]
  203. 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]
  204. 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]
  205. 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]
  206. 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]
  207. 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]
  208. Lu, H.; Huang, H. FOXO1: A Potential Target for Human Diseases. Curr. Drug Targets 2011, 12, 1235–1244. [Google Scholar] [CrossRef]
  209. 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]
  210. 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]
  211. 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]
  212. 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]
  213. 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]
  214. 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]
  215. 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]
  216. 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]
  217. 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]
  218. 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]
  219. 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]
  220. 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]
  221. 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]
  222. 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]
  223. 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]
  224. 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]
  225. 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]
  226. 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]
  227. 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]
  228. 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]
  229. 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]
  230. 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]
  231. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA A Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
  232. 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]
  233. 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]
  234. 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]
  235. 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]
  236. 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]
  237. 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]
  238. 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]
  239. 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]
  240. 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]
  241. 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]
  242. 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]
  243. 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]
  244. 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]
  245. 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]
  246. 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]
  247. 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]
  248. 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]
  249. 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]
  250. 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]
  251. 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]
  252. 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]
  253. 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]
  254. 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]
  255. 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]
  256. 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]
  257. 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]
  258. 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]
  259. 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]
  260. 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]
  261. 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]
  262. 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]
  263. 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]
  264. 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]
  265. 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]
  266. 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]
  267. 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]
  268. 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]
  269. 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]
  270. 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]
  271. 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]
  272. 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]
  273. 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]
  274. 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]
  275. 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]
  276. 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]
  277. 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]
  278. 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]
  279. 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]
  280. 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]
  281. 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]
  282. 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]
  283. 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]
  284. 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]
  285. 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]
  286. 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]
  287. 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]
  288. 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]
  289. 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]
  290. 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]
  291. 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]
  292. Althaus, K.; Greinacher, A. MYH9-Related Platelet Disorders. Semin. Thromb. Hemost. 2009, 35, 189–203. [Google Scholar] [CrossRef]
  293. 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]
  294. 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]
  295. Kunishima, S. May-Hegglin anomaly--from genome research to clinical laboratory. Rinsho Byori 2003, 51, 898–904. [Google Scholar] [PubMed]
  296. 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]
  297. 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]
  298. 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]
  299. 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]
  300. 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]
  301. 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]
  302. 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]
  303. 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]
  304. 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]
  305. 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]
  306. 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]
  307. 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]
  308. 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]
  309. 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]
  310. 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]
  311. 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]
  312. 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]
  313. 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]
  314. 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]
  315. 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]
  316. 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]
  317. 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]
  318. 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]
  319. 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]
  320. 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]
  321. 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]
  322. 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]
  323. 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]
  324. 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]
  325. 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]
  326. 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]
  327. 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]
  328. Garnock-Jones, K.P. Ripasudil: First Global Approval. Drugs 2014, 74, 2211–2215. [Google Scholar] [CrossRef]
  329. 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]
  330. 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]
  331. 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]
  332. 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]
  333. 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]
  334. 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]
  335. 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]
  336. 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]
  337. 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]
  338. 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]
  339. 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]
  340. 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]
  341. 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]
  342. 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]
  343. 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]
  344. 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]
  345. 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]
  346. 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]
  347. Pandya, P.; Orgaz, J.L.; Sanz-Moreno, V. Modes of invasion during tumour dissemination. Mol. Oncol. 2017, 11, 5–27. [Google Scholar] [CrossRef]
  348. 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]
  349. 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]
  350. 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]
  351. Mizuno, K. Signaling mechanisms and functional roles of cofilin phosphorylation and dephosphorylation. Cell Signal 2013, 25, 457–469. [Google Scholar] [CrossRef]
  352. 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]
  353. 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]
  354. 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]
  355. 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]
  356. 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]
  357. 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]
  358. 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]
  359. 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]
  360. 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]
  361. 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]
  362. 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]
  363. 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]
  364. 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]
  365. 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]
  366. 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]
  367. 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]
  368. 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]
  369. 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]
  370. 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]
  371. 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]
  372. 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]
  373. 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]
  374. 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]
  375. 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]
  376. 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]
  377. 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]
Figure 1. The expression and alteration profile of MYH9 at the pancancer level. (A) MYH9 mRNA expression from 32 TCGA datasets. (B) The alteration profile of MYH9 from the same 32 TCGA datasets. The expression and alteration frequency data were obtained from cBioPortal (www.cbioportal.org).
Figure 1. The expression and alteration profile of MYH9 at the pancancer level. (A) MYH9 mRNA expression from 32 TCGA datasets. (B) The alteration profile of MYH9 from the same 32 TCGA datasets. The expression and alteration frequency data were obtained from cBioPortal (www.cbioportal.org).
Ijms 25 09435 g001
Figure 3. The figure shows the specific kinases involved in the phosphorylation of serine and threonine residues of both RLCs and heavy chains. PKC, protein kinase C; MLCK, myosin light chain kinase; ROCK, Rho-associated protein kinase; TRPM7, transient receptor potential melastatin 7; PKCβ, protein kinase Cβ; CK II, casein kinase II. The figure was adapted from Pecci et al. [28].
Figure 3. The figure shows the specific kinases involved in the phosphorylation of serine and threonine residues of both RLCs and heavy chains. PKC, protein kinase C; MLCK, myosin light chain kinase; ROCK, Rho-associated protein kinase; TRPM7, transient receptor potential melastatin 7; PKCβ, protein kinase Cβ; CK II, casein kinase II. The figure was adapted from Pecci et al. [28].
Ijms 25 09435 g003
Figure 4. This figure illustrates the regulation of myosin II filament formation and activity. Specific serine residues on the myosin heavy chain (S1916, S1927, and S1943) are phosphorylated by various kinases, including MHCKA, MHCKB, MHCKC, TRPM6, TRPM7, PKC, and CK2. The phosphorylation of these sites is depicted as favoring the filamentous state of myosin, which is essential for mechanotransduction and ATP hydrolysis-driven interaction with actin filaments. Phosphatases are the enzymes responsible for dephosphorylation, which may reverse the phosphorylation effect, potentially leading to a shift back to the monomeric state of myosin. Mts1, also known as S100A4, is a calcium-binding protein that regulates myosin II function by modulating filament assembly. It binds to the myosin heavy chain, influencing the balance between monomeric and filamentous forms of myosin II. Mts1 typically inhibits filament formation, thereby controlling myosin’s contractile activity and its ability to interact with actin.
Figure 4. This figure illustrates the regulation of myosin II filament formation and activity. Specific serine residues on the myosin heavy chain (S1916, S1927, and S1943) are phosphorylated by various kinases, including MHCKA, MHCKB, MHCKC, TRPM6, TRPM7, PKC, and CK2. The phosphorylation of these sites is depicted as favoring the filamentous state of myosin, which is essential for mechanotransduction and ATP hydrolysis-driven interaction with actin filaments. Phosphatases are the enzymes responsible for dephosphorylation, which may reverse the phosphorylation effect, potentially leading to a shift back to the monomeric state of myosin. Mts1, also known as S100A4, is a calcium-binding protein that regulates myosin II function by modulating filament assembly. It binds to the myosin heavy chain, influencing the balance between monomeric and filamentous forms of myosin II. Mts1 typically inhibits filament formation, thereby controlling myosin’s contractile activity and its ability to interact with actin.
Ijms 25 09435 g004
Figure 5. The figure shows the structure and orientation of cell migration in the 2D environment. At the front, actin filaments within lamellipodia and filopodia are oriented with their rapidly polymerizing ends in the forward direction. In the main body, actin and myosin filaments form bipolar structures to aid in cell retraction. NM IIA and NM IIB show distinct localizations inside the cell, with NM IIA predominantly being found at the leading edge where actin dynamics are most active. NM IIB is predominant toward the rear end. The region between the leading and trailing edges contains varying concentrations of NM IIA and NM IIB. Additional molecules, such as RhoA, Rac1, Cdc42, Ca2+ ions, and αPKC, also play significant roles in this cellular organization and migration process.
Figure 5. The figure shows the structure and orientation of cell migration in the 2D environment. At the front, actin filaments within lamellipodia and filopodia are oriented with their rapidly polymerizing ends in the forward direction. In the main body, actin and myosin filaments form bipolar structures to aid in cell retraction. NM IIA and NM IIB show distinct localizations inside the cell, with NM IIA predominantly being found at the leading edge where actin dynamics are most active. NM IIB is predominant toward the rear end. The region between the leading and trailing edges contains varying concentrations of NM IIA and NM IIB. Additional molecules, such as RhoA, Rac1, Cdc42, Ca2+ ions, and αPKC, also play significant roles in this cellular organization and migration process.
Ijms 25 09435 g005
Figure 6. The formation of plasma membrane blebs consists of three phases: initiation, expansion, and retraction. Blebbing stimuli, such as Ca2+ influx and apoptosis, induce the initiation of membrane protrusion. Actomyosin contractility drives the expansion of blebs, which are devoid of the F-actin cortex. Rho-ROCK signaling then drives bleb retraction via actomyosin contractility. NM IIA contractile forces promote bleb retraction.
Figure 6. The formation of plasma membrane blebs consists of three phases: initiation, expansion, and retraction. Blebbing stimuli, such as Ca2+ influx and apoptosis, induce the initiation of membrane protrusion. Actomyosin contractility drives the expansion of blebs, which are devoid of the F-actin cortex. Rho-ROCK signaling then drives bleb retraction via actomyosin contractility. NM IIA contractile forces promote bleb retraction.
Ijms 25 09435 g006
Figure 7. A schematic representation of the MYH9 exons with common mutations found in patients with MYH9-RD. The color coding of exon organization is as follows: black, motor domain; green, neck; orange, coiled coil domain; and brown, non-helical tail.
Figure 7. A schematic representation of the MYH9 exons with common mutations found in patients with MYH9-RD. The color coding of exon organization is as follows: black, motor domain; green, neck; orange, coiled coil domain; and brown, non-helical tail.
Ijms 25 09435 g007
Table 1. This table summarizes the key intramolecular interactions within the myosin II molecule that help maintain its 10S or “OFF” state.
Table 1. This table summarizes the key intramolecular interactions within the myosin II molecule that help maintain its 10S or “OFF” state.
InteractionInteraction TypeComponents InvolvedSpecific RegionsSignificance
BF1Head–HeadBlocked Head (BH) ↔ Free Head (FH)BH: Loop I365–N381, Helix T382–L390 ↔ FH: Helix E727–Y734, Loop E735–D748Stabilizes the myosin heads in the pre-power stroke state, preventing ATP hydrolysis and actin binding.
BF2Head–HeadBH RLC ↔ FH RLCN terminal lobes: BH: Helix A, A–B linker ↔ FH: Helix D, A–B linkerStrengthens head–head interaction, crucial for regulating muscle activity in the 10S state.
TB1Head–TailBlocked Head (BH) ↔ Tail (Seg3)Seg3: L1604–E1612 ↔ BH: Loop L450–F460Provides weak electrostatic interaction that contributes to maintaining the 10S state.
TB2Head–TailBlocked Head (BH) ↔ Tail (Seg2)Seg2: L1431–D1436 ↔ BH: Helix K72–D74 (SH3 domain)Physically blocks BH converter domain movement, preventing ATP turnover.
TB3Head–TailBlocked Head (BH) ↔ Tail (Seg2)Seg2: Q1445–L1452 ↔ BH: R718, L766 (near converter domain)Inhibits necessary movements in the BH, trapping ATP hydrolysis products.
TB4Head–TailBlocked Head (BH) ↔ TailTail: L1494–L1498 ↔ BH: Helix D (ELC N-lobe)Stabilizes 10S conformation by reinforcing the interaction between the tail and BH.
TB5Head–TailBlocked Head (BH) ↔ Tail (Seg3)Seg3: A1577–R1584 ↔ BH: Helix E (RLC)Further stabilizes the 10S structure by anchoring Seg3 to the BH regulatory domain.
TB6Head–TailBlocked Head (BH) RLC ↔ Tail (Seg3)BH RLC N terminal extension ↔ Seg3: Residues 1560–1572Crucial for maintaining the 10S state by strengthening the head–tail interaction, involving the phosphorylation domain (PD).
TF1Head–TailFree Head (FH) ↔ Tail (Seg1)FH: CM Loop T404–K420 ↔ Seg1: M925–A941Inhibits FH actin binding, keeping the myosin in an inactive state.
TF2Head–TailFree Head (FH) ↔ Tail (Seg1)FH: Loop 2 K626–T658 ↔ Seg1: M925–A941Further inhibits actin binding by the FH, reinforcing the 10S state.
TF3Head–TailFree Head (FH) RLC ↔ Tail (Seg1)FH RLC Helix A ↔ Seg1: L850–Q856Influences regulatory movements of the FH RLC, contributing to the stability of the 10S conformation.
TT1Tail–TailSegment 1 (Seg1) ↔ Segment 3 (Seg3)Weak electrostatic interactions across several regionsSupports the compact folding of the tail, crucial for maintaining the 10S state.
TT2Tail–TailSegment 1 (Seg1) ↔ Segment 3 (Seg3)Seg1: R910–M925 ↔ Seg3: L1628–E1647Maintains close alignment of the tail segments, reinforcing the 10S conformation.
Table 2. Altered NM IIA regulation in various cancer.
Table 2. Altered NM IIA regulation in various cancer.
Tumor TypeMechanismReferences
Squamous cell carcinoma
Human head and neck: dual role as tumor promoter and tumor suppressor
[200,201]
Tumor suppressor:
Mouse head and neck: tumor suppressor
[202]
Skin (mouse): tumor suppressor
[202]
Tongue (mouse): tumor suppressor
[203]
Melanoma
Mouse: tumor suppressor
[204]
Tumor promoter:
Gliomas
Glioma: acts as tumor promoter and interacts with THBS1 to promote glioma cell malignancy
[205]
Glioblastoma: promotes pro-tumorigenic vascular functions
[206]
Nasopharyngeal carcinoma
Promotes tumor stemness and EMT, leading to NPC progression
[207,208,209,210,211]
Lung carcinoma
In non-small cell lung carcinoma, NM IIA is associated with poor prognosis
[212,213]
NM IIA is involved in development of resistance to EGFR Tyrosine kinase inhibitors in NSCLC
[214]
NM IIA promotes malignant behavior of lung adenocarcinoma
[215]
Hepatocellular carcinoma
NM IIA overexpression is associated with stemness and poor prognosis
[216]
Low NM IIA level is associated with reduced cell proliferation, metastasis, and resistance to sorafenib
[217]
Esophageal squamous cell carcinoma
NM II overexpression is associated with poor prognosis
[218,219,220]
Pancreatic cancer
High levels of NM IIA promote EMT and metastasis
[221]
Gastric cancer
NM II overexpression is associated with poor prognosis
[222,223]
Colorectal cancer
NM IIA promotes CRC metastasis by interacting with MAPK/AKT, ATG9B/ integrin β1, and LIMK/actinin-4
[224,225,226]
Breast cancer
Phosphorylation of NM IIA at Ser1943 residue results in increased cell motility and chemotaxis
[95]
In TNBC cells, myosin-IIA, along with integrin β1 and MLCK, contributes to adaptive resistance development against MEK inhibitors
[227]
NM IIA modulates biophysical characteristics of ECM during cancer progression
[228]
Renal cancer
NM IIA facilitates nuclear translocation of chemokine receptor CXCR4 which drives metastasis
[229]
Overexpression of NM IIA drives progression of clear cell RCC
[230]
Prostate cancer
NM IIA maintains nuclear stability during cell migration
[231]
Osteosarcoma
High levels of NM IIA activate Wnt/β-catenin pathway to promote tumorigenesis
[232]
Thyroid cancer
In papillary thyroid carcinoma, NM IIA promotes secretion of matrix metalloproteinase 1 (MMP1) to increase cell proliferation and metastasis
[233]
In differentiated thyroid carcinoma, high levels of NM IIA contribute to development of radioresistance
[234]
Acute myeloid leukemia
Increased NM IIA expression enhances actomyosin contractility of AML cells
[235]
Diffuse large B cell lymphoma
NM IIA activates Wnt-β-catenin-STAT3 pathway to promote disease progression
[236]
Table 3. Drugs targeting MYH9.
Table 3. Drugs targeting MYH9.
DrugsMode of ActionReferences
Blebbistatin
Non-competitive myosin-II inhibitor
[320]
Cinobufotalin
Reduces expression of MYH9 in HCC and lung cancer
[217,255]
Apatinib
Inhibits phosphorylation of VEGFR2 in glioma cells
[205]
Disulfiram
Directly targets MYH9 and Ras GTPase-activating-like protein IQGAP1 to inhibit growth of TNBC cells
[323]
C70-EDA
Inhibits NM IIA filament assembly in A549 lung adenocarcinoma cells
[324]
J13
Disrupts MYH9 interaction with actin
[325]
Staurosporine
Inhibits phosphorylation of MYH9 at Ser1943 residue in gastric cancer
[222]
Homoharringtonine
Upregulates MYH9 expression in AML and CML cell lines
[326]
Fasudil
Rho-kinase inhibitor and calcium channel blocker
[327]
Ripasudil and Netarsudil
ROCK1 and ROCK2 inhibitor
[328,329]
Belumosudil
Selective ROCK2 inhibitor
[330,331]
Y27632
ROCK1 and ROCK2 inhibitor
[332]
GSK269962A
Selective ROCK1 inhibitor
[333]
AT13148
Dual ROCK/AKT inhibitor
[334]
BDP5290
MRCK inhibitor
[335]
Rhodblock 6
Rho kinase inhibitor
[336]
RKI-1447 and RKI-18
Rho kinase inhibitor
[337,338]
Thiosemicarbazone iron chelators
Inhibits ROCK1/p-MLC2 pathway
[339]
Combretastatin (CA-4)
Increases RLC phosphorylation
[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.

Share and Cite

MDPI and ACS Style

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

AMA Style

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 Style

Feroz, 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

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop