Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters

Database
Language
Journal subject
Affiliation country
Publication year range
1.
J Cell Sci ; 134(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34013964

ABSTRACT

Myosin XIX (Myo19) is an actin-based motor that competes with adaptors of microtubule-based motors for binding to the outer mitochondrial transmembrane proteins Miro1 and Miro2 (collectively Miro, also known as RhoT1 and RhoT2, respectively). Here, we investigate which mitochondrial and cellular processes depend on the coordination of Myo19 and microtubule-based motor activities. To this end, we created Myo19-deficient HEK293T cells. Mitochondria in these cells were not properly fragmented at mitosis and were partitioned asymmetrically to daughter cells. Respiratory functions of mitochondria were impaired and ROS generation was enhanced. On a cellular level, cell proliferation, cytokinesis and cell-matrix adhesion were negatively affected. On a molecular level, Myo19 regulates focal adhesions in interphase, and mitochondrial fusion and mitochondrially associated levels of fission protein Drp1 and adaptor proteins dynactin and TRAK1 at prometaphase. These alterations were due to a disturbed coordination of Myo19 and microtubule-based motor activities by Miro.


Subject(s)
Actins , Myosins , Actins/genetics , Actins/metabolism , HEK293 Cells , Humans , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myosins/metabolism , rho GTP-Binding Proteins/metabolism
2.
J Biol Chem ; 296: 100136, 2021.
Article in English | MEDLINE | ID: mdl-33268376

ABSTRACT

To migrate, cells assume a polarized morphology, extending forward with a leading edge with their trailing edge retracting back toward the cell body. Both cell extension and retraction critically depend on the organization and dynamics of the actin cytoskeleton, and the small, monomeric GTPases Rac and Rho are important regulators of actin. Activation of Rac induces actin polymerization and cell extension, whereas activation of Rho enhances acto-myosin II contractility and cell retraction. To coordinate migration, these processes must be carefully regulated. The myosin Myo9b, a Rho GTPase-activating protein (GAP), negatively regulates Rho activity and deletion of Myo9b in leukocytes impairs cell migration through increased Rho activity. However, it is not known whether cell motility is regulated by global or local inhibition of Rho activity by Myo9b. Here, we addressed this question by using Myo9b-deficient macrophage-like cells that expressed different recombinant Myo9b constructs. We found that Myo9b accumulates in lamellipodial extensions generated by Rac-induced actin polymerization as a function of its motor activity. Deletion of Myo9b in HL-60-derived macrophages altered cell morphology and impaired cell migration. Reintroduction of Myo9b or Myo9b motor and GAP mutants revealed that local GAP activity rescues cell morphology and migration. In summary, Rac activation leads to actin polymerization and recruitment of Myo9b, which locally inhibits Rho activity to enhance directional cell migration.


Subject(s)
Actin Cytoskeleton/metabolism , GTPase-Activating Proteins/metabolism , Macrophages/cytology , Macrophages/metabolism , Myosins/metabolism , Pseudopodia/metabolism , Cell Movement/physiology , Cells, Cultured , GTPase-Activating Proteins/genetics , Humans , Myosins/genetics
3.
J Biol Chem ; 294(18): 7202-7218, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30737279

ABSTRACT

Whereas myosin 18B (Myo18B) is known to be a critical sarcomeric protein, the function of myosin 18A (Myo18A) is unclear, although it has been implicated in cell motility and Golgi shape. Here, we show that homozygous deletion (homozygous tm1a, tm1b, or tm1d alleles) of Myo18a in mouse is embryonic lethal. Reminiscent of Myo18b, Myo18a was highly expressed in the embryo heart, and cardiac-restricted Myo18a deletion in mice was embryonic lethal. Surprisingly, using Western blot analysis, we were unable to detect the known isoforms of Myo18A, Myo18Aα and Myo18Aß, in mouse heart using a custom C-terminal antibody. However, alternative anti-Myo18A antibodies detected a larger than expected protein, and RNA-Seq analysis indicated that a novel Myo18A transcript is expressed in mouse ventricular myocytes (and human heart). Cloning and sequencing revealed that this cardiac isoform, denoted Myo18Aγ, lacks the PDZ-containing N terminus of Myo18Aα but includes an alternative N-terminal extension and a long serine-rich C terminus. EGFP-tagged Myo18Aγ expressed in ventricular myocytes localized to the level of A-bands in sarcomeres, and Myo18a knockout embryos at day 10.5 exhibited disorganized sarcomeres with wavy thick filaments. We additionally generated myeloid-restricted Myo18a knockout mice to investigate the role of Myo18A in nonmuscle cells, exemplified by macrophages, which express more Myo18Aß than Myo18Aα, but no defects in cell shape, motility, or Golgi shape were detected. In summary, we have identified a previously unrecognized sarcomere component, a large novel isoform (denoted Myo18Aγ) of Myo18A. Thus, both members of class XVIII myosins are critical components of cardiac sarcomeres.


Subject(s)
Myocardium/metabolism , Myosins/metabolism , Sarcomeres/metabolism , Animals , Gene Deletion , Genes, Lethal , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Knockout , Myosins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
J Cell Sci ; 131(17)2018 09 10.
Article in English | MEDLINE | ID: mdl-30111583

ABSTRACT

Mitochondrial distribution in cells is critical for cellular function and proper inheritance during cell division. In mammalian cells, mitochondria are transported predominantly along microtubules by kinesin and dynein motors that bind indirectly via TRAK1 and TRAK2 to outer mitochondrial membrane proteins Miro1 and Miro2 (Miro1/2). Here, using proximity labelling, we identified Miro1/2 as potential binding partners of myosin XIX (Myo19). Interaction studies show that Miro1 binds directly to a C-terminal fragment of the Myo19 tail region and that Miro1/2 recruit the Myo19 tail in vivo This recruitment is regulated by the nucleotide state of the N-terminal Rho-like GTPase domain of Miro1/2. Notably, Myo19 protein stability in cells depends on its association with Miro1/2. Downregulation of Miro1/2 or overexpression of the adaptor proteins TRAK1 and TRAK2 caused a reduction in Myo19 protein levels. Myo19 regulates the subcellular distribution of mitochondria, and downregulation, as well as overexpression, of Myo19 induced perinuclear collapse of mitochondria, phenocopying loss of the kinesin KIF5, dynein or their mitochondrial receptors Miro1/2. These results suggest that Miro1 and Miro2 coordinate microtubule- and actin-based mitochondrial movement.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Myosins/metabolism , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dyneins/genetics , Dyneins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Kinesins/genetics , Kinesins/metabolism , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Myosins/chemistry , Myosins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Domains , rho GTP-Binding Proteins/genetics
5.
FEBS Lett ; 584(3): 493-9, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19968988

ABSTRACT

Immune cells navigate through different environments where they experience different mechanical forces. Responses to external forces are determined by the mechanical properties of a cell and they depend to a large extent on the actin-rich cell cortex. We report here that Myo1G, a previously uncharacterised member of class I myosins, is expressed specifically in haematopoietic tissues and cells. It is associated with the plasma membrane. This association is dependent on a conserved PH-domain-like myosin I tail homology motif and the head domain. However, the head domain does not need to be a functional motor. Knockdown of Myo1G in Jurkat cells decreased cell elasticity significantly. We propose that Myo1G regulates cell elasticity by deformations of the actin network at the cell cortex.


Subject(s)
Cell Membrane/metabolism , Elasticity/physiology , Hematopoiesis/physiology , Myosins/physiology , Actins/metabolism , Animals , Cell Line , Cell Line, Tumor , Hematopoiesis/genetics , In Vitro Techniques , Lymph Nodes/metabolism , Mice , Microscopy, Atomic Force , Microscopy, Fluorescence , Myosins/genetics , Myosins/metabolism , RNA, Small Interfering , Spleen/metabolism , Thymus Gland/metabolism
6.
J Biol Chem ; 284(6): 3663-71, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19059909

ABSTRACT

The mammalian class IX myosin Myo9b can move considerable distances along actin filaments before it dissociates. This is remarkable, because it is single headed and because the rate-limiting step in its ATPase cycle is ATP hydrolysis. Thus, it spends most of its cycling time in the ATP-bound state that has a weak affinity for F-actin in other myosins. It has been speculated that the very extended loop 2 in the Myo9b head domain comprises an additional actin-binding site that prevents it from dissociation in the weak binding states. Here we show that two regions in the loop 2 determine the F-actin concentrations needed to maximally activate the steady-state ATPase activity. Together these two regions regulate the amount capable of binding F-actin and the affinity of the nucleotide-free state. The isolated loop 2 behaved like an entropic spring and bound stoichiometrically and with high affinity to F-actin. Subfragment 1 from skeletal muscle myosin II bound to F-actin simultaneously with the isolated loop 2 of Myo9b and could not displace it. Furthermore, the present results imply also a regulatory role for the tail region. Taken together, the results demonstrate that the extended loop 2 in Myo9b binds F-actin and influences the binding of the conventional stereo-specific actin-binding site.


Subject(s)
Actins/chemistry , Adenosine Triphosphate/chemistry , Myosins/chemistry , Actins/genetics , Actins/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Binding Sites/physiology , Cell Line , Hydrolysis , Myosins/genetics , Myosins/metabolism , Protein Binding/physiology , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Rats
7.
Mol Biol Cell ; 20(24): 5074-85, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19828736

ABSTRACT

The ependymal multiciliated epithelium in the brain restricts the cerebrospinal fluid to the cerebral ventricles and regulates its flow. We report here that mice deficient for myosin IXa (Myo9a), an actin-dependent motor molecule with a Rho GTPase-activating (GAP) domain, develop severe hydrocephalus with stenosis and closure of the ventral caudal 3rd ventricle and the aqueduct. Myo9a is expressed in maturing ependymal epithelial cells, and its absence leads to impaired maturation of ependymal cells. The Myo9a deficiency further resulted in a distorted ependyma due to irregular epithelial cell morphology and altered organization of intercellular junctions. Ependymal cells occasionally delaminated, forming multilayered structures that bridged the CSF-filled ventricular space. Hydrocephalus formation could be significantly attenuated by the inhibition of the Rho-effector Rho-kinase (ROCK). Administration of ROCK-inhibitor restored maturation of ependymal cells, but not the morphological distortions of the ependyma. Similarly, down-regulation of Myo9a by siRNA in Caco-2 adenocarcinoma cells increased Rho-signaling and induced alterations in differentiation, cell morphology, junction assembly, junctional signaling, and gene expression. Our results demonstrate that Myo9a is a critical regulator of Rho-dependent and -independent signaling mechanisms that guide epithelial differentiation. Moreover, Rho-kinases may represent a new target for therapeutic intervention in some forms of hydrocephalus.


Subject(s)
Cell Differentiation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hydrocephalus/metabolism , Hydrocephalus/pathology , Myosins/metabolism , Administration, Oral , Amides/pharmacology , Animals , Animals, Newborn , Caco-2 Cells , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Shape/drug effects , Cerebral Aqueduct/drug effects , Cerebral Aqueduct/metabolism , Cerebral Aqueduct/pathology , Constriction, Pathologic/pathology , Ependyma/drug effects , Ependyma/metabolism , Ependyma/pathology , Epithelial Cells/drug effects , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Mice , Mice, Knockout , Myosins/deficiency , Pyridines/pharmacology , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL