Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters








Publication year range
1.
Commun Biol ; 7(1): 1305, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39394274

ABSTRACT

Lysosomes, crucial cellular organelles, undergo bidirectional transport along microtubules, mediated by motor proteins such as cytoplasmic dynein-1 (dynein) and various kinesins. While the kinesin-3 family member KIF1C is established in mediating anterograde vesicle transport, its role in lysosomal transport remains unclear. Our study reveals that KIF1C unexpectedly supports the retrograde transport of lysosomes, driven by dynein, and contributes to their perinuclear localization. Notably, while KIF1C facilitates this perinuclear positioning, its motor activity is not required and, instead, exerts an inhibitory effect on this process. Mechanistically, KIF1C facilitates this process by interacting with the dynein-activating adaptor Hook3, which associates with the lysosome-anchored protein RUFY3. This regulatory mechanism is critical for the efficient degradation of cargo in autophagic and endocytic pathways. Our findings identify an unconventional, non-motor role for KIF1C in activating dynein-driven lysosomal transport, expanding our understanding of its functional diversity in cellular trafficking.


Subject(s)
Dyneins , Kinesins , Lysosomes , Microtubule-Associated Proteins , Kinesins/metabolism , Kinesins/genetics , Lysosomes/metabolism , Humans , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , HeLa Cells , Biological Transport , Protein Transport
2.
Glia ; 72(2): 411-432, 2024 02.
Article in English | MEDLINE | ID: mdl-37904612

ABSTRACT

Astrocytes, a type of glial cells, play critical roles in promoting the protection and repair of damaged tissues after brain injury. Inflammatory cytokines and growth factors can affect gene expression in astrocytes in injured brains, but signaling pathways and transcriptional mechanisms that regulate tissue protective functions of astrocytes are still poorly understood. In this study, we investigated the molecular mechanisms regulating the function of reactive astrocytes induced in mouse models of stab wound (SW) brain injury and collagenase-induced intracerebral hemorrhage (ICH). We show that basic fibroblast growth factor (bFGF), whose expression is up-regulated in mouse brains after SW injury and ICH, acts synergistically with inflammatory cytokines to activate E2F1-mediated transcription of a gene encoding the Ror-family protein Ror2, a receptor for Wnt5a, in cultured astrocytes. We also found that subsequent activation of Wnt5a/Ror2 signaling in astrocytes results in nuclear accumulation of antioxidative transcription factor Nrf2 at least partly by increased expression of p62/Sqstm1, leading to promoted expression of several Nrf2 target genes, including heme oxygenase 1. Finally, we provide evidence demonstrating that enhanced activation of Wnt5a/Ror2 signaling in astrocytes reduces cellular damage caused by hemin, a degradation product of hemoglobin, and promotes repair of the damaged blood brain barrier after brain hemorrhage.


Subject(s)
Brain Injuries , NF-E2-Related Factor 2 , Animals , Mice , Astrocytes/metabolism , Brain Injuries/genetics , Brain Injuries/metabolism , Cytokines/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction , Wnt-5a Protein/metabolism
3.
Genes Cells ; 28(4): 307-318, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36811220

ABSTRACT

Ror1 signaling regulates cell polarity, migration, proliferation, and differentiation during developmental morphogenesis, and plays an important role in regulating neurogenesis in the embryonic neocortices. However, the role of Ror1 signaling in the brains after birth remains largely unknown. Here, we found that expression levels of Ror1 in the mouse neocortices increase during the postnatal period, when astrocytes mature and start expressing GFAP. Indeed, Ror1 is highly expressed in cultured postmitotic mature astrocytes. RNA-Seq analysis revealed that Ror1 expressed in cultured astrocytes mediates upregulated expression of genes related to fatty acid (FA) metabolism, including the gene encoding carnitine palmitoyl-transferase 1a (Cpt1a), the rate-limiting enzyme of mitochondrial fatty acid ß-oxidation (FAO). We also found that Ror1 promotes the degradation of lipid droplets (LDs) accumulated in the cytoplasm of cultured astrocytes after oleic acid loading, and that suppressed expression of Ror1 decreases the amount of FAs localized at mitochondria, intracellular ATP levels, and expression levels of peroxisome proliferator-activated receptor α (PPARα) target genes, including Cpt1a. Collectively, these findings indicate that Ror1 signaling promotes PPARα-mediated transcription of FA metabolism-related genes, thereby facilitating the availability of FAs derived from LDs for mitochondrial FAO in the mature astrocytes.


Subject(s)
Astrocytes , PPAR alpha , Animals , Mice , Astrocytes/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Fatty Acids/metabolism , Lipid Metabolism , Mitochondria/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism
4.
Cancer Sci ; 114(2): 561-573, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36314076

ABSTRACT

Ror1 plays a crucial role in cancer progression by regulating cell proliferation and migration. Ror1 is expressed abundantly in various types of cancer cells and cancer stem-like cells. However, the molecular mechanisms regulating expression of Ror1 in these cells remain largely unknown. Ror1 and its putative ligand Wnt5a are expressed highly in malignant gliomas, especially in glioblastomas, and the extents of Ror1 expression are correlated positively with poorer prognosis in patients with gliomas. We show that Ror1 expression can be upregulated in glioblastoma cells under spheroid culture, but not adherent culture conditions. Notch and hypoxia signaling pathways have been shown to be activated in spheroid-forming glioblastoma stem-like cells (GSCs), and Ror1 expression in glioblastoma cells is indeed suppressed by inhibiting either Notch or hypoxia signaling. Meanwhile, either forced expression of the Notch intracellular domain (NICD) in or hypoxic culture of glioblastoma cells result in enhanced expression of Ror1 in the cells. Consistently, we show that both NICD and hypoxia-inducible factor 1 alpha bind to upstream regions within the Ror1 gene more efficiently in GSCs under spheroid culture conditions. Furthermore, we provide evidence indicating that binding of Wnt5a to Ror1, upregulated by Notch and hypoxia signaling pathways in GSCs, might promote their spheroid-forming ability. Collectively, these findings indicate for the first time that Notch and hypoxia signaling pathways can elicit a Wnt5a-Ror1 axis through transcriptional activation of Ror1 in glioblastoma cells, thereby promoting their stem cell-like property.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/metabolism , Glioma/pathology , Signal Transduction , Hypoxia/pathology , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Brain Neoplasms/pathology , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
5.
J Biol Chem ; 298(7): 102090, 2022 07.
Article in English | MEDLINE | ID: mdl-35654143

ABSTRACT

Invadopodia on cancer cells play crucial roles in tumor invasion and metastasis by degrading and remodeling the surrounding extracellular matrices and driving cell migration in complex 3D environments. Previous studies have indicated that microtubules (MTs) play a crucial role in elongation of invadopodia, but not their formation, probably by regulating delivery of membrane and secretory proteins within invadopodia. However, the identity of the responsible MT-based molecular motors and their regulation has been elusive. Here, we show that KIF1C, a member of kinesin-3 family, is localized to the tips of invadopodia and is required for their elongation and the invasion of cancer cells. We also found that c-Src phosphorylates tyrosine residues within the stalk domain of KIF1C, thereby enhancing its association with tyrosine phosphatase PTPD1, that in turn activates MT-binding ability of KIF1C, probably by relieving the autoinhibitory interaction between its motor and stalk domains. These findings shed new insights into how c-Src signaling is coupled to the MT-dependent dynamic nature of invadopodia and also advance our understanding of the mechanism of KIF1C activation through release of its autoinhibition.


Subject(s)
Genes, src , Kinesins , Neoplasm Invasiveness , Podosomes , Cell Line, Tumor , Humans , Kinesins/genetics , Microtubules/metabolism , Phosphorylation , Podosomes/metabolism , Protein Tyrosine Phosphatases, Non-Receptor , Tyrosine/metabolism
6.
Front Cell Dev Biol ; 10: 891763, 2022.
Article in English | MEDLINE | ID: mdl-35493090

ABSTRACT

The Ror-family proteins, Ror1 and Ror2, act as receptors or co-receptors for Wnt5a and its related Wnt proteins to activate non-canonical Wnt signaling. Ror1 and/or Ror2-mediated signaling plays essential roles in regulating cell polarity, migration, proliferation and differentiation during developmental morphogenesis, tissue-/organo-genesis and regeneration of adult tissues following injury. Ror1 and Ror2 are expressed abundantly in developing tissues in an overlapping, yet distinct manner, and their expression in adult tissues is restricted to specific cell types such as tissue stem/progenitor cells. Expression levels of Ror1 and/or Ror2 in the adult tissues are increased following injury, thereby promoting regeneration or repair of these injured tissues. On the other hand, disruption of Wnt5a-Ror2 signaling is implicated in senescence of tissue stem/progenitor cells that is related to the impaired regeneration capacity of aged tissues. In fact, Ror1 and Ror2 are implicated in age-related diseases, including tissue fibrosis, atherosclerosis (or arteriosclerosis), neurodegenerative diseases, and cancers. In these diseases, enhanced and/or sustained (chronic) expression of Ror1 and/or Ror2 is observed, and they might contribute to the progression of these diseases through Wnt5a-dependent and -independent manners. In this article, we overview recent advances in our understanding of the roles of Ror1 and Ror2-mediated signaling in the development, tissue regeneration and age-related diseases, and discuss their potential to be therapeutic targets for chronic inflammatory diseases and cancers.

7.
J Neurochem ; 156(6): 834-847, 2021 03.
Article in English | MEDLINE | ID: mdl-33460120

ABSTRACT

PRMT1, a major arginine methyltransferase, plays critical roles in transcription, DNA damage response, and cell proliferation. Although we have previously discovered the crucial roles of PRMT1 for oligodendrocyte lineage progression in the central nervous system of neural stem cell-specific PRMT1 conditional knockout (PRMT1-CKO) mice, the context of other glial cell states that may cause the hypomyelination phenotype in PRMT1-CKO mice has not been explored so far. Here, we performed RNA-seq of the neonatal cortices of PRMT1-CKO mice to reveal overall gene expression changes and show the up-regulation of inflammatory signaling which is generally mediated by astrocytes and microglia in advance of the myelination defects. In particular, qRT-PCR analyses revealed Interleukin-6 (Il-6), a major central nervous system cytokine, was dramatically increased in the PRMT1-CKO brains. The gene expression changes led to augmentation of glial fibrillary acidic protein and Vimentin protein levels in PRMT1-CKO mice, showing severe reactive astrogliosis after birth. We further show that IBA1-positive and CD68-positive activated microglia were increased in PRMT1-CKO mice, in spite of intact Prmt1 gene expression in purified microglia from the mutant mice. Our results indicate that PRMT1 loss in the neural stem cell lineage causes disruptive changes in all glial types perturbing postnatal brain development and myelination.


Subject(s)
Astrocytes , Brain/growth & development , Central Nervous System/metabolism , Microglia , Protein-Arginine N-Methyltransferases/genetics , Animals , Animals, Newborn , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Encephalitis/physiopathology , Female , Interleukin-6/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Myelin Sheath , Neural Stem Cells/metabolism , Pregnancy , RNA, Small Interfering/pharmacology , Signal Transduction
8.
Dev Dyn ; 250(1): 27-38, 2021 01.
Article in English | MEDLINE | ID: mdl-31925877

ABSTRACT

The Ror-family receptor tyrosine kinases (RTKs), consisting of Ror1 and Ror2, play crucial roles in morphogenesis and formation of various tissues/organs, including the bones and skeletal muscles, the so-called musculoskeletal system, during embryonic development, by acting as receptors or coreceptors for a noncanonical Wnt protein Wnt5a. Furthermore, several lines of evidence have indicated that Ror1 and/or Ror2 play critical roles in the regeneration and maintenance of the musculoskeletal system in adults. Considering the anatomical and functional relationship between the skeleton and skeletal muscles, their structural and functional association might be tightly regulated during their embryonic development, development after birth, and their regeneration after injury in adults. Importantly, in addition to their congenital anomalies, much attention has been paid onto the age-related disorders of the musculoskeletal system, including osteopenia and sarcopenia, which affect severely the quality of life. In this article, we overview recent advances in our understanding of the roles of Ror1- and/or Ror2-mediated signaling in the embryonic development, regeneration in adults, and congenital and age-related disorders of the musculoskeletal system and discuss possible therapeutic approaches to locomotive syndromes by modulating Ror1- and/or Ror2-mediated signaling.


Subject(s)
Musculoskeletal Development , Musculoskeletal Diseases/enzymology , Musculoskeletal System/enzymology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/metabolism , Animals , Humans , Ligands , Wnt Signaling Pathway
9.
FASEB J ; 34(2): 3413-3428, 2020 02.
Article in English | MEDLINE | ID: mdl-31922321

ABSTRACT

Ror2 signaling has been shown to regulate the cell cycle progression in normal and cancer cells. However, the molecular mechanism of the cell cycle progression upon activation of Ror2 signaling still remains unknown. Here, we found that the expression levels of Ror2 in G1-arrested NIH/3T3 fibroblasts are low and are rapidly increased following the cell cycle progression induced by basic fibroblast growth factor (bFGF) stimulation. By expressing wild-type or a dominant negative mutant of E2F1, we show that E2F1 mediates bFGF-induced expression of Ror2, and that E2F1 binds to the promoter of the Ror2 gene to activate its expression. We also found that G1/S phase transition of bFGF-stimulated NIH/3T3 cells is delayed by the suppressed expression of Ror2. RNA-seq analysis revealed that the suppressed expression of Ror2 results in the decreased expression of various E2F target genes concomitantly with increased expression of Forkhead box O (FoxO) target genes, including p21Cip1 , and p27Kip1 . Moreover, the inhibitory effect of Ror2 knockdown on the cell cycle progression can be restored by suppressed expression of p21Cip1 , p27Kip1 ,or FoxO3a. Collectively, these findings indicate that E2F1-Ror2 signaling mediates the transcriptional activation and inhibition of E2F1-driven and FoxO3a-driven cell cycle-regulated genes, respectively, thereby promoting G1/S phase transition of bFGF-stimulated NIH/3T3 cells.


Subject(s)
E2F1 Transcription Factor/metabolism , G1 Phase , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , S Phase , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , E2F1 Transcription Factor/genetics , Fibroblast Growth Factors/metabolism , Forkhead Box Protein O3/metabolism , Mice , NIH 3T3 Cells , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction
10.
Hum Mol Genet ; 27(7): 1174-1185, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29360985

ABSTRACT

Congenital muscular dystrophies (CMDs) are characterized by progressive weakness and degeneration of skeletal muscle. In several forms of CMD, abnormal glycosylation of α-dystroglycan (α-DG) results in conditions collectively known as dystroglycanopathies, which are associated with central nervous system involvement. We recently demonstrated that fukutin, the gene responsible for Fukuyama congenital muscular dystrophy, encodes the ribitol-phosphate transferase essential for dystroglycan function. Brain pathology in patients with dystroglycanopathy typically includes cobblestone lissencephaly, mental retardation, and refractory epilepsy; however, some patients exhibit average intelligence, with few or almost no structural defects. Currently, there is no effective treatment for dystroglycanopathy, and the mechanisms underlying the generation of this broad clinical spectrum remain unknown. Here, we analysed four distinct mouse models of dystroglycanopathy: two brain-selective fukutin conditional knockout strains (neuronal stem cell-selective Nestin-fukutin-cKO and forebrain-selective Emx1-fukutin-cKO), a FukutinHp strain with the founder retrotransposal insertion in the fukutin gene, and a spontaneous Large-mutant Largemyd strain. These models exhibit variations in the severity of brain pathology, replicating the clinical heterogeneity of dystroglycanopathy. Immunofluorescence analysis of the developing cortex suggested that residual glycosylation of α-DG at embryonic day 13.5 (E13.5), when cortical dysplasia is not yet apparent, may contribute to subsequent phenotypic heterogeneity. Surprisingly, delivery of fukutin or Large into the brains of mice at E12.5 prevented severe brain malformation in Emx1-fukutin-cKO and Largemyd/myd mice, respectively. These findings indicate that spatiotemporal persistence of functionally glycosylated α-DG may be crucial for brain development and modulation of glycosylation during the fetal stage could be a potential therapeutic strategy for dystroglycanopathy.


Subject(s)
Brain/embryology , Dystroglycans/metabolism , Fetus/embryology , Gene Transfer Techniques , Genetic Therapy , Malformations of Cortical Development/therapy , Animals , Brain/pathology , Dystroglycans/genetics , Female , Fetus/pathology , Glycosylation , Male , Malformations of Cortical Development/embryology , Malformations of Cortical Development/genetics , Malformations of Cortical Development/pathology , Mice , Mice, Transgenic
11.
Dev Dyn ; 247(1): 24-32, 2018 01.
Article in English | MEDLINE | ID: mdl-28470690

ABSTRACT

The Ror-family of receptor tyrosine kinases (RTKs) are involved critically in tissue genesis and organogenesis during development. In mammals, Ror1 and Ror2, members of the Ror-family RTKs, have been shown to mediate cell polarity, migration, proliferation, and differentiation through the activation of noncanonical Wnt signaling by acting as receptors or co-receptors for Wnt5a. Nematodes bearing mutations within the cam-1 gene, encoding a Ror2 ortholog, exhibit defects in various developmental processes of the nervous system, including neuronal cell migration, polarization, axonal extension, and synaptic transmission. In mice, Ror2 and/or Ror1 are also shown to play roles in regulating neurite extension, synapse formation, and synaptic transmission of hippocampal neurons, indicating that the Ror-family RTKs have evolutionarily conserved functions at least in part in neurons during development. Furthermore, Ror2 and/or Ror1 are expressed in neural stem/progenitor cells of the developing brain and in astrocytes of the adult brain after injury, and they play important roles in regulating cell proliferation under these different contexts. In this article, we overview recent advances in our understanding of the roles of the Ror-family RTKs in the development and repair of the nervous system and discuss their potential for therapeutic targets to neurodegenerative diseases. Developmental Dynamics 247:24-32, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Animals , Neural Stem Cells/metabolism , Signal Transduction/physiology
12.
J Biol Chem ; 292(38): 15939-15951, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28790171

ABSTRACT

The Ror family receptor tyrosine kinases, Ror1 and Ror2, play important roles in regulating developmental morphogenesis and tissue- and organogenesis, but their roles in tissue regeneration in adult animals remain largely unknown. In this study, we examined the expression and function of Ror1 and Ror2 during skeletal muscle regeneration. Using an in vivo skeletal muscle injury model, we show that expression of Ror1 and Ror2 in skeletal muscles is induced transiently by the inflammatory cytokines, TNF-α and IL-1ß, after injury and that inhibition of TNF-α and IL-1ß by neutralizing antibodies suppresses expression of Ror1 and Ror2 in injured muscles. Importantly, expression of Ror1, but not Ror2, was induced primarily in Pax7-positive satellite cells (SCs) after muscle injury, and administration of neutralizing antibodies decreased the proportion of Pax7-positive proliferative SCs after muscle injury. We also found that stimulation of a mouse myogenic cell line, C2C12 cells, with TNF-α or IL-1ß induced expression of Ror1 via NF-κB activation and that suppressed expression of Ror1 inhibited their proliferative responses in SCs. Intriguingly, SC-specific depletion of Ror1 decreased the number of Pax7-positive SCs after muscle injury. Collectively, these findings indicate for the first time that Ror1 has a critical role in regulating SC proliferation during skeletal muscle regeneration. We conclude that Ror1 might be a suitable target in the development of diagnostic and therapeutic approaches to manage muscular disorders.


Subject(s)
Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Line , Cell Proliferation , Gene Expression Regulation, Enzymologic , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Development , NF-kappa B/metabolism , PAX7 Transcription Factor/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
13.
Glia ; 65(1): 182-197, 2017 01.
Article in English | MEDLINE | ID: mdl-27726178

ABSTRACT

Ror2 receptor tyrosine kinase plays crucial roles in developmental morphogenesis and tissue-/organo-genesis. In the developing brain, Ror2 is expressed in neural stem/progenitor cells (NPCs) and involved in the regulation of their stemness. However, it remains largely unknown about its role in the adult brain. In this study, we show that Ror2 is up-regulated in reactive astrocytes in the neocortices within 3 days following stab-wound injury. Intriguingly, Ror2-expressing astrocytes were detected primarily at the area surrounding the injury site, where astrocytes express Nestin, a marker of NPCs, and proliferate in response to injury. Furthermore, we show by using astrocyte-specific Ror2 knockout (KO) mice that a loss of Ror2 in astrocytes attenuates injury-induced proliferation of reactive astrocytes. It was also found that basic fibroblast growth factor (bFGF) is strongly up-regulated at 1 day post injury in the neocortices, and that stimulation of cultured quiescent astrocytes with bFGF restarts their cell cycle and induces expression of Ror2 during the G1 phase predominantly in proliferating cells. By using this culture method, we further show that the proportions of Ror2-expressing astrocytes increase following treatment with the histone deacetylases inhibitors including valproic acid, and that bFGF stimulation increases the levels of Ror2 expression within the respective cells. Moreover, we show that bFGF-induced cell cycle progression into S phase is inhibited or promoted in astrocytes from Ror2 KO mice or NPCs stably expressing Ror2-GFP, respectively. Collectively, these findings indicate that Ror2 plays a critical role in regulating the cell cycle progression of reactive astrocytes following brain injury, GLIA 2016. GLIA 2017;65:182-197.


Subject(s)
Astrocytes/metabolism , Brain Injuries/enzymology , Cell Cycle/physiology , Cell Division , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Animals , Brain Injuries/pathology , Cell Division/physiology , Cells, Cultured , Mice, Knockout , Nestin/metabolism , Neural Stem Cells/metabolism
14.
Int Rev Cell Mol Biol ; 314: 117-48, 2015.
Article in English | MEDLINE | ID: mdl-25619716

ABSTRACT

Wnt5a is involved in the activation of noncanonical Wnt signaling, including planar cell polarity (PCP) and Wnt-Ca(2+) pathways. The Ror-family of receptor tyrosine kinases is composed of Ror1 and Ror2 in mammals. Ror2 acts as a receptor or coreceptor for Wnt5a and regulates Wnt5a-induced activation of PCP pathway, and Wnt5a-Ror2 axis indeed plays critical roles in the developmental morphogenesis by regulating cell polarity and migration. Furthermore, Wnt5a-Ror2 axis is constitutively activated in cancer cells and confers highly motile and invasive properties on cancer cells through the expression of matrix metalloproteinase genes and enhanced formation of invadopodia. Meanwhile, Wnt5a also exhibits a tumor-suppressive function in certain cancers, including breast and colorectal carcinomas. Thus, it is of great importance to understand the respective molecular mechanisms governing Wnt5a-mediated tumor-progressive and tumor-suppressive functions, in order to develop novel and proper diagnostic and therapeutic strategies targeting Wnt5a signaling for human cancers.


Subject(s)
Calcium Signaling , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Cell Polarity/genetics , Collagenases/genetics , Collagenases/metabolism , Humans , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/genetics , Wnt-5a Protein
15.
Genes Cells ; 19(4): 287-96, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24475942

ABSTRACT

Activation of Wnt/ß-catenin signal in muscle satellite cells (mSCs) of aged mice during myogenic differentiation has been appreciated as an important age-related feature of the skeletal muscles, resulting in impairment of their regenerative ability following muscle injury. However, it remains elusive about molecules involved in this age-related alteration of Wnt/ß-catenin signal in myogenic cells. To clarify this issue, we carried out expression analyses of Wnt receptor genes using real-time RT-PCR in mSCs isolated from the skeletal muscles of young and aged mice. Here, we show that expression of Frizzled1 (Fzd1) was detected at high levels in mSCs of aged mice. Higher expression levels of Fzd1 were also detected in mSC-derived myogenic cells from aged mice and associated with activation of Wnt/ß-catenin signal during their myogenic differentiation in vitro. We also provide evidence that suppressed expression of Fzd1 in myogenic cells from aged mice results in a significant increase in myogenic differentiation, and its forced expression in those from young mice results in its drastic inhibition. These findings indicate the critical role of Fzd1 in altered myogenic differentiation associated with aging.


Subject(s)
Cell Differentiation/physiology , Frizzled Receptors/metabolism , Satellite Cells, Skeletal Muscle/cytology , Wnt Proteins/metabolism , beta Catenin/metabolism , Age Factors , Aging , Animals , Cells, Cultured , Male , Mice, Inbred ICR , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction
16.
Genes Cells ; 18(7): 608-19, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23755735

ABSTRACT

Activation of Wnt5a-Ror2 signaling has been shown to be associated with epithelial-to-mesenchymal transition (EMT) of epidermoid carcinoma cells via induction of matrix metalloproteinase-2 (MMP-2). Because EMT has also been implicated in the progression of tissue fibrosis, we examined the possible association of Wnt5a-Ror2 signaling with renal fibrosis. Here, we show that expression of Wnt5a and Ror2 is induced in a damaged mouse kidney after unilateral ureteral obstruction (UUO) treatment. Immunofluorescent analysis showed that Ror2 expression is clearly induced in tubular epithelial cells during renal fibrosis, and these Ror2-expressing cells also express Snail and vimentin, markers of mesenchymal cells, suggesting that Ror2 might be induced in epithelial cells undergoing EMT. We also found that MMP-2 expression is induced at Ror2-positive epithelium adjacent to significantly disrupted tubular basement membrane (TBM). Interestingly, reduced expression of MMP-2 is detected at epithelium in damaged kidneys from Ror2(+/-) mice compared with those from wild-type Ror2(+/+) mice. Importantly, extents of TBM disruption are apparently reduced in damaged kidneys from Ror2(+/-) mice compared with those from wild-type mice. Collectively, these findings indicate that activation of Wnt5a-Ror2 signaling in epithelial cells undergoing EMT may play an important role in disrupting TBM via MMP-2 induction during renal fibrosis.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Fibrosis/metabolism , Kidney Diseases/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/metabolism , Animals , Disease Models, Animal , Epithelial Cells/pathology , Fibrosis/pathology , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction , Wnt Proteins/genetics , Wnt-5a Protein
17.
Clin Calcium ; 23(6): 809-15, 2013 Jun.
Article in Japanese | MEDLINE | ID: mdl-23719492

ABSTRACT

Signal transduction, elicited by Wnt-family of secreted proteins, can be classified intoß-catenin-dependent canonical Wnt signaling and -independent non-canonical Wnt signaling. Non-canonical Wnt signaling contains planar cell polarity (PCP) pathway and Ca (+ +) pathway, which play central roles in developmental morphogenesis, by regulating cellular functions, including cell polarity and cell migration. In this article, we will overview the molecular basis of non-canonical Wnt signaling, with a particular emphasis on the roles of non-canonical Wnt signaling mediated by Wnt5a and its cognate receptors, Ror-family of receptor tyrosine kinases (Ror1, Ror2) , and will introduce up-to-date information on non-canonical Wnt signaling obtained from recent studies about pathological conditions, including cancer progression and chronic inflammation.


Subject(s)
Wnt Signaling Pathway/physiology , Animals , Cell Polarity/physiology , Humans , Receptors, Cell Surface/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism
18.
J Cell Sci ; 125(Pt 8): 2017-29, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22328498

ABSTRACT

The Ror family receptor tyrosine kinases (RTKs), Ror1 and Ror2, have been shown to play crucial roles in developmental morphogenesis by acting as receptors or co-receptors to mediate Wnt5a-induced signaling. Although Ror1, Ror2 and Wnt5a are expressed in the developing brain, little is known about their roles in the neural development. Here we show that Ror1, Ror2 and their ligand Wnt5a are highly expressed in neocortical neural progenitor cells (NPCs). Small interfering RNA (siRNA)-mediated suppression of Ror1, Ror2 or Wnt5a in cultured NPCs isolated from embryonic neocortex results in the reduction of ßIII-tubulin-positive neurons that are produced from NPCs possibly through the generation of T-box brain 2 (Tbr2)-positive intermediate progenitors. BrdU-labeling experiments further reveal that the proportion of proliferative and neurogenic NPCs, which are positive for neural progenitor cell marker (Pax6) but negative for glial cell marker (glial fibrillary acidic protein; GFAP), is reduced within a few days in culture following knockdown of these molecules, suggesting that Ror1, Ror2 and Wnt5a regulate neurogenesis through the maintenance of NPCs. Moreover, we show that Dishevelled 2 (Dvl2) is involved in Wnt5a-Ror1 and Wnt5a-Ror2 signaling in NPCs, and that suppressed expression of Dvl2 indeed reduces the proportion of proliferative and neurogenic NPCs. Interestingly, suppressed expression of either Ror1 or Ror2 in NPCs in the developing neocortex results in the precocious differentiation of NPCs into neurons, and their forced expression results in delayed differentiation. Collectively, these results indicate that Wnt5a-Ror1 and Wnt5a-Ror2 signaling pathways play roles in maintaining proliferative and neurogenic NPCs during neurogenesis of the developing neocortex.


Subject(s)
Neocortex/embryology , Neocortex/enzymology , Neural Stem Cells/enzymology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Animals , Cell Proliferation , Cells, Cultured , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred ICR , Neocortex/cytology , Neural Stem Cells/cytology , Neurogenesis , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein
19.
Methods Mol Biol ; 839: 201-14, 2012.
Article in English | MEDLINE | ID: mdl-22218903

ABSTRACT

Planar cell polarity (PCP) pathway of Wnt signaling plays a crucial role to establish the polarization of cells during tissue development. Our recent findings using in vitro analyses have revealed that Ror2, a member of the Ror-family receptor tyrosine kinases, acts as a receptor or co-receptor for Wnt5a and plays a crucial role for Wnt5a-induced polarized cell migration through activating PCP pathway. Indeed, analyses of both Wnt5a and Ror2 mutant mice have shown that Wnt5a-Ror2 signaling is involved in establishing the PCP in epithelial tissues in vivo, indicating that in vitro analyses of polarized cell migration and PCP signaling induced by Wnt5a can be useful tools to explore putative regulators involved in Wnt/PCP pathway. Here, we introduce in vitro methods using cultured cells to monitor polarized cell migration and PCP signaling induced by Wnt5a.


Subject(s)
Cell Polarity , Signal Transduction , Wnt Proteins/metabolism , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Genes, Reporter/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Microtubules/metabolism , NIH 3T3 Cells , Phosphoproteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Transcription Factor AP-1/metabolism , Wnt-5a Protein
20.
J Biol Chem ; 286(7): 5157-65, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21149453

ABSTRACT

Bone morphogenetic proteins (BMPs) regulate many mammalian physiologic and pathophysiologic processes. These proteins bind with the kinase receptors BMPR-I and BMPR-II, thereby activating Smad transcription factor. In this study, we demonstrate that neogenin, a receptor for netrins and proteins of the repulsive guidance molecule family, is a receptor for BMPs and modulates Smad signal transduction. Neogenin was found to bind directly with BMP-2, BMP-4, BMP-6, and BMP-7. Knockdown of neogenin in C2C12 cells resulted in the enhancement of the BMP-2-induced processes of osteoblastic differentiation and phosphorylation of Smad1, Smad5, and Smad8. Conversely, overexpression of neogenin in C2C12 cells suppressed these processes. Our results also indicated that BMP-induced activation of RhoA was mediated by neogenin. Inhibition of RhoA promoted BMP-2-induced processes of osteoblastic differentiation and phosphorylation of Smad1/5/8. However, treatment with Y-27632, an inhibitor of Rho-associated protein kinase, did not modulate BMP-induced phosphorylation of Smad1/5/8. Taken together, our findings suggest that neogenin negatively regulates the functions of BMP and that this effect of neogenin is mediated by the activation of RhoA.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Membrane Proteins/metabolism , Osteoblasts/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Proteins/genetics , Cell Differentiation/physiology , Enzyme Activation/physiology , HEK293 Cells , Humans , Membrane Proteins/genetics , Osteoblasts/cytology , Phosphorylation/physiology , Protein Binding/physiology , Smad Proteins/genetics , Smad Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL