72 results on '"Shigeyuki Nada"'
Search Results
2. β-catenin-promoted cholesterol metabolism protects against cellular senescence in naked mole-rat cells
- Author
-
Woei-Yaw Chee, Yuriko Kurahashi, Junhyeong Kim, Kyoko Miura, Daisuke Okuzaki, Tohru Ishitani, Kentaro Kajiwara, Shigeyuki Nada, Hideyuki Okano, and Masato Okada
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Woei-Yaw Chee et al. explore the molecular mechanisms of β-catenin involvement in cholesterol metabolism suppressing cellular senescence in naked mole-rat cells. The results of this study suggest that β-catenin in NMRs functions to offset senescence by regulating cholesterol metabolism, which may contribute to increased longevity in naked mole rats.
- Published
- 2021
- Full Text
- View/download PDF
3. Structural basis for the assembly of the Ragulator-Rag GTPase complex
- Author
-
Ryo Yonehara, Shigeyuki Nada, Tomokazu Nakai, Masahiro Nakai, Ayaka Kitamura, Akira Ogawa, Hirokazu Nakatsumi, Keiichi I. Nakayama, Songling Li, Daron M. Standley, Eiki Yamashita, Atsushi Nakagawa, and Masato Okada
- Subjects
Science - Abstract
mTORC1 activity is controlled through Rag GTPases, which are anchored to the lysosome through the Ragulator. Here, the authors give molecular insights into Ragulator-Rag GTPase assembly and present the crystal structures of the Ragulator alone and in complex with the RagA-C roadblock domains.
- Published
- 2017
- Full Text
- View/download PDF
4. Polarization of M2 macrophages requires Lamtor1 that integrates cytokine and amino-acid signals
- Author
-
Tetsuya Kimura, Shigeyuki Nada, Noriko Takegahara, Tatsusada Okuno, Satoshi Nojima, Sujin Kang, Daisuke Ito, Keiko Morimoto, Takashi Hosokawa, Yoshitomo Hayama, Yuichi Mitsui, Natsuki Sakurai, Hana Sarashina-Kida, Masayuki Nishide, Yohei Maeda, Hyota Takamatsu, Daisuke Okuzaki, Masaki Yamada, Masato Okada, and Atsushi Kumanogoh
- Subjects
Science - Abstract
The role of nutrient-sensing pathways in regulation of innate immune response is unexplored. Here the authors show that IL-4 activates the amino-acid sensing pathway in macrophages and leads to polarization of anti-inflammatory M2 macrophages via the transcription factor liver X receptor.
- Published
- 2016
- Full Text
- View/download PDF
5. Erratum: Polarization of M2 macrophages requires Lamtor1 that integrates cytokine and amino-acid signals
- Author
-
Tetsuya Kimura, Shigeyuki Nada, Noriko Takegahara, Tatsusada Okuno, Satoshi Nojima, Sujin Kang, Daisuke Ito, Keiko Morimoto, Takashi Hosokawa, Yoshitomo Hayama, Yuichi Mitsui, Natsuki Sakurai, Hana Sarashina-Kida, Masayuki Nishide, Yohei Maeda, Hyota Takamatsu, Daisuke Okuzaki, Masaki Yamada, Masato Okada, and Atsushi Kumanogoh
- Subjects
Science - Abstract
Nature Communications 7 Article number: 13130 (2016); Published 12 October 2016; Updated 27 February 2017 In this Article, there are errors in the labelling of the x axis in Fig. 5g,h that were introduced during the production process. The second label on the x axis of each graph in Fig. 5g should have been ‘ΦWT’ instead of ‘ΦKO’.
- Published
- 2017
- Full Text
- View/download PDF
6. The mTOR pathway controls cell proliferation by regulating the FoxO3a transcription factor via SGK1 kinase.
- Author
-
Shunsuke Mori, Shigeyuki Nada, Hironobu Kimura, Shoji Tajima, Yusuke Takahashi, Ayaka Kitamura, Chitose Oneyama, and Masato Okada
- Subjects
Medicine ,Science - Abstract
The mechanistic target of rapamycin (mTOR) functions as a component of two large complexes, mTORC1 and mTORC2, which play crucial roles in regulating cell growth and homeostasis. However, the molecular mechanisms by which mTOR controls cell proliferation remain elusive. Here we show that the FoxO3a transcription factor is coordinately regulated by mTORC1 and mTORC2, and plays a crucial role in controlling cell proliferation. To dissect mTOR signaling, mTORC1 was specifically inactivated by depleting p18, an essential anchor of mTORC1 on lysosomes. mTORC1 inactivation caused a marked retardation of cell proliferation, which was associated with upregulation of cyclin-dependent kinase inhibitors (CDKIs). Although Akt was activated by mTORC1 inactivation, FoxO3a was upregulated via an epigenetic mechanism and hypophosphorylated at Ser314, which resulted in its nuclear accumulation. Consistently, mTORC1 inactivation induced downregulation of serum- and glucocorticoid-inducible kinase 1 (SGK1), the kinase responsible for Ser314 phosphorylation. Expression of FoxO3a mutated at Ser314 suppressed cell proliferation by inducing CDKI expression. SGK1 overexpression suppressed CDKI expression in p18-deficient cells, whereas SGK1 knockdown induced CDKI expression in wild-type cells, resulting in the suppression of cell proliferation. These results suggest that mTORC1, in coordination with mTORC2, controls cell proliferation by regulating FoxO3a gene expression and SGK1-mediated phosphorylation of FoxO3a at Ser314.
- Published
- 2014
- Full Text
- View/download PDF
7. The lysosomal Ragulator complex activates NLRP3 inflammasome in vivo via HDAC6
- Author
-
Kohei Tsujimoto, Tatsunori Jo, Daiki Nagira, Hachiro Konaka, Jeong Hoon Park, Shin‐ichiro Yoshimura, Akinori Ninomiya, Fuminori Sugihara, Takehiro Hirayama, Eri Itotagawa, Yusei Matsuzaki, Yuki Takaichi, Wataru Aoki, Shotaro Saita, Shuhei Nakamura, Andrea Ballabio, Shigeyuki Nada, Masato Okada, Hyota Takamatsu, Atsushi Kumanogoh, Tsujimoto, Kohei, Jo, Tatsunori, Nagira, Daiki, Konaka, Hachiro, Park, Jeong Hoon, Yoshimura, Shin-Ichiro, Ninomiya, Akinori, Sugihara, Fuminori, Hirayama, Takehiro, Itotagawa, Eri, Matsuzaki, Yusei, Takaichi, Yuki, Aoki, Wataru, Saita, Shotaro, Nakamura, Shuhei, Ballabio, Andrea, Nada, Shigeyuki, Okada, Masato, Takamatsu, Hyota, and Kumanogoh, Atsushi
- Subjects
Inflammation ,α-tocopherol ,General Immunology and Microbiology ,Inflammasomes ,General Neuroscience ,alpha-Tocopherol ,Peritonitis ,HDAC6 ,Histone Deacetylase 6 ,General Biochemistry, Genetics and Molecular Biology ,NLRP3 inflammasome ,Uric Acid ,Mice, Inbred C57BL ,Mice ,Ragulator complex ,NLR Family, Pyrin Domain-Containing 3 Protein ,Humans ,Animals ,Lysosomes ,Molecular Biology - Abstract
The cellular activation of the NLRP3 inflammasome is spatiotemporally orchestrated by various organelles, but whether lysosomes contribute to this process remains unclear. Here, we show the vital role of the lysosomal membrane-tethered Ragulator complex in NLRP3 inflammasome activation. Deficiency of Lamtor1, an essential component of the Ragulator complex, abrogated NLRP3 inflammasome activation in murine macrophages and human monocytic cells. Myeloid-specific Lamtor1-deficient mice showed marked attenuation of NLRP3-associated inflammatory disease severity, including LPS-induced sepsis, alum-induced peritonitis, and monosodium urate (MSU)-induced arthritis. Mechanistically, Lamtor1 interacted with both NLRP3 and histone deacetylase 6 (HDAC6). HDAC6 enhances the interaction between Lamtor1 and NLRP3, resulting in NLRP3 inflammasome activation. DL-all-rac-α-tocopherol, a synthetic form of vitamin E, inhibited the Lamtor1-HDAC6 interaction, resulting in diminished NLRP3 inflammasome activation. Further, DL-all-rac-α-tocopherol alleviated acute gouty arthritis and MSU-induced peritonitis. These results provide novel insights into the role of lysosomes in the activation of NLRP3 inflammasomes by the Ragulator complex.
- Published
- 2023
8. A novel TGF-β-responsive enhancer regulates SRC expression and epithelial–mesenchymal transition-associated cell migration
- Author
-
Soshi Noshita, Yuki Kubo, Kentaro Kajiwara, Daisuke Okuzaki, Shigeyuki Nada, and Masato Okada
- Abstract
The non-receptor tyrosine kinase SRC is overexpressed and/or hyperactivated in various human cancers, and facilitates cancer progression by promoting invasion and metastasis. However, the mechanisms underlying SRC upregulation are poorly understood. In this study, we demonstrated that transforming growth factor-β (TGF-β) induces SRC expression at the transcriptional level by activating a novel intragenic SRC enhancer. In the human breast epithelial cell line MCF10A, TGF-β1 stimulation upregulated theSRC1Apromoter, resulting in increased SRC mRNA and protein levels. Chromatin immunoprecipitation (ChIP)-sequencing analysis revealed that SMAD complex is recruited to three enhancer regions ~15 kb upstream and downstream of theSRCpromoter, and one of them is capable of activating theSRCpromoter in response to TGF-β. In addition, JUN, which is a member of the activator protein (AP)-1 family, also localizes to the enhancer and regulates TGF-β-induced SRC expression. Furthermore, the total amount of active SRC also increased, coinciding with the TGF-β-induced SRC expression. In TGF-β-induced epithelial–mesenchymal transition (EMT), TGF-β-induced SRC upregulation plays a crucial role in EMT-associated cell migration by activating the SRC/focal adhesion kinase (FAK) circuit. Overall, these results suggest that TGF-β-induced SRC upregulation promotes cancer cell invasion and metastasis in a subset of human malignancies.
- Published
- 2022
- Full Text
- View/download PDF
9. The lysosomal Ragulator complex plays an essential role in leukocyte trafficking by activating myosin II
- Author
-
Masato Okada, Kohei Tsujimoto, Takeshi Nakatani, Atsushi Kumanogoh, Masayuki Nishide, JeongHoon Park, Hachiro Konaka, Tetsuya Kimura, Shyohei Koyama, Yasuhiro Kato, Tatsunori Jo, Hyota Takamatsu, Takayoshi Morita, Yoshitomo Hayama, and Shigeyuki Nada
- Subjects
Male ,0301 basic medicine ,Leukocyte migration ,Neutrophils ,Science ,Phosphatase ,Antigen-presenting cells ,General Physics and Astronomy ,Motility ,mTORC1 ,macromolecular substances ,Mechanistic Target of Rapamycin Complex 1 ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,Myosin-Light-Chain Phosphatase ,03 medical and health sciences ,0302 clinical medicine ,Myosin ,Leukocytes ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Myosin Type II ,Multidisciplinary ,Chemistry ,Intracellular Signaling Peptides and Proteins ,Cell migration ,Actomyosin ,Dendritic Cells ,General Chemistry ,Ragulator complex ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Amoeboid migration ,Female ,Myosin-light-chain phosphatase ,Lysosomes ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Lysosomes are involved in nutrient sensing via the mechanistic target of rapamycin complex 1 (mTORC1). mTORC1 is tethered to lysosomes by the Ragulator complex, a heteropentamer in which Lamtor1 wraps around Lamtor2–5. Although the Ragulator complex is required for cell migration, the mechanisms by which it participates in cell motility remain unknown. Here, we show that lysosomes move to the uropod in motile cells, providing the platform where Lamtor1 interacts with the myosin phosphatase Rho-interacting protein (MPRIP) independently of mTORC1 and interferes with the interaction between MPRIP and MYPT1, a subunit of myosin light chain phosphatase (MLCP), thereby increasing myosin II–mediated actomyosin contraction. Additionally, formation of the complete Ragulator complex is required for leukocyte migration and pathophysiological immune responses. Together, our findings demonstrate that the lysosomal Ragulator complex plays an essential role in leukocyte migration by activating myosin II through interacting with MPRIP., Myosin II–mediated contractility is required for leukocyte migration. Here, authors show that lysosomes are involved in leukocyte migration by providing the platform where Ragulator complex interacts with the myosin phosphatase Rho-interacting protein (MPRIP) independently of mTORC1 and interferes with the interaction between MPRIP and a subunit of myosin light chain phosphatase (MLCP).
- Published
- 2021
10. A TGF-β-responsive enhancer regulates SRC expression and epithelial–mesenchymal transition-associated cell migration.
- Author
-
Soshi Noshita, Yuki Kubo, Kentaro Kajiwara, Daisuke Okuzaki, Shigeyuki Nada, and Masato Okada
- Subjects
CELL migration ,EPITHELIAL-mesenchymal transition ,PROTEIN-tyrosine kinases ,CANCER invasiveness ,CANCER cells ,BREAST - Abstract
The non-receptor tyrosine kinase SRC is overexpressed and/or hyperactivated in various human cancers, and facilitates cancer progression by promoting invasion and metastasis. However, the mechanisms underlying SRC upregulation are poorly understood. In this study, we demonstrate that transforming growth factor-β (TGF-β) induces SRC expression at the transcriptional level by activating an intragenic the SRC enhancer. In the human breast epithelial cell line MCF10A, TGF-β1 stimulation upregulated one of the SRC promotors, the 1A promoter, resulting in increased SRC mRNA and protein levels. Chromatin immunoprecipitation (ChIP)-sequencing analysis revealed that the SMAD complex is recruited to three enhancer regions ∼15 kb upstream and downstream of the SRC promoter, and one of them is capable of activating the SRC promoter in response to TGF-β. JUN, a member of the activator protein (AP)-1 family, localises to the enhancer and regulates TGF-β-induced SRC expression. Furthermore, TGF-β-induced SRC upregulation plays a crucial role in epithelial–mesenchymal transition (EMT)-associated cell migration by activating the SRC–focal adhesion kinase (FAK) circuit. Overall, these results suggest that TGF-β-induced SRC upregulation promotes cancer cell invasion and metastasis in a subset of human malignancies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. β-catenin-promoted cholesterol metabolism protects against cellular senescence in naked mole-rat cells
- Author
-
Hideyuki Okano, Kyoko Miura, Yuriko Kurahashi, Junhyeong Kim, Tohru Ishitani, Daisuke Okuzaki, Woei Yaw Chee, Masato Okada, Kentaro Kajiwara, and Shigeyuki Nada
- Subjects
0301 basic medicine ,Senescence ,Apolipoprotein B ,QH301-705.5 ,Longevity ,Medicine (miscellaneous) ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Lipid droplet ,medicine ,Animals ,Biology (General) ,Liver X receptor ,Wnt Signaling Pathway ,Naked mole-rat ,Cellular Senescence ,beta Catenin ,biology ,Chemistry ,Cholesterol ,Mole Rats ,Wnt signaling pathway ,Lipid Droplets ,Fibroblasts ,biology.organism_classification ,Cell biology ,Oxidative Stress ,030104 developmental biology ,Apolipoproteins ,biology.protein ,NIH 3T3 Cells ,lipids (amino acids, peptides, and proteins) ,General Agricultural and Biological Sciences ,030217 neurology & neurosurgery ,Oxidative stress ,Cell signalling - Abstract
The naked mole-rat (NMR; Heterocephalus glaber) exhibits cancer resistance and an exceptionally long lifespan of approximately 30 years, but the mechanism(s) underlying increased longevity in NMRs remains unclear. In the present study, we report unique mechanisms underlying cholesterol metabolism in NMR cells, which may be responsible for their anti-senescent properties. NMR fibroblasts expressed β-catenin abundantly; this high expression was linked to increased accumulation of cholesterol-enriched lipid droplets. Ablation of β-catenin or inhibition of cholesterol synthesis abolished lipid droplet formation and induced senescence-like phenotypes accompanied by increased oxidative stress. β-catenin ablation downregulated apolipoprotein F and the LXR/RXR pathway, which are involved in cholesterol transport and biogenesis. Apolipoprotein F ablation also suppressed lipid droplet accumulation and promoted cellular senescence, indicating that apolipoprotein F mediates β-catenin signaling in NMR cells. Thus, we suggest that β-catenin in NMRs functions to offset senescence by regulating cholesterol metabolism, which may contribute to increased longevity in NMRs., Woei-Yaw Chee et al. explore the molecular mechanisms of β-catenin involvement in cholesterol metabolism suppressing cellular senescence in naked mole-rat cells. The results of this study suggest that β-catenin in NMRs functions to offset senescence by regulating cholesterol metabolism, which may contribute to increased longevity in naked mole rats.
- Published
- 2021
12. p18/Lamtor1-mTORC1 Signaling Controls Development of Mucin-producing Goblet Cells in the Intestine
- Author
-
Masato Okada, Daisuke Yamazaki, Hiroaki Miki, Shizuka Ito, Kentaro Kajiwara, Shigeyuki Nada, and Tetsuya Kimura
- Subjects
endocrine system ,endocrine system diseases ,Physiology ,medicine.medical_treatment ,Cellular differentiation ,Crypt ,Enteroendocrine cell differentiation ,Kruppel-Like Transcription Factors ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,Conditional gene knockout ,medicine ,Animals ,CDX2 Transcription Factor ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Chemistry ,Growth factor ,030302 biochemistry & molecular biology ,Cell Biology ,General Medicine ,Intestinal epithelium ,Cell biology ,KLF4 ,Goblet Cells ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) plays a pivotal role in controlling cell growth and metabolism in response to nutrients and growth factors. The activity of mTORC1 is dually regulated by amino acids and growth factor signaling, and amino acid-dependent mTORC1 activity is regulated by mTORC1 interaction with the Ragulator-Rag GTPase complex, which is localized to the surface of lysosomes via a membrane-anchored protein, p18/Lamtor1. However, the physiological function of p18-Ragulator-dependent mTORC1 signaling remains elusive. The present study evaluated the function of p18-mediated mTORC1 signaling in the intestinal epithelia using p18 conditional knockout mice. In p18 knockout colonic crypts, mTORC1 was delocalized from lysosomes, and in vivo mTORC1 activity was markedly decreased. Histologically, p18 knockout crypts exhibited significantly increased proliferating cells and dramatically decreased mucin-producing goblet cells, while overall crypt architecture and enteroendocrine cell differentiation were unaffected. Furthermore, p18 knockout crypts normally expressed transcription factors implicated in crypt differentiation, such as Cdx2 and Klf4, indicating that p18 ablation did not affect the genetic program of cell differentiation. Analysis of colon crypt organoid cultures revealed that both p18 ablation and rapamycin treatment robustly suppressed development of mucin-producing goblet cells. Hence, p18-mediated mTORC1 signaling could promote the anabolic metabolism required for robust mucin production in goblet cells to protect the intestinal epithelia from various external stressors.Key words: mTORC1, p18/lamtor1, intestinal epithelium, goblet cells, mucin.
- Published
- 2020
- Full Text
- View/download PDF
13. The Ragulator complex serves as a substrate-specific mTORC1 scaffold in regulating the nuclear translocation of transcription factor EB
- Author
-
Tetsuya Kimura, Yoshitomo Hayama, Daisuke Okuzaki, Shigeyuki Nada, and Masato Okada
- Subjects
Cell Nucleus ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Cell Biology ,Intracellular Membranes ,Mechanistic Target of Rapamycin Complex 1 ,Biochemistry ,Mice ,Protein Transport ,RAW 264.7 Cells ,Animals ,Lysosomes ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Signal Transduction - Abstract
The mammalian target of rapamycin complex 1 (mTORC1) signaling pathway is activated by intracellular nutritional sufficiency and extracellular growth signals. It has been reported that mTORC1 acts as a hub that integrates these inputs to orchestrate a number of cellular responses, including translation, nucleotide synthesis, lipid synthesis, and lysosome biogenesis. However, little is known about specific control of mTORC1 signaling downstream of this complex. Here, we demonstrate that Ragulator, a heteropentameric protein complex required for mTORC1 activation in response to amino acids, is critical for inhibiting the nuclear translocation of transcription factor EB (TFEB). We established a unique RAW264.7 clone that lacked Ragulator but retained total mTORC1 activity. In a nutrition-sufficient state, the nuclear translocation of TFEB was markedly enhanced in the clone despite total mTORC1 kinase activity. In addition, as a cellular phenotype, the number of lysosomes was increased by tenfold in the Ragulator-deficient clone compared with that of control cells. These findings indicate that mTORC1 essentially requires the Ragulator complex for regulating the subcellular distribution of TFEB. Our findings also suggest that other scaffold proteins may be associated with mTORC1 for the specific regulation of downstream signaling.
- Published
- 2021
14. d-Serine Mediates Cellular Proliferation for Kidney Remodeling
- Author
-
Naotsugu Ichimaru, Maiko Nakane, Masataka Kawamura, Shigeyuki Nada, Yoshitaka Isaka, Tomonori Kimura, Atsushi Hesaka, Daisuke Okuzaki, Masato Okada, Shinsuke Sakai, Masashi Mita, and Yusuke Tsukamoto
- Subjects
Kidney ,medicine.medical_specialty ,business.industry ,Renal function ,General Medicine ,Cell cycle ,Nephrectomy ,Genetically modified organism ,Gene expression profiling ,Serine ,Mice ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,Living Donors ,medicine ,Animals ,Humans ,Biomarker (medicine) ,business ,Function (biology) ,Cell Proliferation ,Original Investigation - Abstract
Background: D-Serine, a long-term undetected enantiomer of serine, is a biomarker that reflects kidney function and disease activity. The physiological functions of D-serine have been unclear. Methods: Dynamics of D-serine was assessed by measuring D-serine in human samples of living kidney donors using two-dimensional high-performance liquid chromatography, and by auto-radiographic studies in mice. Effects of D-serine on kidney were examined by gene expression profiling and metabolic studies using unilateral nephrectomy mice, and genetically modified cells. Results: Unilateral nephrectomy in human living kidney donors decreases urinary excretion and thus increases the blood level of D-serine. D-Serine is quickly and dominantly distributed to the kidney upon injection in mice, suggesting that the kidney is a main target organ. Treatment of D-serine at low dose promotes the enlargement of remnant kidney in mouse model. Mechanistically, D-serine activates the cell cycle for tissue remodeling through an mTOR-related pathway. Conclusions: D-Serine is a physiological molecule that promotes kidney remodeling. Besides its function as a biomarker, D-serine has a physiological activity that influences kidney function.
- Published
- 2021
15. Atg5-mediated autophagy controls apoptosis/anoikis via p53/Rb pathway in naked mole-rat fibroblasts
- Author
-
Norikazu Yabuta, Kentaro Kajiwara, Woei-Yaw Chee, Junhyeong Kim, Shigeyuki Nada, and Masato Okada
- Subjects
0301 basic medicine ,Male ,ATG5 ,Biophysics ,Apoptosis ,Biochemistry ,Retinoblastoma Protein ,Autophagy-Related Protein 5 ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Autophagy ,Cell Adhesion ,Animals ,Anoikis ,Cell adhesion ,Molecular Biology ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Proliferation ,Skin ,Gene knockdown ,Chemistry ,Cell growth ,Mole Rats ,Cell Biology ,Fibroblasts ,Cell biology ,Mitochondria ,Up-Regulation ,030104 developmental biology ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Tumor Suppressor Protein p53 ,Autophagy-Related Protein 12 ,Signal Transduction - Abstract
The naked mole-rat (NMR, Heterocephalus glaber) is the longest-living known rodent species, with a maximum lifespan of over 30 years. NMRs exhibit negligible senescence, exceptional resistance to cancer, and high basal autophagy activity compared with mouse. The molecular mechanisms and physiological roles underlying the high basal autophagy activity in NMRs remain to be elucidated. We identified that the Atg12-Atg5 conjugate, a critical component of autophagosome formation, was highly expressed in NMR skin fibroblasts (NSFs) compared with that in mouse skin fibroblasts. Phenotypic analysis of Atg5 knockdown NSFs revealed that high basal autophagy activity in NSFs was associated with abundant expression of the Atg12-Atg5 conjugate. Atg5 knockdown in NSFs led to accumulation of dysfunctional mitochondria, and suppressed cell proliferation and cell adhesion ability, promoting apoptosis/anoikis accompanied by upregulation of the apoptosis-related genes, Bax and Noxa. Furthermore, inhibition of the p53/Rb pro-apoptotic pathway with SV40 large T antigen abolished Atg5 knockdown-induced increases in apoptosis/anoikis. Taken together, these findings suggest that high basal autophagy activity in NMR cells, mediated by Atg5, contributes to suppression of p53/Rb-induced apoptosis, which could benefit the longevity of NMR cells.
- Published
- 2020
16. Genetic dissection of Ragulator structure and function in amino acid-dependent regulation of mTORC1
- Author
-
Shigeyuki Nada and Masato Okada
- Subjects
Phosphatase ,GTPase ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Lysosome ,medicine ,Humans ,Amino Acids ,Molecular Biology ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,chemistry.chemical_classification ,0303 health sciences ,biology ,General Medicine ,Regulatory-Associated Protein of mTOR ,Ragulator complex ,Amino acid ,Cell biology ,medicine.anatomical_structure ,chemistry ,Gene Expression Regulation ,biology.protein ,Ras Homolog Enriched in Brain Protein ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Function (biology) ,RHEB - Abstract
Ragulator is a heteropentameric protein complex consisting of two roadblock heterodimers wrapped by the membrane anchor p18/Lamtor1. The Ragulator complex functions as a lysosomal membrane scaffold for Rag GTPases to recruit and activate mechanistic target of rapamycin complex 1 (mTORC1). However, the roles of Ragulator structure in the regulation of mTORC1 function remain elusive. In this study, we disrupted Ragulator structure by directly anchoring RagC to lysosomes and monitored the effect on amino acid-dependent mTORC1 activation. Expression of lysosome-anchored RagC in p18-deficient cells resulted in constitutive lysosomal localization and amino acid-independent activation of mTORC1. Co-expression of Ragulator in this system restored the amino acid dependency of mTORC1 activation. Furthermore, ablation of Gator1, a suppressor of Rag GTPases, induced amino acid-independent activation of mTORC1 even in the presence of Ragulator. These results demonstrate that Ragulator structure is essential for amino acid-dependent regulation of Rag GTPases via Gator1. In addition, our genetic analyses revealed new roles of amino acids in the regulation of mTORC1 as follows: amino acids could activate a fraction of mTORC1 in a Rheb-independent manner, and could also drive negative-feedback regulation of mTORC1 signalling via protein phosphatases. These intriguing findings contribute to our overall understanding of the regulatory mechanisms of mTORC1 signalling.
- Published
- 2020
17. Ubiquitination of Src promotes its secretion via small extracellular vesicles
- Author
-
Kentaro Kajiwara, Kentaro Tanaka, Shigeyuki Nada, Masato Okada, and Yuko Ito
- Subjects
0301 basic medicine ,biology ,Endosome ,Chemistry ,Mutant ,Biophysics ,Cell Biology ,Biochemistry ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Ubiquitin ,Downregulation and upregulation ,030220 oncology & carcinogenesis ,Invadopodia ,biology.protein ,Secretion ,Molecular Biology ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Upregulation of the Src tyrosine kinase is implicated in the progression of cancer. The oncogenic potential of Src is suppressed via several negative regulation systems including degradation via the ubiquitin-proteasome pathway. Here, we show that ubiquitination of Src promotes its secretion via small extracellular vesicles (sEVs) to suppress its oncogenic potential. In MDCK cells expressing a modified Src that can be activated by hydroxytamoxifen, activated Src was transported to late endosomes/lysosomes and secreted via sEVs. The secretion of Src was suppressed by ablation of Cbl E3-ligase, suggesting the contribution of ubiquitination to this process. Activated Src was ubiquitinated at multiple sites, and Lys429 was identified as a critical site for sEV-mediated secretion. Mutation of Src at Lys429 (R429) caused resistance to ubiquitination and decreased its secretion via sEVs. The activated R429 mutant was also transported to late endosomes/lysosomes, whereas its incorporation into intraluminal vesicles was reduced. Activation of the R429 mutant induced a greater FAK activation than that of wild-type Src, thereby potentiating Src-induced invasive phenotypes, such as invadopodia formation and invasive activity. These findings demonstrate that ubiquitination of activated Src at Lys429 promotes its secretion via sEVs, suggesting a potential strategy to suppress the oncogenic function of upregulated Src.
- Published
- 2020
18. Lysosomal Protein Lamtor1 Controls Innate Immune Responses via Nuclear Translocation of Transcription Factor EB
- Author
-
Yoshitomo Hayama, Hana Sarashina-Kida, Takeshi Nakatani, Yasuhiro Kato, Hyota Takamatsu, Shohei Koyama, Tetsuya Kimura, Takeshi Tsuda, Shigeyuki Nada, Yohei Maeda, Yoshito Takeda, Yuhei Kinehara, Satoshi Nojima, Daisuke Ito, Masayuki Nishide, Yoshimitsu Nakanishi, Taro Koba, Sujin Kang, Masato Okada, Atsushi Kumanogoh, and Takashi Hosokawa
- Subjects
0301 basic medicine ,Autophagosome ,Immunology ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Cell Line ,Mice ,03 medical and health sciences ,Lysosome ,Conditional gene knockout ,Autophagy ,medicine ,Animals ,Immunology and Allergy ,Amino Acids ,Phosphorylation ,Adaptor Proteins, Signal Transducing ,Cell Nucleus ,Mice, Knockout ,Innate immune system ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Chemistry ,Macrophages ,TOR Serine-Threonine Kinases ,Proteins ,Signal transducing adaptor protein ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,Protein Transport ,RAW 264.7 Cells ,030104 developmental biology ,medicine.anatomical_structure ,TFEB ,Lysosomes ,Signal Transduction - Abstract
Amino acid metabolism plays important roles in innate immune cells, including macrophages. Recently, we reported that a lysosomal adaptor protein, Lamtor1, which serves as the scaffold for amino acid–activated mechanistic target of rapamycin complex 1 (mTORC1), is critical for the polarization of M2 macrophages. However, little is known about how Lamtor1 affects the inflammatory responses that are triggered by the stimuli for TLRs. In this article, we show that Lamtor1 controls innate immune responses by regulating the phosphorylation and nuclear translocation of transcription factor EB (TFEB), which has been known as the master regulator for lysosome and autophagosome biogenesis. Furthermore, we show that nuclear translocation of TFEB occurs in alveolar macrophages of myeloid-specific Lamtor1 conditional knockout mice and that these mice are hypersensitive to intratracheal administration of LPS and bleomycin. Our observation clarified that the amino acid–sensing pathway consisting of Lamtor1, mTORC1, and TFEB is involved in the regulation of innate immune responses.
- Published
- 2018
- Full Text
- View/download PDF
19. Lamtor1 Is Critically Required for CD4+ T Cell Proliferation and Regulatory T Cell Suppressive Function
- Author
-
Shimon Sakaguchi, Daisuke Ito, Shigeyuki Nada, Yuhei Kinehara, Hyota Takamatsu, Takashi Hosokawa, Yoshitomo Hayama, Syohei Koyama, Atsushi Kumanogoh, Tetsuya Kimura, Masayuki Nishide, Norihisa Mikami, Kazuya Yamashita, Daisuke Okuzaki, Masato Okada, Takeshi Nakatani, Naganari Ohkura, Yasuhiro Kato, Satoshi Nojima, Tatsusada Okuno, and Sujin Kang
- Subjects
0301 basic medicine ,Regulatory T cell ,ZAP70 ,Immunology ,FOXP3 ,CD28 ,Biology ,Cell biology ,03 medical and health sciences ,Interleukin 21 ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Biochemistry ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,030215 immunology ,Interleukin 3 - Abstract
Mechanistic target of rapamycin complex (mTORC)1 integrates intracellular sufficiency of nutrients and regulates various cellular functions. Previous studies using mice with conditional knockout of mTORC1 component proteins (i.e., mTOR, Raptor, and Rheb) gave conflicting results on the roles of mTORC1 in CD4+ T cells. Lamtor1 is the protein that is required for amino acid sensing and activation of mTORC1; however, the roles of Lamtor1 in T cells have not been investigated. In this article, we show that Lamtor1-deficient CD4+ T cells exhibited marked reductions in proliferation, IL-2 production, mTORC1 activity, and expression of purine- and lipid-synthesis genes. Polarization of Th17 cells, but not Th1 and Th2 cells, diminished following the loss of Lamtor1. Accordingly, CD4-Cre–driven Lamtor1-knockout mice exhibited reduced numbers of CD4+ and CD8+ T cells at rest, and they were completely resistant to experimental autoimmune encephalomyelitis. In contrast, genetic ablation of Lamtor1 in Foxp3+ T cells resulted in severe autoimmunity and premature death. Lamtor1-deficient regulatory T cells survived ex vivo as long as wild-type regulatory T cells; however, they exhibited a marked loss of suppressive function and expression of signature molecules, such as CTLA-4. These results indicate that Lamtor1 plays essential roles in CD4+ T cells. Our data suggest that Lamtor1 should be considered a novel therapeutic target in immune systems.
- Published
- 2017
- Full Text
- View/download PDF
20. Constitutive activation of Src family kinases in mouse embryos that lack Csk
- Author
-
Shigeyuki Nada, Takeshi Yagi, Hiroyuki Takeda, Tomoyuki Tokunaga, Hachiro Nakagawa, Yoji Ikawa, Masato Okada, and Shinichi Aizawa
- Subjects
Protein tyrosine kinase -- Influence ,Neural tube -- Abnormalities ,Biological sciences - Abstract
C-Src kinase (csk), a cytoplasmic protein-tyrosine kinase, inhibits the kinase activity of Src family members by phosphorylating the negative regulatory tyrosine residue of Src. Csk-deficient embryos of mice exhibit characteristic abnormality in the development of neural tissues when the expression of p60 (super c-src) increases. An elevation in the kinase activity of p60 (super c-src) in csk-deficient embryos is due to a substantial reduction of phosphate on Tyr-527. Csk acts as anti-oncogene through its negative regulation of Src family kinases.
- Published
- 1993
21. Role of Ragulator in the Regulation of Mechanistic Target of Rapamycin Signaling in Podocytes and Glomerular Function
- Author
-
Masato Okada, Shigeyuki Nada, Ken Inoki, Junying Wang, Yao Yao, and Sei Yoshida
- Subjects
Male ,0301 basic medicine ,Kidney Glomerulus ,030232 urology & nephrology ,mTORC1 ,Bioinformatics ,Podocyte ,Diabetic nephropathy ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Lysosome ,Animals ,Medicine ,Phosphatidylinositol ,Mechanistic target of rapamycin ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,biology ,Podocytes ,business.industry ,TOR Serine-Threonine Kinases ,General Medicine ,medicine.disease ,Ragulator complex ,Cell biology ,Basic Research ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Nephrology ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Lysosomes ,business ,Signal Transduction ,RHEB - Abstract
Aberrant activation of mechanistic target of rapamycin complex 1 (mTORC1) in glomerular podocytes leads to glomerular insufficiency and may contribute to the development of glomerular diseases, including diabetic nephropathy. Thus, an approach for preventing mTORC1 activation may allow circumvention of the onset and progression of mTORC1-dependent podocyte injury and glomerular diseases. mTORC1 activation requires inputs from both growth factors and nutrients that inactivate the tuberous sclerosis complex (TSC), a key suppressor of mTORC1, on the lysosome. Previous studies in mice revealed that the growth factor-phosphatidylinositol 3-kinase pathway and mTORC1 are essential for maintaining normal podocyte function, suggesting that direct inhibition of the phosphatidylinositol 3-kinase pathway or mTORC1 may not be an ideal approach to sustaining physiologic podocyte functions under certain disease conditions. Here, we report the role of the Ragulator complex, which recruits mTORC1 to lysosomes in response to nutrient availability in podocytes. Notably, podocytes lacking Ragulator maintain basal mTORC1 activity. Unlike podocyte-specific mTORC1-knockout mice, mice lacking functional Ragulator in podocytes did not show abnormalities in podocyte or glomerular function. However, aberrant mTORC1 activation induced by active Rheb in podocyte-specific TSC1-knockout (podo-TSC1 KO) mice did require Ragulator. Moreover, ablation of Ragulator in the podocytes of podo-TSC1 KO mice or streptozotocin-induced diabetic mice significantly blocked the development of pathologic renal phenotypes. These observations suggest that the blockade of mTORC1 recruitment to lysosomes may be a useful clinical approach to attenuate aberrant mTORC1 activation under certain disease conditions.
- Published
- 2016
- Full Text
- View/download PDF
22. Src mediates TGF-β-induced intraocular pressure elevation in glaucoma
- Author
-
Kentaro Kajiwara, Shigeyuki Nada, Teruhisa Tsukamoto, and Masato Okada
- Subjects
0301 basic medicine ,Male ,Intraocular pressure ,genetic structures ,Physiology ,Clinical Biochemistry ,Dasatinib ,Glaucoma ,Focal adhesion ,Extracellular matrix ,03 medical and health sciences ,Transforming Growth Factor beta2 ,0302 clinical medicine ,Trabecular Meshwork ,Rats, Inbred BN ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Cells, Cultured ,Intraocular Pressure ,Adaptor Proteins, Signal Transducing ,Chemistry ,Cell Biology ,medicine.disease ,eye diseases ,Cell biology ,Enzyme Activation ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,src-Family Kinases ,030220 oncology & carcinogenesis ,Tissue Plasminogen Activator ,sense organs ,Trabecular meshwork ,Proto-oncogene tyrosine-protein kinase Src ,Transforming growth factor ,medicine.drug ,Signal Transduction - Abstract
Glaucoma, a progressive and irreversible optic neuropathy, is one of the leading causes of vision impairment worldwide. Elevation of intraocular pressure (IOP) due to transforming growth factor-β (TGF-β)-induced dysfunction of the trabecular meshwork is a risk factor for glaucoma, but the underlying molecular mechanisms remain elusive. Here, we show that Src kinase is involved in TGF-β-induced IOP elevation. We observed that dasatinib, a potent Src inhibitor, suppressed TGF-β2-induced IOP in rat eyes. Mechanistic analyses in human trabecular meshwork cells showed that TGF-β2 activated Src signaling and concomitantly increased cytoskeletal remodeling, cell adhesion, and extracellular matrix (ECM) accumulation. Src was activated via TGF-β2-induced upregulation of the Src scaffolding protein CasL, which mediates the assembly of focal adhesions, cytoskeletal remodeling, and ECM deposition. Activation of Src suppressed the expression of tissue plasminogen activator, thereby attenuating ECM degradation. Furthermore, the Src inhibitor ameliorated TGF-β2-induced changes in the contractile and adhesive characteristics of trabecular meshwork cells, and ECM deposition. These findings underscore the crucial role of Src activity in TGF-β-induced IOP elevation and identify Src signaling as a potential therapeutic target in glaucoma.
- Published
- 2018
23. Structural basis for the assembly of the Ragulator-Rag GTPase complex
- Author
-
Eiki Yamashita, Tomokazu Nakai, Masato Okada, Songling Li, Hirokazu Nakatsumi, Shigeyuki Nada, Daron M. Standley, Atsushi Nakagawa, M. Nakai, Keiichi I. Nakayama, Ryo Yonehara, Akira Ogawa, and Ayaka Kitamura
- Subjects
0301 basic medicine ,Pentameric protein ,Science ,General Physics and Astronomy ,GTPase ,mTORC1 ,Plasma protein binding ,Nutrient sensing ,Mechanistic Target of Rapamycin Complex 1 ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Humans ,lcsh:Science ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,GTPase complex ,Multidisciplinary ,Chemistry ,Cell growth ,Signal transducing adaptor protein ,hemic and immune systems ,General Chemistry ,Cell biology ,030104 developmental biology ,Multiprotein Complexes ,lcsh:Q ,Protein Multimerization ,Lysosomes ,030217 neurology & neurosurgery ,Protein Binding - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) plays a central role in regulating cell growth and metabolism by responding to cellular nutrient conditions. The activity of mTORC1 is controlled by Rag GTPases, which are anchored to lysosomes via Ragulator, a pentameric protein complex consisting of membrane-anchored p18/LAMTOR1 and two roadblock heterodimers. Here we report the crystal structure of Ragulator in complex with the roadblock domains of RagA-C, which helps to elucidate the molecular basis for the regulation of Rag GTPases. In the structure, p18 wraps around the three pairs of roadblock heterodimers to tandemly assemble them onto lysosomes. Cellular and in vitro analyses further demonstrate that p18 is required for Ragulator-Rag GTPase assembly and amino acid-dependent activation of mTORC1. These results establish p18 as a critical organizing scaffold for the Ragulator-Rag GTPase complex, which may provide a platform for nutrient sensing on lysosomes., mTORC1 activity is controlled through Rag GTPases, which are anchored to the lysosome through the Ragulator. Here, the authors give molecular insights into Ragulator-Rag GTPase assembly and present the crystal structures of the Ragulator alone and in complex with the RagA-C roadblock domains.
- Published
- 2017
- Full Text
- View/download PDF
24. Lamtor1 Is Critically Required for CD4
- Author
-
Takashi, Hosokawa, Tetsuya, Kimura, Shigeyuki, Nada, Tatsusada, Okuno, Daisuke, Ito, Sujin, Kang, Satoshi, Nojima, Kazuya, Yamashita, Takeshi, Nakatani, Yoshitomo, Hayama, Yasuhiro, Kato, Yuhei, Kinehara, Masayuki, Nishide, Norihisa, Mikami, Syohei, Koyama, Hyota, Takamatsu, Daisuke, Okuzaki, Naganari, Ohkura, Shimon, Sakaguchi, Masato, Okada, and Atsushi, Kumanogoh
- Subjects
Mice, Knockout ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,TOR Serine-Threonine Kinases ,Cell Differentiation ,Mechanistic Target of Rapamycin Complex 1 ,Lipid Metabolism ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Mice, Inbred C57BL ,Mice ,T-Lymphocyte Subsets ,Multiprotein Complexes ,Animals ,Humans ,Interleukin-2 ,Th17 Cells ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Cell Proliferation - Abstract
Mechanistic target of rapamycin complex (mTORC)1 integrates intracellular sufficiency of nutrients and regulates various cellular functions. Previous studies using mice with conditional knockout of mTORC1 component proteins (i.e., mTOR, Raptor, and Rheb) gave conflicting results on the roles of mTORC1 in CD4
- Published
- 2017
25. p18/Lamtor1-mTORC1 Signaling Controls Development of Mucin-producing Goblet Cells in the Intestine.
- Author
-
Shizuka Ito, Shigeyuki Nada, Daisuke Yamazaki, Tetsuya Kimura, Kentaro Kajiwara, Hiroaki Miki, and Masato Okada
- Published
- 2020
- Full Text
- View/download PDF
26. Polarization of M2 macrophages requires Lamtor1 that integrates cytokine and amino-acid signals
- Author
-
Yoshitomo Hayama, Noriko Takegahara, Takashi Hosokawa, Daisuke Okuzaki, Natsuki Sakurai, Daisuke Ito, Hana Sarashina-Kida, Hyota Takamatsu, Tetsuya Kimura, Yuichi Mitsui, Satoshi Nojima, Keiko Morimoto, Yohei Maeda, Masayuki Nishide, Tatsusada Okuno, Sujin Kang, Masaki Yamada, Atsushi Kumanogoh, Shigeyuki Nada, and Masato Okada
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Multidisciplinary ,Science ,medicine.medical_treatment ,General Physics and Astronomy ,Signal transducing adaptor protein ,General Chemistry ,mTORC1 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Amino acid ,03 medical and health sciences ,030104 developmental biology ,Cytokine ,chemistry ,Biochemistry ,medicine ,Macrophage ,Signal transduction ,Liver X receptor ,PI3K/AKT/mTOR pathway - Abstract
Macrophages play crucial roles in host defence and tissue homoeostasis, processes in which both environmental stimuli and intracellularly generated metabolites influence activation of macrophages. Activated macrophages are classified into M1 and M2 macrophages. It remains unclear how intracellular nutrition sufficiency, especially for amino acid, influences on macrophage activation. Here we show that a lysosomal adaptor protein Lamtor1, which forms an amino-acid sensing complex with lysosomal vacuolar-type H+-ATPase (v-ATPase), and is the scaffold for amino acid-activated mTORC1 (mechanistic target of rapamycin complex 1), is critically required for M2 polarization. Lamtor1 deficiency, amino-acid starvation, or inhibition of v-ATPase and mTOR result in defective M2 polarization and enhanced M1 polarization. Furthermore, we identified liver X receptor (LXR) as the downstream target of Lamtor1 and mTORC1. Production of 25-hydroxycholesterol is dependent on Lamtor1 and mTORC1. Our findings demonstrate that Lamtor1 plays an essential role in M2 polarization, coupling immunity and metabolism.
- Published
- 2016
- Full Text
- View/download PDF
27. The late endosome/lysosome-anchored p18-mTORC1 pathway controls terminal maturation of lysosomes
- Author
-
Shigeyuki Nada, Taeko Soma-Nagae, Yusuke Takahashi, Masato Okada, Chitose Oneyama, and Shunsuke Mori
- Subjects
endocrine system ,endocrine system diseases ,Endosome ,Biophysics ,Endosomes ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Biochemistry ,Gene Knockout Techniques ,Mice ,Lysosome ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Late endosome ,Adaptor Proteins, Signal Transducing ,LAMP1 ,TOR Serine-Threonine Kinases ,Lysosome-Associated Membrane Glycoproteins ,Proteins ,rab7 GTP-Binding Proteins ,Cell Biology ,Cell biology ,medicine.anatomical_structure ,Membrane protein ,rab GTP-Binding Proteins ,Multiprotein Complexes ,Lysosomes ,Intracellular ,Biogenesis - Abstract
The late endosome/lysosome membrane adaptor p18 (or LAMTOR1) serves as an anchor for the mammalian target of rapamycin complex 1 (mTORC1) and is required for its activation on lysosomes. The loss of p18 causes severe defects in cell growth as well as endosome dynamics, including membrane protein transport and lysosome biogenesis. However, the mechanisms underlying these effects on lysosome biogenesis remain unknown. Here, we show that the p18-mTORC1 pathway is crucial for terminal maturation of lysosomes. The loss of p18 causes aberrant intracellular distribution and abnormal sizes of late endosomes/lysosomes and an accumulation of late endosome specific components, including Rab7, RagC, and LAMP1; this suggests that intact late endosomes accumulate in the absence of p18. These defects are phenocopied by inhibiting mTORC1 activity with rapamycin. Loss of p18 also suppresses the integration of late endosomes and lysosomes, resulting in the defective degradation of tracer proteins. These results suggest that the p18-mTORC1 pathway plays crucial roles in the late stages of lysosomal maturation, potentially in late endosome–lysosome fusion, which is required for processing of various macromolecules.
- Published
- 2012
- Full Text
- View/download PDF
28. The guanine nucleotide exchange factor Arhgef5 plays crucial roles in Src-induced podosome formation
- Author
-
Chitose Oneyama, Miho Kuroiwa, Masato Okada, and Shigeyuki Nada
- Subjects
rho GTP-Binding Proteins ,RHOA ,Podosome ,GTPase ,SH3 domain ,src Homology Domains ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Adhesion ,Animals ,Guanine Nucleotide Exchange Factors ,cdc42 GTP-Binding Protein ,biology ,Cell Biology ,Cell biology ,Enzyme Activation ,src-Family Kinases ,Cdc42 GTP-Binding Protein ,Invadopodia ,NIH 3T3 Cells ,biology.protein ,Cell Surface Extensions ,Guanine nucleotide exchange factor ,rhoA GTP-Binding Protein ,Rho Guanine Nucleotide Exchange Factors ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Podosomes and invadopodia are actin-rich membrane protrusions that play a crucial role in cell adhesion and migration, and extracellular matrix remodeling in normal and cancer cells. The formation of podosomes and invadopodia is promoted by upregulation of some oncogenic molecules and is closely related to the invasive potential of cancer cells. However, the molecular mechanisms underlying the podosome and invadopodium formation still remain unclear. Here, we show that a guanine nucleotide exchange factor (GEF) for Rho family GTPases (Arhgef5) is crucial for Src-induced podosome formation. Using an inducible system for Src activation, we found that Src-induced podosome formation depends upon the Src SH3 domain, and identified Arhgef5 as a Src SH3-binding protein. RNA interference (RNAi)-mediated depletion of Arhgef5 caused robust inhibition of Src-dependent podosome formation. Overexpression of Arhgef5 promoted actin stress fiber remodeling through activating RhoA, and the activation of RhoA or Cdc42 was required for Src-induced podosome formation. Arhgef5 was tyrosine-phosphorylated by Src and bound to Src to positively regulate its activity. Furthermore, the pleckstrin homology (PH) domain of Arhgef5 was required for podosome formation, and Arhgef5 formed a ternary complex with Src and phosphoinositide 3-kinase when Src and/or Arhgef5 were upregulated. These findings provide novel insights into the molecular mechanisms of podosome and invadopodium formation induced by Src upregulation.
- Published
- 2011
- Full Text
- View/download PDF
29. Ragulator-Rag Complex Targets mTORC1 to the Lysosomal Surface and Is Necessary for Its Activation by Amino Acids
- Author
-
Yasemin Sancak, Roberto Zoncu, Shigeyuki Nada, Liron Bar-Peled, David M. Sabatini, Andrew L. Markhard, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Sancak, Yasemin, Bar-Peled, Liron, Zoncu, Roberto, Markhard, Andrew L., and Sabatini, David M.
- Subjects
GTPase ,mTORC1 ,Plasma protein binding ,0302 clinical medicine ,Amino Acids ,chemistry.chemical_classification ,0303 health sciences ,biology ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,Signal transducing adaptor protein ,Ragulator complex ,Recombinant Proteins ,3. Good health ,Cell biology ,Amino acid ,Protein Transport ,Biochemistry ,030220 oncology & carcinogenesis ,Drosophila ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,Signal Transduction ,RHEB ,Cell signaling ,MAP Kinase Signaling System ,Mechanistic Target of Rapamycin Complex 1 ,Protein Serine-Threonine Kinases ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Lysosomal-Associated Membrane Protein 2 ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,030304 developmental biology ,Biochemistry, Genetics and Molecular Biology(all) ,Neuropeptides ,Lysosome-Associated Membrane Glycoproteins ,Proteins ,Intracellular Membranes ,Regulatory-Associated Protein of mTOR ,chemistry ,Multiprotein Complexes ,Mutation ,biology.protein ,RNA ,Ras Homolog Enriched in Brain Protein ,CELLBIO ,Lysosomes ,Transcription Factors - Abstract
The mTORC1 kinase promotes growth in response to growth factors, energy levels, and amino acids, and its activity is often deregulated in disease. The Rag GTPases interact with mTORC1 and are proposed to activate it in response to amino acids by promoting mTORC1 translocation to a membrane-bound compartment that contains the mTORC1 activator, Rheb. We show that amino acids induce the movement of mTORC1 to lysosomal membranes, where the Rag proteins reside. A complex encoded by the MAPKSP1, ROBLD3, and c11orf59 genes, which we term Ragulator, interacts with the Rag GTPases, recruits them to lysosomes, and is essential for mTORC1 activation. Constitutive targeting of mTORC1 to the lysosomal surface is sufficient to render the mTORC1 pathway amino acid insensitive and independent of Rag and Ragulator, but not Rheb, function. Thus, Rag-Ragulator-mediated translocation of mTORC1 to lysosomal membranes is the key event in amino acid signaling to mTORC1., National Institutes of Health (U.S.) (Grant CA103866), National Institutes of Health (U.S.) (Grant AI47389), United States. Dept. of Defense (W81XWH-07-0448), W. M. Keck Foundation, Jane Coffin Childs Memorial Fund for Medical Research, LAM Foundation (Fellowship)
- Published
- 2010
- Full Text
- View/download PDF
30. ZO-1- and ZO-2-Dependent Integration of Myosin-2 to Epithelial Zonula Adherens
- Author
-
Kazuaki Umeda, Shigeyuki Nada, Shoichiro Tsukita, Yuji Yamazaki, Sachiko Tsukita, Masami Wada, and Masato Okada
- Subjects
RHOA ,Pyridines ,PDZ domain ,Myosins ,Biology ,Zonula Occludens-2 Protein ,Models, Biological ,Epithelium ,Adherens junction ,Mice ,Myosin ,Fluorescence Resonance Energy Transfer ,Animals ,Molecular Biology ,Actin ,Cadherin ,Membrane Proteins ,Epithelial Cells ,Adherens Junctions ,Articles ,Cell Biology ,Transfection ,Cadherins ,Phosphoproteins ,Amides ,Actins ,Rats ,Cell biology ,Gene Expression Regulation ,Zonula Occludens-1 Protein ,biology.protein - Abstract
For the zonula adherens (ZA) to be established by linear arrangement of adherens junctions (AJs) in epithelial sheet cells, critical for the epithelial cell sheet formation and intercellular barrier function, myosin-2 is supposedly integrated into the ZA with the result of overlapping localization of E-cadherin/actin/myosin-2. Here, we immunofluorescently showed that myosin-2 failed to be integrated into the ZA in cultured epithelial-type ZO1(ko)/2(kd) Eph4 cells lacking ZO-1 and -2 (zonula occludens-1 and -2) by knockout and knockdown, respectively. Instead, a linearized but fragmented arrangement of AJs was formed in the way that it was positive for E-cadherin/actin, but negative for myosin-2 (designated prezonula-AJ). Transfection of full-length ZO-1 or ZO-2, or ZO-1 lacking its PDZ (PSD-95/discs large/zonula occludens-1)-1/2 domains (but not one lacking PDZ-1/2/3) into ZO1(ko)/2(kd) Eph4 cells restored the junctional integration of myosin-2 with prezonula-AJ to establish the ZA. Transfection of dominant-active RhoA or Rho-kinase (ROCK), as well as administration of lysophosphatidic acid or Y27632, which activates RhoA or inhibits ROCK, respectively, suggested that RhoA regulated the junctional integration of myosin-2 into ZA in a manner such that ROCK played a necessary but not-sufficient role. Fluorescence resonance energy transfer analyses revealed that spatiotemporal Rho-activation occurred in a ZO-1/2–dependent way to establish ZA from primordial forms in epithelial cells.
- Published
- 2008
- Full Text
- View/download PDF
31. The Lipid Raft-Anchored Adaptor Protein Cbp Controls the Oncogenic Potential of c-Src
- Author
-
Chitose Oneyama, Shigeyuki Nada, Kazunobu Saito, Tomoya Hikita, Marc-Werner Dobenecker, Masato Okada, Alexander Tarakhovsky, and Kengo Enya
- Subjects
Mice, Nude ,Biology ,medicine.disease_cause ,Cell Fractionation ,environment and public health ,CSK Tyrosine-Protein Kinase ,Mice ,Membrane Microdomains ,Downregulation and upregulation ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Lipid raft ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,Mice, Inbred BALB C ,Gene Transfer Techniques ,Signal transducing adaptor protein ,Membrane Proteins ,Cell Biology ,Cell cycle ,Fibroblasts ,Protein-Tyrosine Kinases ,Phosphoproteins ,Molecular biology ,Cell biology ,Cell Transformation, Neoplastic ,src-Family Kinases ,Phosphorylation ,Carcinogenesis ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The tyrosine kinase c-Src is upregulated in various human cancers irrespective of its negative regulator Csk, but the regulatory mechanisms remain unclear. Here, we show that a lipid raft-anchored Csk adaptor, Cbp/PAG, is directly involved in controlling the oncogenicity of c-Src. Using Csk-deficient cells that can be transformed by c-Src overexpression, we found that Cbp expression is markedly downregulated by c-Src activation and re-expression of Cbp efficiently suppresses c-Src transformation as well as tumorigenesis. Cbp-deficient cells are more susceptible to v-Src transformation than their parental cells. Upon phosphorylation, Cbp specifically binds to activated c-Src and sequesters it in lipid rafts, resulting in an efficient suppression of c-Src function independent of Csk. In some human cancer cells and tumors, Cbp is downregulated and the introduction of Cbp significantly suppresses tumorigenesis. These findings indicate a potential role for Cbp as a suppressor of c-Src-mediated tumor progression.
- Published
- 2008
- Full Text
- View/download PDF
32. Ablation of Csk in neural crest lineages causes corneal anomaly by deregulating collagen fibril organization and cell motility
- Author
-
Masato Okada, Reiko Yagi, Yasuo Uchiyama, Chitose Oneyama, Shigeyuki Nada, Masato Koike, Atsuko Takatsuka, and Christian Schmedt
- Subjects
Corneal endothelium ,Stromal cell ,Motility ,RAC1 ,Mice, Transgenic ,CDC42 ,Cell motility ,Src family kinases ,Biology ,Collagen fibril organization ,CSK Tyrosine-Protein Kinase ,Mesoderm ,Cornea ,Mice ,Cell Movement ,Csk ,Fluorescence Resonance Energy Transfer ,Animals ,Cdc42 ,Promoter Regions, Genetic ,Molecular Biology ,Mice, Knockout ,Tyrosine-protein kinase CSK ,Retinoblastoma-Like Protein p130 ,Neural crest ,Cas ,Cell Biology ,Protein-Tyrosine Kinases ,Mice, Mutant Strains ,eye diseases ,Cell biology ,Cre–loxP ,Protein zero ,Phenotype ,src-Family Kinases ,Neural Crest ,Immunology ,Cell polarity ,RNA Interference ,Collagen ,Myelin P0 Protein ,Rac1 ,Developmental Biology - Abstract
Src family kinases (SFKs) have been implicated in the regulation of cell motility. To verify their in vivo roles during development, we generated mutant mice in which Csk, a negative regulator of SFKs, was inactivated in neural crest lineages using the Protein zero promoter in a Cre-loxP system. Inactivation of Csk caused deformities in various tissues of neural crest origins, including facial dysplasia and corneal opacity. In the cornea, the stromal collagen fibril was disorganized and there was an overproduction of collagen 1a1 and several metalloproteases. The corneal endothelium failed to overlie the central region of the eye and the peripheral endothelium displayed a disorganized cytoskeleton. Corneal mesenchymal cells cultured from mutant mice showed attenuated cell motility. In these cells, p130 Crk-associated substrate (Cas) was hyperphosphorylated and markedly downregulated. The expression of a dominant negative Cas (Cas Delta SD) could suppress the cell motility defects. Fluorescence resonance energy transfer analysis revealed that activation of Rac1 and Cdc42 was depolarized in Csk-inactivated cells, which was restored by the expression of either Csk or Cas Delta SD. These results demonstrate that the SFKs/Csk circuit plays crucial roles in corneal development by controlling stromal organization and by ensuring cell motility via the Cas-Rac/Cdc42 pathways.
- Published
- 2008
- Full Text
- View/download PDF
33. Functional dissection of transformation by c-Src and v-Src
- Author
-
Tomoya Hikita, Masato Okada, Chitose Oneyama, and Shigeyuki Nada
- Subjects
Kinase ,Angiogenesis ,Cell ,Cell Biology ,Biology ,Cyclin D1 ,medicine.anatomical_structure ,Tumor progression ,v-Src ,Immunology ,Genetics ,biology.protein ,medicine ,Cancer research ,STAT3 ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The c-src proto-oncogene product, c-Src, is frequently over-expressed and activated in various human malignant cancers, implicating a role for c-Src in cancer progression. To verify the role of c-Src, we analyzed the transforming ability of c-Src in mouse embryonic fibroblasts that lack Csk, a negative regulator of Src family kinases. Although Csk deficiency is not sufficient for cell transformation, c-Src over-expression induced characteristic transformed phenotypes including anchorage-independent growth and tumorigenecity. These phenotypes were dose-dependently inhibited by the re-expression of Csk, indicating that there is a certain threshold for c-Src transformation, which is determined by the c-Src : Csk ratio. In contrast to v-Src, c-Src induced the phosphorylation of a limited number of cellular proteins and elicited a restricted change in gene expression profiles. The activation of some critical targets for v-Src transformation, such as STAT3, was not significantly induced by c-Src transformation. Several genes that are involved in cancer progression, that is, cyclin D1 and HIF-1α, were induced by v-Src, but not by c-Src. Furthermore, v-Src tumors exhibited aggressive growth and extensive angiogenesis, while c-Src tumors grew more slowly accompanied by the induction of hematomas. These findings demonstrate that c-Src has the potential to induce cell transformation, but it requires coordination with an additional pathway(s) to promote tumor progression in vivo.
- Published
- 2007
- Full Text
- View/download PDF
34. Tyrosine phosphorylation controls cortactin binding to two enterohaemorrhagic Escherichia coli effectors: Tir and EspFu/TccP
- Author
-
Takeshi Honda, Masato Okada, Tetsuya Iida, Niels Nijstad, Toshio Kodama, Vlademir Vicente Cantarelli, Said K. Abolghait, and Shigeyuki Nada
- Subjects
Brush border ,Virulence Factors ,Immunology ,Virulence ,Receptors, Cell Surface ,macromolecular substances ,Biology ,Escherichia coli O157 ,Microbiology ,Cell Line ,Dephosphorylation ,chemistry.chemical_compound ,Two-Hybrid System Techniques ,hemic and lymphatic diseases ,Virology ,Humans ,Phosphorylation ,Pathogen ,Actin ,Effector ,Escherichia coli Proteins ,Intracellular Signaling Peptides and Proteins ,Tyrosine phosphorylation ,Fibroblasts ,Actins ,Cell biology ,chemistry ,biology.protein ,Tyrosine ,bacteria ,Carrier Proteins ,Cortactin ,HeLa Cells - Abstract
Summary Enterohaemorrhagic Escherichia coli (EHEC) is an important food-borne pathogen that, upon infection, causes destruction of the microvilli brush border of intestinal cells. EHEC is able to recruit several host cell proteins and induce actin accumulation beneath its adherence site, forming a pedestal-like structure upon which the bacterium is firmly attached. Injection of bacterial effectors into the host cells is required to trigger the recruitment and activation of proteins, such as cortactin, neural Wiskott–Aldrich syndrome protein (N-WASP) and Arp2/3 complex, directly involved in the actin polymerization process. We found that cortactin, an actin-binding protein, has a pivotal role during pedestal formation by EHEC. Cortactin was found to bind directly to two important virulence factors of EHEC, Tir and EspFu, which are translocated into the host cells during infection. Binding of cortactin to these effectors is dependent upon tyrosine phosphorylation and a balance between tyrosine phosphorylation and dephosphorylation of cortactin is required to regulate pedestal formation by EHEC.
- Published
- 2007
- Full Text
- View/download PDF
35. C-terminal Src kinase controls development and maintenance of mouse squamous epithelia
- Author
-
Masato Okada, Yasuo Uchiyama, Chitose Oneyama, Shigeyuki Nada, Satoshi Itami, Christian Schmedt, Satoshi Waguri, Yasuyuki Sumikawa, and Reiko Yagi
- Subjects
Keratinocytes ,Immunoblotting ,Mice, Transgenic ,RAC1 ,Biology ,Tacrolimus ,Article ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,CSK Tyrosine-Protein Kinase ,Mice ,Esophagus ,Microscopy, Electron, Transmission ,Cell Adhesion ,Animals ,Src family kinase ,Cell adhesion ,Molecular Biology ,Cells, Cultured ,Cytoskeleton ,Skin ,Inflammation ,Mice, Knockout ,Tyrosine-protein kinase CSK ,General Immunology and Microbiology ,Tumor Necrosis Factor-alpha ,General Neuroscience ,Protein-Tyrosine Kinases ,Immunohistochemistry ,Matrix Metalloproteinases ,Cell biology ,Keratin 5 ,Phenotype ,src-Family Kinases ,Mutation ,Cancer research ,Keratin-5 ,Signal transduction ,Immunosuppressive Agents ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Carboxy-terminal Src kinase (Csk) is a negative regulator of Src family kinases, which play pivotal roles in controlling cell adhesion, migration, and cancer progression. To elucidate the in vivo role of Csk in epithelial tissues, we conditionally inactivated Csk in squamous epithelia using the keratin-5 promoter/Cre-loxP system in mice. The mutant mice developed apparent defects in the skin, esophagus, and forestomach, with concomitant hyperplasia and chronic inflammation. Histology of the mutant epidermis revealed impaired cell-cell adhesion in basal cell layers. Analysis of primary keratinocytes showed that the defective cell-cell adhesion was caused by cytoskeletal remodeling via activation of the Rac1 pathway. Mutant keratinocytes also showed elevated expression of mesenchymal proteins, matrix metalloproteinases (MMPs), and the proinflammatory cytokine TNF-alpha. Inhibition of the expression of TNF-alpha and MMP9 by the anti-inflammatory reagent FK506 could cure the epidermal hyperplasia, suggesting a causal link between inflammation and epidermal hyperplasia. These observations demonstrate that the Src/Csk circuit plays crucial roles in development and maintenance of epithelia by controlling cytoskeletal organization as well as phenotypic conversion linked to inflammatory events.
- Published
- 2007
- Full Text
- View/download PDF
36. Constitutive activation of neuronal Src causes aberrant dendritic morphogenesis in mouse cerebellar Purkinje cells
- Author
-
Shigeki Yuasa, Shigeyuki Nada, Takenori Kotani, Masato Okada, and Nobuhiro Morone
- Subjects
Genetically modified mouse ,Calbindins ,Morphogenesis ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,SRC Family Tyrosine Kinase ,Proto-Oncogene Proteins c-fyn ,Mice ,Purkinje Cells ,S100 Calcium Binding Protein G ,FYN ,Microscopy, Electron, Transmission ,Microtubule ,Cerebellum ,Animals ,Cells, Cultured ,Kinase ,General Neuroscience ,Gene Expression Regulation, Developmental ,Dendrites ,General Medicine ,Immunohistochemistry ,Cell biology ,Mice, Inbred C57BL ,Genes, src ,Animals, Newborn ,Neural development ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Src family tyrosine kinases are essential for neural development, but their in vivo functions remain elusive because of functional compensation among family members. To elucidate the roles of individual Src family members in vivo, we generated transgenic mice expressing the neuronal form of c-Src (n-Src), Fyn, and their constitutively active forms in cerebellar Purkinje cells using the L7 promoter. The expression of the constitutively active n-Src retarded the postnatal development of Purkinje cells and disrupted dendritic morphogenesis, whereas the wild-type n-Src had only moderate effects. Neither wild-type nor constitutively active Fyn over-expression significantly affected Purkinje-cell morphology. The aberrant Purkinje cells in n-Src transgenic mice retained multiple dendritic shafts extending in non-polarized directions and were located heterotopically in the molecular layer. Ultrastructural observation of the dendritic shafts revealed that the microtubules of n-Src transgenic mice were more densely and irregularly arranged, and had structural deformities. In primary culture, Purkinje cells from n-Src transgenic mice developed abnormally thick dendritic shafts and large growth-cone-like structures with poorly extended dendrites, which could be rescued by treatment with a selective inhibitor of Src family kinases, PP2. These results suggest that n-Src activity regulates the dendritic morphogenesis of Purkinje cells through affecting microtubule organization.
- Published
- 2007
- Full Text
- View/download PDF
37. Disruption of the Paternal Necdin Gene Diminishes TrkA Signaling for Sensory Neuron Survival
- Author
-
Taichi Uetsuki, Shigeyuki Nada, Masato Okada, Kazuaki Yoshikawa, Masashi Yamada, Akari Hosokawa, Ken-ichiro Kuwako, and Isao Nishimura
- Subjects
Male ,Cell Survival ,Population ,Gene Expression ,Apoptosis ,Nerve Tissue Proteins ,Tropomyosin receptor kinase A ,PC12 Cells ,Receptor, Nerve Growth Factor ,Receptor tyrosine kinase ,Fathers ,Genomic Imprinting ,Mice ,Pregnancy ,Neurobiology of Disease ,Ganglia, Spinal ,medicine ,Animals ,Low-affinity nerve growth factor receptor ,Neurons, Afferent ,Receptor, trkA ,education ,Protein kinase A ,Mice, Knockout ,education.field_of_study ,biology ,General Neuroscience ,Nociceptors ,Nuclear Proteins ,Sensory neuron ,Rats ,Mice, Inbred C57BL ,Nerve growth factor ,medicine.anatomical_structure ,nervous system ,Mice, Inbred CBA ,biology.protein ,Cancer research ,Phosphorylation ,Female ,Prader-Willi Syndrome ,Signal Transduction - Abstract
Necdin is a multifunctional signaling protein that stabilizes terminal differentiation of postmitotic neurons. The human necdin gene in chromosome 15q11-q12 is maternally imprinted, paternally transcribed, and not expressed in Prader-Willi syndrome, a human genomic imprinting-associated neurodevelopmental disorder. Although necdin-deficient mice display several abnormal phenotypes reminiscent of this syndrome, little is known about molecular mechanisms that lead to the neurodevelopmental defects. Here, we demonstrate that paternally expressed necdin is required for physiological development of nerve growth factor (NGF)-dependent sensory neurons. Mouse embryos defective in the paternal necdin allele displayed absent necdin expression in the dorsal root ganglia, in which the tropomyosin-related kinase A (TrkA) receptor tyrosine kinase and the p75 neurotrophin receptor were expressed in a normal manner. Necdin interacted with both TrkA and p75 to facilitate the association between these receptors. NGF-induced phosphorylation of TrkA and mitogen-activated protein kinase was significantly diminished in the necdin-null sensory ganglia. Furthermore, the mice lacking the paternal necdin allele displayed augmented apoptosis in the sensory gangliain vivoand had a reduced population of substance P-containing neurons. These mutant mice showed significantly high tolerance to thermal pain, which is often seen in individuals with Prader-Willi syndrome. These results suggest that paternally expressed necdin facilitates TrkA signaling to promote the survival of NGF-dependent nociceptive neurons.
- Published
- 2005
- Full Text
- View/download PDF
38. ABCA5 Resides in Lysosomes, and ABCA5 Knockout Mice Develop Lysosomal Disease-Like Symptoms
- Author
-
Yoshiyuki Kubo, Akihito Yamaguchi, Tsuyoshi Nishi, Chiemi Takenaka, Megumi Ohigashi, Kyoko Tamura, Sayaka Sekiya, Shigeyuki Nada, and Akitsugu Yamamoto
- Subjects
Male ,Thyroid Hormones ,Subfamily ,Molecular Sequence Data ,Thyroid Gland ,ATP-binding cassette transporter ,ABCA8 ,Cell Line ,Mice ,Complementary DNA ,Mammalian Genetic Models with Minimal or Complex Phenotypes ,Animals ,Exophthalmos ,Humans ,Abnormalities, Multiple ,Amino Acid Sequence ,RNA, Messenger ,Cloning, Molecular ,Molecular Biology ,Mice, Knockout ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Myocardium ,Cell Biology ,Immunohistochemistry ,Molecular biology ,Rats ,Transport protein ,Cell biology ,Protein Transport ,Membrane protein ,ABCA1 ,Knockout mouse ,biology.protein ,ATP-Binding Cassette Transporters ,Female ,Lysosomes ,Gene Deletion - Abstract
ATP binding cassette (ABC) transporters are membrane proteins that are widely distributed in prokaryotes and eukaryotes, and most of them transport substrates across membranes (10, 12, 35). In general, they transport drugs, toxins, peptides, lipid derivatives, and so on, coupled with ATP hydrolysis at their nucleotide binding domains (NBDs) (12). NBDs are characterized by three motifs, Walker A, B, and C (signature) (35). In humans and rodents, ABC transporters are divided into seven subfamilies, A to G, and some of them are known to participate in physiological phenomena. In particular, subfamily A of ABC transporters is an interesting group that includes 13 members found in humans; some members of this group are known to transport lipids and their derivatives and to be related to genetic diseases (8). For example, ABCA1 has been reported to participate in cholesterol trafficking (18, 22), and a genetic defect of it causes Tangier disease, which is characterized clinically by the accumulation of cholesteryl ester in various tissues (9). ABCA1 is also involved in the engulfment of apoptotic cells by macrophages (17). ABCA4 (3, 13) is specifically expressed in the retina and transports N-retinylidene-phosphatidyl-ethanolamine (37), and a genetic defect of it causes Stargardt disease, which is clinically characterized by the progressive loss of central vision and progressive atrophy of the retinal pigment epithelium overlying the macula (1). These examples indicate that the physiological roles of subfamily A members are likely to differ from those of other ABC transporters, such as MDR1, for which knockout mice did not exhibit remarkable abnormalities without the administration of drugs and toxins (29). However, the functions and substrates have been determined for only two members of subfamily A. In order to determine the physiological roles of ABC proteins in the brain or their neuron-specific functions, we tried to identify a novel member of subfamily A in newborn mouse brain and neural cells derived from P19 cells by reverse transcription (RT)-PCR. As a result of full-length cDNA cloning, we found an orthologue of human ABCA5 (mABCA5). The human and rat ABCA5 cDNA have already been isolated, and their mRNAs have been reported to be expressed mainly in brain, lung, and testis (23). The human ABCA5 gene is located on chromosome 17q23.4 and forms a gene cluster with ABCA6, ABCA8, ABCA9, and ABCA10 (10). Similar gene clustering has been found on mouse chromosome 11 (2). However, further characterization of the ABCA5 protein has not been reported. In this study, we determined the subcellular localization of the ABCA5 protein and examined its functions after generating knockout mice.
- Published
- 2005
- Full Text
- View/download PDF
39. Potentiation of the ligand-binding activity of integrin α3β1 via association with tetraspanin CD151
- Author
-
Masato Okada, Shigeyuki Nada, Ryoko Nishiuchi, Junichi Takagi, Yasuhiro Sumida, Noriko Sanzen, Yoshinao Wada, Kiyotoshi Sekiguchi, and Hitoshi Hasegawa
- Subjects
Protein Conformation ,Placenta ,Integrin ,CD18 ,Tetraspanin 24 ,Ligands ,CD49c ,Collagen receptor ,Antigens, CD ,Cell Line, Tumor ,Chlorocebus aethiops ,Cell Adhesion ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Cell adhesion ,Multidisciplinary ,biology ,Integrin alpha3beta1 ,Antibodies, Monoclonal ,Biological Sciences ,Ligand (biochemistry) ,Molecular biology ,Cell biology ,Integrin alpha M ,Multiprotein Complexes ,COS Cells ,biology.protein ,RNA Interference ,Integrin, beta 6 ,Laminin ,Protein Binding - Abstract
CD151, one of the tetraspanins, forms a stable complex with integrin alpha3beta1, the major laminin receptor on the cell surface. We found that 8C3, an anti-CD151 mAb obtained by screening for reactivity with integrin alpha3beta1-CD151 complexes, was capable of dissociating CD151 from integrin alpha3beta1, thereby allowing us to deplete CD151 from purified integrin alpha3beta1-CD151 complexes. The CD151-free integrin alpha3beta1 thus obtained showed a significant reduction in its ability to bind to laminin-10/11, a high-affinity ligand for integrin alpha3beta1, with a concomitant reduction in its reactivity with mAb AG89, which recognizes activated beta1-containing integrins. These results raised the possibility that the association of integrin alpha3beta1 with CD151 potentiates the ligand-binding activity of the integrin through sustaining its activated conformation. In support of this possibility, the ligand-binding activity was restored when CD151-free integrin alpha3beta1 was reassociated with purified CD151. 8C3-induced dissociation of CD151 from integrin alpha3beta1 was also demonstrated on the surface of living cells by fluorescent resonance energy transfer imaging, accompanied by a concomitant reduction in the cell adhesion to laminin-10/11-coated substrates. CD151 knock-down by RNA interference also resulted in a reduction in the adhesive activity of the cells. Taken together, these results indicate that CD151 association modulates the ligand-binding activity of integrin alpha3beta1 through stabilizing its activated conformation not only with purified proteins but also in a physiological context.
- Published
- 2005
- Full Text
- View/download PDF
40. Anteriorization of neural fate by inhibitor of β-catenin and T cell factor (ICAT), a negative regulator of Wnt signaling
- Author
-
Susumu Ohwada, Shinji Takada, Kiyotoshi Satoh, Tetsu Akiyama, Takefumi Ishidao, Takao Senda, Shigeyuki Nada, Mana Kasai, Kenichi Tago, Yoshimi Hasegawa, and Tsutomu Nakamura
- Subjects
Male ,T cell ,Cellular differentiation ,Cell Cycle Proteins ,Biology ,Mice ,Prosencephalon ,Proto-Oncogene Proteins ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Neurons ,Multidisciplinary ,Stem Cells ,Wnt signaling pathway ,Cell Differentiation ,Biological Sciences ,Zebrafish Proteins ,Embryo, Mammalian ,Embryonic stem cell ,Cell biology ,Mice, Inbred C57BL ,Repressor Proteins ,Wnt Proteins ,medicine.anatomical_structure ,Catenin ,Female ,Stem cell ,WNT3A ,Signal Transduction ,Transcription Factors - Abstract
Inhibitor of β-catenin and T cell factor (ICAT) inhibits Wnt signaling by interfering with the interaction between β-catenin and T cell factor. Here we show that ICAT –/– embryos exhibit malformation of the forebrain and craniofacial bones and lack the kidney. Analysis of the neuronal differentiation of embryonic stem cells revealed that Wnt3a redirects the fate of neural progenitors to a posterior character, whereas ICAT induces forebrain cells by inhibiting Wnt signaling. Furthermore, ICAT –/– embryonic stem cells were found to differentiate into neuronal cells possessing a posterior character. These results suggest that ICAT plays an important role in the anteriorization of neural cells by inhibiting the posteriorizing activity of Wnt signaling.
- Published
- 2004
- Full Text
- View/download PDF
41. Mechanism of Csk-mediated Down-regulation of Src Family Tyrosine Kinases in Epidermal Growth Factor Signaling
- Author
-
Shigeyuki Nada, Hidetada Matsuoka, and Masato Okada
- Subjects
Scaffold protein ,Yellow fluorescent protein ,Time Factors ,Green Fluorescent Proteins ,Immunoblotting ,Down-Regulation ,In Vitro Techniques ,Biology ,SRC Family Tyrosine Kinase ,Transfection ,Models, Biological ,Biochemistry ,CSK Tyrosine-Protein Kinase ,Epidermal growth factor ,Fluorescence Resonance Energy Transfer ,Animals ,Phosphorylation ,Molecular Biology ,Binding Sites ,Tyrosine-protein kinase CSK ,Epidermal Growth Factor ,Cell Membrane ,Cell Biology ,Protein-Tyrosine Kinases ,Precipitin Tests ,Fusion protein ,Protein Structure, Tertiary ,Cell biology ,Luminescent Proteins ,src-Family Kinases ,Microscopy, Fluorescence ,Spectrophotometry ,COS Cells ,Mutation ,biology.protein ,Signal transduction ,Plasmids ,Signal Transduction - Abstract
The Src family tyrosine kinases (SFKs) play pivotal roles as molecular switches that link a variety of extracellular cues to intracellular signaling pathway. The function of SFK is regulated by phosphorylation at the C-terminal regulatory site mediated by Csk. Recently a novel SFK target Cbp (or PAG) was identified as a membrane-anchored scaffold protein for Csk. To establish the mechanism of Csk/Cbp-mediated regulation of SFK in vivo, we observed dynamic changes in the interaction of Csk with Cbp by utilizing fusion proteins with modified green fluorescent proteins: cyan fluorescent protein (CFP) or yellow fluorescent protein (YFP). Upon SFK activation induced by epidermal growth factor stimulation, fluorescent resonance energy transfer (FRET) response was detected transiently at membrane ruffles in COS1 cells co-expressing CFP-Csk and Cbp-YFP and in cells expressing a single-molecule FRET indicator consisting of CskSH2 and Cbp. Suppression of SFK by PP2 or use of a mutant Cbp that lacks the Csk binding site abolished the FRET response, although a dominant-negative form of Csk enhanced and sustained the FRET response, demonstrating that the FRET response is dependent upon the SFK activity. These observations show that Csk/Cbp-mediated down-regulation of SFK takes place at membrane ruffles in an early stage of epidermal growth factor signaling and suggest that the Csk/Cbp-based FRET indicators are useful for monitoring the status of SFK in living cells.
- Published
- 2004
- Full Text
- View/download PDF
42. Csk defines the ability of integrin-mediated cell adhesion and migration in human colon cancer cells: implication for a potential role in cancer metastasis
- Author
-
Masato Okada, Akio Konishi, Takao Yamori, Naoki Morishita, Shigeyuki Nada, Hidetada Matsuoka, and William Rengifo-Cam
- Subjects
Integrins ,Cancer Research ,medicine.medical_specialty ,Integrin ,Cell Communication ,Protein tyrosine phosphatase ,CSK Tyrosine-Protein Kinase ,Cell Movement ,Proto-Oncogene Proteins ,Internal medicine ,Cell Adhesion ,Genetics ,medicine ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Cell adhesion ,Molecular Biology ,beta Catenin ,Tyrosine-protein kinase CSK ,biology ,Phosphotransferases ,Protein-Tyrosine Kinases ,Cadherins ,Actin cytoskeleton ,Actins ,Cell biology ,Cytoskeletal Proteins ,src-Family Kinases ,Endocrinology ,Colonic Neoplasms ,Cancer cell ,Trans-Activators ,biology.protein ,Phosphorylation ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Progression of human colon cancer is often associated with elevated expression and activity of the Src family tyrosine kinase (SFK). SFK is ordinarily in equilibrium between inactive and primed states by a balance of negative regulatory kinase Csk and its counteracting tyrosine phosphatase(s), both of which act on the regulatory C-terminal tyrosine of SFK. To evaluate the contribution of the regulatory system of SFK in cancer progression, we here modulated the equilibrium status of SFK by introducing wild-type or dominant-negative Csk in human epithelial colon cancer cells, HCT15 and HT29. Overexpression of wild-type Csk induced decreased SFK activation, increased cell-cell contacts mediated by E-cadherin, decreased the number of focal contacts and decreased cell adhesion/migration and in vitro invasiveness. Conversely, expression of a dominant-negative Csk resulted in elevated SFK activation, enhanced phosphorylation of FAK and paxilllin, enhanced cell scattering, an increased number of focal contacts, dramatic rearrangement of actin cytoskeleton and increased cell adhesion/migration and in vitro invasiveness. In these scattered cells, however, localization, expression and phosphorylation of either E-cadherin or beta-catenin were not significantly affected, suggesting that the E-cadherin-mediated cell-cell contact is indirectly regulated by SFK. Furthermore, all these events occurred absolutely dependent on integrin-mediated cell adhesion. These findings demonstrate that Csk defines the ability of integrin-SFK-mediated cell adhesion signaling that influences the metastatic potential of cancer cells.
- Published
- 2004
- Full Text
- View/download PDF
43. Transmembrane phosphoprotein Cbp senses cell adhesion signaling mediated by Src family kinase in lipid rafts
- Author
-
Takaki Shima, Shigeyuki Nada, and Masato Okada
- Subjects
Integrins ,DNA, Complementary ,Time Factors ,Integrin ,Biology ,environment and public health ,Culture Media, Serum-Free ,Cell membrane ,Focal adhesion ,Mice ,Membrane Microdomains ,FYN ,Cell Movement ,Cell Adhesion ,medicine ,Animals ,Src family kinase ,Phosphorylation ,Cell adhesion ,Cytoskeleton ,Multidisciplinary ,Cell Membrane ,Brain ,Membrane Proteins ,Biological Sciences ,Lipid Metabolism ,Phosphoproteins ,Fibronectins ,Cell biology ,Cell Adhesion Process ,src-Family Kinases ,medicine.anatomical_structure ,biology.protein ,Tyrosine ,RNA Interference ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Cbp, a C-terminal Src kinase (Csk)-binding protein, is a transmembrane phosphoprotein that has been implicated in the regulation of the Src family kinase (SFK) through recruiting Csk, a negative regulator of SFK, to a membrane microdomain of lipid rafts. To examine the contribution of Cbp to cell adhesion signaling mediated by SFK, we investigated the kinase responsible for phosphorylating Cbp and the mode of phosphorylation during the cell adhesion process. The results obtained by using mutant mice or cells that lack Csk and/or a member of SFK, Fyn, reveal that Cbp is phosphorylated predominantly by raft-localized Fyn in vivo . Upon cell adhesion onto fibronectin, Cbp becomes transiently phosphorylated (consistent with SFK activation) and recruits Csk to lipid rafts. These events are completed before the full activation of focal adhesion kinase, indicating that the transient activation and down-regulation of SFK in lipid rafts are earlier events in cell adhesion signaling. In Csk-deficient cells, continuous hyperactivation of SFK leads to continuous hyperphosphorylation of Cbp, accompanied by impaired cell spreading and migration. Silencing of Cbp by RNA interference also induced impaired cell spreading. These findings suggest that Cbp could serve as a sensor of SFK activity in early stages of cell adhesion signaling, and that Csk-mediated down-regulation of SFK is essential to allow dynamic cellular events involved in the regulation of cell spreading and migration.
- Published
- 2003
- Full Text
- View/download PDF
44. Cloning of rat ABCA7 and its preferential expression in platelets
- Author
-
Mari Sasaki, Akihito Yamaguchi, Yoshiyuki Kubo, Shigeyuki Nada, and Ayako Shoji
- Subjects
Blood Platelets ,medicine.drug_class ,Biophysics ,CHO Cells ,Biology ,Monoclonal antibody ,Biochemistry ,Epitope ,Western blot ,Cricetinae ,Complementary DNA ,medicine ,Animals ,Humans ,Tissue Distribution ,Cloning, Molecular ,Rats, Wistar ,Molecular Biology ,Peptide sequence ,chemistry.chemical_classification ,Molecular mass ,medicine.diagnostic_test ,Antibodies, Monoclonal ,Cell Biology ,Molecular biology ,Rats ,Amino acid ,Epitope mapping ,chemistry ,Multigene Family ,ATP-Binding Cassette Transporters ,Female ,Epitope Mapping - Abstract
We cloned the full-length cDNA of a rat orthologue of ABCA7 (rABCA7) from rat platelets. The cDNA of rABCA7 is 6510bp in length and encodes a protein of 2170 amino acids. The amino acid sequence of rABCA7 exhibits homology to those of mouse ABCA7 (92.5% identical in amino acid sequence) and human ABCA7 (76.6%). We obtained two clones of monoclonal antibodies against rABCA7 recognizing different epitopes. Analysis of CHO cells stably expressing rABCA7 by confocal laser-scanning microscopy indicated that rABCA7 is mainly located in the plasma membrane. Western blot analysis of rat tissues revealed that rABCA7 was preferentially expressed in platelets and that its apparent molecular mass was 250kDa. This is the first report of the tissue distribution of rABCA7 at the protein level and is the first reported case of ABC transporters being expressed in platelets, suggesting their important role in platelet function.
- Published
- 2003
- Full Text
- View/download PDF
45. p18/LAMTOR1
- Author
-
Shigeyuki Nada, Yusuke Takahashi, Masato Okada, and Shunsuke Mori
- Subjects
MAPK/ERK pathway ,medicine.anatomical_structure ,Endosome ,Lysosome ,Autophagy ,medicine ,mTORC1 ,GTPase ,Biology ,PI3K/AKT/mTOR pathway ,Late endosome ,Cell biology - Abstract
p18/LAMTOR1 is a membrane protein specifically localized to the surface of late endosomes/lysosomes that serves as an anchor for the "Ragulator" complex, which contains p14/LAMTOR2, MP1/LAMTOR3, HBXIP, and C7orf59. The Ragulator interacts with RagAB/CD GTPases and V-ATPase and plays crucial roles for activation of mammalian target of rapamycin complex 1 (mTORC1) on the lysosomal surface. Activated mTORC1 orchestrates various cellular functions, for example, macromolecule biosynthesis, energy metabolism, autophagy, cell growth, responses to growth factors, and the trafficking and maturation of lysosomes. The Ragulator can also regulate a branch of the MAPK pathway by recruiting MEK1 to MP1/LAMTOR3. These findings suggest that p18/LAMTOR1 creates a core platform for intracellular signaling pathways that function via late endosomes/lysosomes.
- Published
- 2014
- Full Text
- View/download PDF
46. p18/LAMTOR1: a late endosome/lysosome-specific anchor protein for the mTORC1/MAPK signaling pathway
- Author
-
Shigeyuki, Nada, Shunsuke, Mori, Yusuke, Takahashi, and Masato, Okada
- Subjects
Mice, Knockout ,MAP Kinase Signaling System ,Recombinant Fusion Proteins ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,Mechanistic Target of Rapamycin Complex 1 ,Cell Fractionation ,PC12 Cells ,Rats ,Luminescent Proteins ,Mice ,Protein Transport ,Membrane Microdomains ,Microscopy, Fluorescence ,Multiprotein Complexes ,Protein Interaction Mapping ,Animals ,Humans ,Carrier Proteins ,Lysosomes ,Adaptor Proteins, Signal Transducing ,Signal Transduction - Abstract
p18/LAMTOR1 is a membrane protein specifically localized to the surface of late endosomes/lysosomes that serves as an anchor for the "Ragulator" complex, which contains p14/LAMTOR2, MP1/LAMTOR3, HBXIP, and C7orf59. The Ragulator interacts with RagAB/CD GTPases and V-ATPase and plays crucial roles for activation of mammalian target of rapamycin complex 1 (mTORC1) on the lysosomal surface. Activated mTORC1 orchestrates various cellular functions, for example, macromolecule biosynthesis, energy metabolism, autophagy, cell growth, responses to growth factors, and the trafficking and maturation of lysosomes. The Ragulator can also regulate a branch of the MAPK pathway by recruiting MEK1 to MP1/LAMTOR3. These findings suggest that p18/LAMTOR1 creates a core platform for intracellular signaling pathways that function via late endosomes/lysosomes.
- Published
- 2014
47. Complete Cysteine-scanning Mutagenesis and Site-directed Chemical Modification of the Tn10-encoded Metal-Tetracycline/H+ Antiporter
- Author
-
Tomomi Kimura-Someya, Akihito Yamaguchi, Norihisa Tamura, Shigeyuki Nada, Shinobu Iwaki, and Satoko Konishi
- Subjects
Conformational change ,Chemistry ,Stereochemistry ,Antiporter ,Molecular Sequence Data ,Cell Biology ,Biochemistry ,Antiporters ,Major facilitator superfamily ,Transmembrane protein ,Transmembrane domain ,Bacterial Proteins ,Ethylmaleimide ,DNA Transposable Elements ,Mutagenesis, Site-Directed ,Tn10 ,Amino Acid Sequence ,Cysteine ,Binding site ,Molecular Biology - Abstract
Bacterial Tn10-encoded metal-tetracycline/H(+) antiporter was the first found drug exporter and has been studied as a paradigm of antiporter-type major facilitator superfamily transporters. Here the 400 amino acid residues of this protein were individually replaced by cysteine except for the initial methionine. As a result, we could obtain a complete map of the functionally or structurally important residues. In addition, we could determine the precise boundaries of all the transmembrane segments on the basis of the reactivity with N-ethylmaleimide (NEM). The NEM binding results indicated the presence of a transmembrane water-filled channel in the transporter. The twelve transmembrane segments can be divided into three groups; four are totally embedded in the hydrophobic interior, four face a putative water-filled channel along their full length, and the remaining four face the channel for half their length, the other halves being embedded in the hydrophobic interior. These three types of transmembrane segments are mutually arranged with a 4-fold symmetry. The competitive binding of membrane-permeable and -impermeable SH reagents in intact cells indicates that the transmembrane water-filled channel has a thin barrier against hydrophilic molecules in the middle of the transmembrane region. Inhibition and stimulation of NEM binding in the presence of tetracycline reflects the substrate-induced protection or conformational change of the Tn10-encoded metal-tetracycline/H(+) antiporter. The mutations protected from NEM binding by tetracycline were mainly located around the permeability barrier in the N-terminal half, suggesting the location of the substrate binding site.
- Published
- 2001
- Full Text
- View/download PDF
48. Monoclonal Antibody That Binds to the Central Loop of the Tn10-Encoded Metal Tetracycline/H+ Antiporter of Escherichia coli
- Author
-
Satoshi Murakami, Shigeyuki Nada, Akihito Yamaguchi, Yoshiyuki Kubo, and Shizuka Okamoto
- Subjects
Protein Conformation ,Antiporter ,Molecular Sequence Data ,Mutant ,medicine.disease_cause ,Biochemistry ,Antiporters ,Epitope ,Epitopes ,Bacterial Proteins ,Antibody Specificity ,Escherichia coli ,medicine ,Histidine ,Amino Acid Sequence ,Cysteine ,Binding site ,Molecular Biology ,Peptide sequence ,biology ,Chemistry ,Antibodies, Monoclonal ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,biology.protein ,Binding Sites, Antibody ,Antibody - Abstract
Mouse monoclonal antibodies were prepared using His-tagged Tn10-encoded metal-tetracycline/H+ antiporter [TetA(B)His] as an antigen. From them, those reacting equally with His-tagged and wild-type TetA(B) were selected and named TCL-1. Cysteine-scanning mutants were used to determine the TCL-1 binding site on the TetA(B) protein. First, 12 Cys mutants of TetA(B) in which one residue in a protruding loop region was replaced by cysteine were constructed. Western blot analysis revealed the binding of TCL-1 to all of these Cys-mutants except for R186C. Then, we constructed 13 cysteine-scanning mutants, F179C to T191C. Among them, eight mutants, F179C to T182C, N184C, and T189C to T191C, exhibited TCL-1 binding, whereas the other five, K183C, T185C, R186C, D187C, and N188C, exhibited no or lower TCL-1 binding. These results clearly indicate that the sequence recognized by TCL-1 is 183Lys-X-Thr-Arg-Asp-Asn188 in the central loop region of TetA(B). TCL-1 is the first reported antibody that binds to a region other than the C-terminus of TetA(B), and the recognized amino acid sequence was identified.
- Published
- 2001
- Full Text
- View/download PDF
49. The lysosomal signaling anchor p18/LAMTOR1 controls epidermal development by regulating lysosome-mediated catabolic processes
- Author
-
Taeko Soma-Nagae, Shigeyuki Nada, Mari Kitagawa, Chitose Oneyama, Yusuke Takahashi, Masato Okada, and Shunsuke Mori
- Subjects
Keratinocytes ,endocrine system ,endocrine system diseases ,MAP Kinase Signaling System ,Mice, Transgenic ,mTORC1 ,Biology ,Mice ,Microscopy, Electron, Transmission ,Lysosome ,Organelle ,Autophagy ,medicine ,Animals ,Homeostasis ,Adaptor Proteins, Signal Transducing ,Corneocyte ,Epidermis (botany) ,Signal transducing adaptor protein ,Cell Biology ,NM23 Nucleoside Diphosphate Kinases ,Cell biology ,medicine.anatomical_structure ,Animals, Newborn ,Epidermal Cells ,Cell culture ,Epidermis ,Lysosomes ,Signal Transduction - Abstract
Summary The lysosomal adaptor protein p18 is an essential anchor of a scaffolding complex for the mTORC1 and MAPK pathways, which play crucial roles in controlling cell growth and energy homeostasis. To elucidate the in vivo function of the p18-mediated pathway, we conditionally ablated p18 in the mouse epidermis. Mutant mice were born with severe defects in formation of the stratum corneum and died within 12 h after birth due to dehydration caused by loss of skin barrier function. Mutant epidermal cells can grow and differentiate into granular cells, but exhibit functional defects in corneocyte maturation. Electron microscopy identified abnormal immature cells, overlying the mutant granular cells, which accumulated autophagosomes, glycogen granules and dead nuclei. Cell culture analysis showed that loss of p18 attenuated lysosome function, resulting in accumulation of immature lysosomes and autophagosomes. Analyses of lysosome behavior revealed that p18 is required for functional interaction between lysosomes and target organelles including autophagosomes. These findings suggest that p18-mediated pathways control lysosome-mediated catabolic processes, which are crucial for the development of mouse epidermis.
- Published
- 2013
- Full Text
- View/download PDF
50. The Rho guanine nucleotide exchange factor ARHGEF5 promotes tumor malignancy via epithelial–mesenchymal transition
- Author
-
Kentaro Kajiwara, Miho Kuroiwa, Chitose Oneyama, Kubo Y, Hisataka Sabe, Masato Okada, Yasuhito Onodera, Suguru Nomimura, Jin-Min Nam, Yu Komiya, and Shigeyuki Nada
- Subjects
0301 basic medicine ,Cancer Research ,Cell growth ,Colorectal cancer ,Cell migration ,Biology ,medicine.disease ,medicine.disease_cause ,Metastasis ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Downregulation and upregulation ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Original Article ,Epithelial–mesenchymal transition ,Carcinogenesis ,Molecular Biology ,PI3K/AKT/mTOR pathway - Abstract
Epithelial tumor cells often acquire malignant properties, such as invasion/metastasis and uncontrolled cell growth, by undergoing epithelial–mesenchymal transition (EMT). However, the mechanisms by which EMT contributes to malignant progression remain elusive. Here we show that the Rho guanine nucleotide exchange factor (GEF) ARHGEF5 promotes tumor malignancy in a manner dependent on EMT status. We previously identified ARHGEF5, a member of the Dbl family of GEFs, as a multifunctional mediator of Src-induced cell invasion and tumor growth. In the present study, ARHGEF5 was upregulated during tumor growth factor-β-induced EMT in human epithelial MCF10A cells, and promoted cell migration by activating the Rho-ROCK pathway. ARHGEF5 was necessary for the invasive and in vivo metastatic activity of human colorectal cancer HCT116 cells. These findings underscore the crucial role of ARHGEF5 in cell migration and invasion/metastasis. An in vivo tumorigenesis assay revealed that ARHGEF5 had the potential to promote tumor growth via the phosphatidylinositol 3-kinase (PI3K) pathway. However, ARHGEF5 was not required for tumor growth in epithelial-like human colorectal cancer HCT116 and HT29 cells, whereas the growth of mesenchymal-like SW480 and SW620 cells depended on ARHGEF5. Induction of EMT by tumor necrosis factor-α or Slug in HCT116 cells resulted in the dependence of tumor growth on ARHGEF5. In these mesenchymal-like cells, Akt was activated via ARHGEF5 and its activity was required for tumor growth. Analysis of a transcriptome data set revealed that the combination of ARHGEF5 upregulation and E-cadherin downregulation or Snail upregulation was significantly correlated with poor prognosis in patients with colorectal cancers. Taken together, our findings suggest that EMT-induced ARHGEF5 activation contributes to the progression of tumor malignancy. ARHGEF5 may serve as a potential therapeutic target in a subset of malignant tumors that have undergone EMT.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.