1
|
Panagiotopoulos AA, Kalyvianaki K, Angelidaki A, Dellis D, Panagiotidis CA, Kampa M, Castanas E. The Sequence [RRKLPVGRS] Is a Nuclear Localization Signal for Importin 8 Binding (NLS8): A Chemical Biology and Bioinformatics Study. Int J Mol Sci 2025; 26:2814. [PMID: 40141456 PMCID: PMC11942892 DOI: 10.3390/ijms26062814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Karyopherins, carrier proteins that recognize particular cargo protein patterns known as nuclear localization signals (NLSs), mediate the nuclear translocation of big proteins. In order to better understand the process of nuclear transport of proteins and create the groundwork for the development of innovative treatments that specifically target importins, it is imperative to clarify the intricate interactions between nuclear transporters and their cargo proteins. Until recently, very few NLSs have been documented. In the current work, an in silico method was used to identify NLSs for importin 8. It was determined that the sequence RRKLPVGRS serves as a recognition motif for importin 8 binding a karyopherin that is involved in the nuclear transportation of several important proteins like AGOs, SMADs, RPL23A, and TFE3. The sequence was validated in vitro in the breast cancer cell line T47D. This work subscribes to the effort to clarify the intricate relationships between nuclear transporters and their cargo proteins, in order to better understand the mechanism of nuclear transport of proteins and lay the groundwork for the development of novel therapeutics that target particular importins and have an immediate translational impact.
Collapse
Affiliation(s)
- Athanasios A. Panagiotopoulos
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 71013 Heraklion, Greece; (K.K.); (A.A.); (M.K.); (E.C.)
| | - Konstantina Kalyvianaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 71013 Heraklion, Greece; (K.K.); (A.A.); (M.K.); (E.C.)
| | - Aikaterini Angelidaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 71013 Heraklion, Greece; (K.K.); (A.A.); (M.K.); (E.C.)
| | - Dimitris Dellis
- National Infrastructures for Research and Technology, 11523 Athens, Greece;
| | - Christos A. Panagiotidis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 71013 Heraklion, Greece; (K.K.); (A.A.); (M.K.); (E.C.)
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 71013 Heraklion, Greece; (K.K.); (A.A.); (M.K.); (E.C.)
| |
Collapse
|
2
|
Montgomery GW. Genetic regulation of ovulation rate and multiple births. Reprod Fertil Dev 2024; 36:RD24083. [PMID: 39222471 DOI: 10.1071/rd24083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Ovulation rate in many mammalian species is controlled to regulate the numbers of offspring and maximise reproductive success. Pathways that regulate ovulation rate still respond to genetic and environmental factors and show considerable variation within and between species. Genetic segregation, positional cloning, and association studies have discovered numerous mutations and genetic risk factors that contribute to this variation. Notable among the discoveries has been the role of mutations in bone morphogenetic protein 15 (BMP15 ), growth differentiation factor 9 (GDF9 ) and bone morphogenetic protein receptor type 1B (BMPR1B ) from the intra-ovarian signalling pathway contributing to the evidence that signalling from the oocyte is the key driver in follicle regulation rather than circulating gonadotrophin concentrations. Multiple variants in different domains of BMP15 and GDF9 result in partial or complete loss of function of the proteins providing insights into their functional roles and differential regulation contributing to species differences in ovulation rate. Early success encouraged many more studies in prolific strains of sheep, cattle and goats providing a valuable catalogue of genetic variants of large effect increasing ovulation rate and litter size. More recently, genetic association studies are beginning to identify genetic risk factors with smaller effects. Most genes implicated are from pathways with defined roles in regulation of the ovarian function. However, some genomic regions suggest regulation by novel genes. Continuing genetic and related functional studies will add further to our understanding of the detailed regulation of ovulation rate and litter size with implications for health and animal production systems.
Collapse
Affiliation(s)
- G W Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
3
|
Itoh Y, Miyamoto Y, Tokunaga M, Suzuki T, Takada A, Ninomiya A, Hishinuma T, Matsuda M, Yoneda Y, Oka M, Suzuki R, Matsuura Y, Okamoto T. Importin-7-dependent nuclear translocation of the Flavivirus core protein is required for infectious virus production. PLoS Pathog 2024; 20:e1012409. [PMID: 39146232 PMCID: PMC11326614 DOI: 10.1371/journal.ppat.1012409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024] Open
Abstract
Flaviviridae is a family of positive-stranded RNA viruses, including human pathogens, such as Japanese encephalitis virus (JEV), dengue virus (DENV), Zika virus (ZIKV), and West Nile virus (WNV). Nuclear localization of the viral core protein is conserved among Flaviviridae, and this feature may be targeted for developing broad-ranging anti-flavivirus drugs. However, the mechanism of core protein translocation to the nucleus and the importance of nuclear translocation in the viral life cycle remain unknown. We aimed to identify the molecular mechanism underlying core protein nuclear translocation. We identified importin-7 (IPO7), an importin-β family protein, as a nuclear carrier for Flaviviridae core proteins. Nuclear import assays revealed that core protein was transported into the nucleus via IPO7, whereas IPO7 deletion by CRISPR/Cas9 impaired their nuclear translocation. To understand the importance of core protein nuclear translocation, we evaluated the production of infectious virus or single-round-infectious-particles in wild-type or IPO7-deficient cells; both processes were significantly impaired in IPO7-deficient cells, whereas intracellular infectious virus levels were equivalent in wild-type and IPO7-deficient cells. These results suggest that IPO7-mediated nuclear translocation of core proteins is involved in the release of infectious virus particles of flaviviruses.
Collapse
Affiliation(s)
- Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Laboratory of Biofunctional Molecular Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Makoto Tokunaga
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akira Takada
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Akinori Ninomiya
- Central Instrumentation Laboratory, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tomomi Hishinuma
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Mami Matsuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiro Yoneda
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
4
|
Kitagawa T, Matsumoto T, Ohta T, Yoshida T, Saito Y, Nakayama Y, Hadate Y, Ashihara E, Watanabe T. Linderapyrone analogue LPD-01 as a cancer treatment agent by targeting importin7. J Nat Med 2024; 78:370-381. [PMID: 38265612 DOI: 10.1007/s11418-023-01774-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024]
Abstract
The Wnt/β-catenin signaling pathway plays important roles in several cancer cells, including cell proliferation and development. We previously succeeded in synthesizing a small molecule compound inhibiting the Wnt/β-catenin signaling pathway, named LPD-01 (1), and 1 inhibited the growth of human colorectal cancer (HT-29) cells. In this study, we revealed that 1 inhibits the growth of HT-29 cells stronger than that of another human colorectal cancer (SW480) cells. Therefore, we have attempted to identify the target proteins of 1 in HT-29 cells. Firstly, we investigated the effect on the expression levels of the Wnt/β-catenin signaling pathway-related proteins. As a result, 1 inhibited the expression of target proteins of Wnt/β-catenin signaling pathway (c-Myc and Survivin) and their genes, whereas the amount of transcriptional co-activator (β-catenin) was not decreased, suggesting that 1 inhibited the Wnt/β-catenin signaling pathway without affecting β-catenin. Next, we investigated the target proteins of 1 using magnetic FG beads. Chemical pull-down assay combined with mass spectrometry suggested that 1 directly binds to importin7. As expected, 1 inhibited the nuclear translocation of importin7 cargoes such as Smad2 and Smad3 in TGF-β-stimulated HT-29 cells. In addition, the knockdown of importin7 by siRNA reduced the expression of target genes of Wnt/β-catenin signaling pathway. These results suggest that importin7 is one of the target proteins of 1 for inhibition of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Takahiro Kitagawa
- Laboratory of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Takahiro Matsumoto
- Laboratory of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan.
| | - Tomoe Ohta
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Nagasaki, 859-3298, Japan
| | - Tatsusada Yoshida
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Nagasaki, 859-3298, Japan
| | - Youhei Saito
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Yuji Nakayama
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Yuki Hadate
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Eishi Ashihara
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan
| | - Tetsushi Watanabe
- Laboratory of Public Health, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, 607-8412, Japan.
| |
Collapse
|
5
|
Mbarek H, Gordon SD, Duffy DL, Hubers N, Mortlock S, Beck JJ, Hottenga JJ, Pool R, Dolan CV, Actkins KV, Gerring ZF, Van Dongen J, Ehli EA, Iacono WG, Mcgue M, Chasman DI, Gallagher CS, Schilit SLP, Morton CC, Paré G, Willemsen G, Whiteman DC, Olsen CM, Derom C, Vlietinck R, Gudbjartsson D, Cannon-Albright L, Krapohl E, Plomin R, Magnusson PKE, Pedersen NL, Hysi P, Mangino M, Spector TD, Palviainen T, Milaneschi Y, Penninnx BW, Campos AI, Ong KK, Perry JRB, Lambalk CB, Kaprio J, Ólafsson Í, Duroure K, Revenu C, Rentería ME, Yengo L, Davis L, Derks EM, Medland SE, Stefansson H, Stefansson K, Del Bene F, Reversade B, Montgomery GW, Boomsma DI, Martin NG. Genome-wide association study meta-analysis of dizygotic twinning illuminates genetic regulation of female fecundity. Hum Reprod 2024; 39:240-257. [PMID: 38052102 PMCID: PMC10767824 DOI: 10.1093/humrep/dead247] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2023] [Indexed: 12/07/2023] Open
Abstract
STUDY QUESTION Which genetic factors regulate female propensity for giving birth to spontaneous dizygotic (DZ) twins? SUMMARY ANSWER We identified four new loci, GNRH1, FSHR, ZFPM1, and IPO8, in addition to previously identified loci, FSHB and SMAD3. WHAT IS KNOWN ALREADY The propensity to give birth to DZ twins runs in families. Earlier, we reported that FSHB and SMAD3 as associated with DZ twinning and female fertility measures. STUDY DESIGN, SIZE, DURATION We conducted a genome-wide association meta-analysis (GWAMA) of mothers of spontaneous dizygotic (DZ) twins (8265 cases, 264 567 controls) and of independent DZ twin offspring (26 252 cases, 417 433 controls). PARTICIPANTS/MATERIALS, SETTING, METHODS Over 700 000 mothers of DZ twins, twin individuals and singletons from large cohorts in Australia/New Zealand, Europe, and the USA were carefully screened to exclude twins born after use of ARTs. Genetic association analyses by cohort were followed by meta-analysis, phenome wide association studies (PheWAS), in silico and in vivo annotations, and Zebrafish functional validation. MAIN RESULTS AND THE ROLE OF CHANCE This study enlarges the sample size considerably from previous efforts, finding four genome-wide significant loci, including two novel signals and a further two novel genes that are implicated by gene level enrichment analyses. The novel loci, GNRH1 and FSHR, have well-established roles in female reproduction whereas ZFPM1 and IPO8 have not previously been implicated in female fertility. We found significant genetic correlations with multiple aspects of female reproduction and body size as well as evidence for significant selection against DZ twinning during human evolution. The 26 top single nucleotide polymorphisms (SNPs) from our GWAMA in European-origin participants weakly predicted the crude twinning rates in 47 non-European populations (r = 0.23 between risk score and population prevalence, s.e. 0.11, 1-tail P = 0.058) indicating that genome-wide association studies (GWAS) are needed in African and Asian populations to explore the causes of their respectively high and low DZ twinning rates. In vivo functional tests in zebrafish for IPO8 validated its essential role in female, but not male, fertility. In most regions, risk SNPs linked to known expression quantitative trait loci (eQTLs). Top SNPs were associated with in vivo reproductive hormone levels with the top pathways including hormone ligand binding receptors and the ovulation cycle. LARGE SCALE DATA The full DZT GWAS summary statistics will made available after publication through the GWAS catalog (https://www.ebi.ac.uk/gwas/). LIMITATIONS, REASONS FOR CAUTION Our study only included European ancestry cohorts. Inclusion of data from Africa (with the highest twining rate) and Asia (with the lowest rate) would illuminate further the biology of twinning and female fertility. WIDER IMPLICATIONS OF THE FINDINGS About one in 40 babies born in the world is a twin and there is much speculation on why twinning runs in families. We hope our results will inform investigations of ovarian response in new and existing ARTs and the causes of female infertility. STUDY FUNDING/COMPETING INTEREST(S) Support for the Netherlands Twin Register came from the Netherlands Organization for Scientific Research (NWO) and The Netherlands Organization for Health Research and Development (ZonMW) grants, 904-61-193, 480-04-004, 400-05-717, Addiction-31160008, 911-09-032, Biobanking and Biomolecular Resources Research Infrastructure (BBMRI.NL, 184.021.007), Royal Netherlands Academy of Science Professor Award (PAH/6635) to DIB, European Research Council (ERC-230374), Rutgers University Cell and DNA Repository (NIMH U24 MH068457-06), the Avera Institute, Sioux Falls, South Dakota (USA) and the National Institutes of Health (NIH R01 HD042157-01A1) and the Genetic Association Information Network (GAIN) of the Foundation for the National Institutes of Health and Grand Opportunity grants 1RC2 MH089951. The QIMR Berghofer Medical Research Institute (QIMR) study was supported by grants from the National Health and Medical Research Council (NHMRC) of Australia (241944, 339462, 389927, 389875, 389891, 389892, 389938, 443036, 442915, 442981, 496610, 496739, 552485, 552498, 1050208, 1075175). L.Y. is funded by Australian Research Council (Grant number DE200100425). The Minnesota Center for Twin and Family Research (MCTFR) was supported in part by USPHS Grants from the National Institute on Alcohol Abuse and Alcoholism (AA09367 and AA11886) and the National Institute on Drug Abuse (DA05147, DA13240, and DA024417). The Women's Genome Health Study (WGHS) was funded by the National Heart, Lung, and Blood Institute (HL043851 and HL080467) and the National Cancer Institute (CA047988 and UM1CA182913), with support for genotyping provided by Amgen. Data collection in the Finnish Twin Registry has been supported by the Wellcome Trust Sanger Institute, the Broad Institute, ENGAGE-European Network for Genetic and Genomic Epidemiology, FP7-HEALTH-F4-2007, grant agreement number 201413, National Institute of Alcohol Abuse and Alcoholism (grants AA-12502, AA-00145, AA-09203, AA15416, and K02AA018755) and the Academy of Finland (grants 100499, 205585, 118555, 141054, 264146, 308248, 312073 and 336823 to J. Kaprio). TwinsUK is funded by the Wellcome Trust, Medical Research Council, Versus Arthritis, European Union Horizon 2020, Chronic Disease Research Foundation (CDRF), Zoe Ltd and the National Institute for Health Research (NIHR) Clinical Research Network (CRN) and Biomedical Research Centre based at Guy's and St Thomas' NHS Foundation Trust in partnership with King's College London. For NESDA, funding was obtained from the Netherlands Organization for Scientific Research (Geestkracht program grant 10000-1002), the Center for Medical Systems Biology (CSMB, NVVO Genomics), Biobanking and Biomolecular Resources Research Infrastructure (BBMRI-NL), VU University's Institutes for Health and Care Research (EMGO+) and Neuroscience Campus Amsterdam, University Medical Center Groningen, Leiden University Medical Center, National Institutes of Health (NIH, ROI D0042157-01A, MH081802, Grand Opportunity grants 1 RC2 Ml-1089951 and IRC2 MH089995). Part of the genotyping and analyses were funded by the Genetic Association Information Network (GAIN) of the Foundation for the National Institutes of Health. Computing was supported by BiG Grid, the Dutch e-Science Grid, which is financially supported by NWO. Work in the Del Bene lab was supported by the Programme Investissements d'Avenir IHU FOReSIGHT (ANR-18-IAHU-01). C.R. was supported by an EU Horizon 2020 Marie Skłodowska-Curie Action fellowship (H2020-MSCA-IF-2014 #661527). H.S. and K.S. are employees of deCODE Genetics/Amgen. The other authors declare no competing financial interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Hamdi Mbarek
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Qatar Genome Program, Qatar Foundation, Doha, Qatar
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - David L Duffy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nikki Hubers
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | - Sally Mortlock
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Jeffrey J Beck
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | - René Pool
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | - Conor V Dolan
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ky’Era V Actkins
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | | | - Jenny Van Dongen
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | - Erik A Ehli
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - William G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Matt Mcgue
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Daniel I Chasman
- Harvard Medical School, Harvard University, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Samantha L P Schilit
- Harvard Medical School, Harvard University, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | - Cynthia C Morton
- Harvard Medical School, Harvard University, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | - Guillaume Paré
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Gonneke Willemsen
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | | | | | | | | | | | | | - Eva Krapohl
- Medical Research Council Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Statistical Sciences & Innovation, UCB Biosciences GmbH, Monheim, Germany
| | - Robert Plomin
- Medical Research Council Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Pirro Hysi
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
- NIHR Biomedical Research Centre at Guy’s and St Thomas’ Foundation Trust, London, UK
| | - Timothy D Spector
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Yuri Milaneschi
- Department of Psychiatry, EMGO Institute for Health and Care Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Brenda W Penninnx
- Department of Psychiatry, EMGO Institute for Health and Care Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Adrian I Campos
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Cornelis B Lambalk
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
- Amsterdam University Medical Centers Location VU Medical Center, Amsterdam, The Netherlands
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Ísleifur Ólafsson
- Department of Clinical Biochemistry, National University Hospital of Iceland, Reykjavik, Iceland
| | - Karine Duroure
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Céline Revenu
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Loic Yengo
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Lea Davis
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Eske M Derks
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | | | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Bruno Reversade
- Genome Institute of Singapore, Laboratory of Human Genetics & Therapeutics, A*STAR, Singapore, Singapore
- Smart-Health Initiative, BESE, KAUST, Thuwal, Saudi Arabia
| | - Grant W Montgomery
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
6
|
Ding N, Li H, Zhang Z, Jia H. Inhibition of importin-7 attenuates ventilator-induced lung injury by targeting nuclear translocation of p38. Inflamm Res 2023; 72:971-988. [PMID: 37004548 DOI: 10.1007/s00011-023-01727-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/13/2023] [Accepted: 03/26/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND The ability of p38 to phosphorylate substrates in the nucleus and the role of nuclear p38 in the regulation of inflammation have focused attention on the subcellular localization of the kinase. Although it is clear that p38 shuttles to the nucleus upon stimulation, the mechanisms that regulate p38 nuclear input in response to mechanical stretch remain to be determined. METHODS Cyclic stretch (CS)-induced nuclear translocation of p38 was determined by Western blotting and immunofluorescence. The p38 interacting protein was identified using endogenous pull-down and protein binding assays. The potential role of importin-7 (Imp7) in CS-induced nuclear translocation of p38 and p38-dependent gene expression was confirmed using a series of in vitro and in vivo experiments. Furthermore, we tested the therapeutic potential of intratracheal administration of Imp7 siRNA-loaded nanoparticles in the ventilator-induced lung injury (VILI) mouse model. RESULTS We show that CS induced phosphorylation-dependent nuclear translocation of p38, which required the involvement of microtubules and dynein. Endogenous pull-down assay revealed Imp7 to be a potential p38-interacting protein, and the direct interaction between p38 and Imp7 was confirmed by in vitro and in vivo binding assays. Furthermore, silencing Imp7 inhibited CS-induced nuclear translocation of p38 and subsequent cytokine production. Notably, intratracheal administration of Imp7 siRNA nanoparticles attenuated lung inflammation and histological damage in the VILI mouse model. CONCLUSIONS Our findings uncover a key role for Imp7 in the process of p38 nuclear import after CS stimulation and highlight the potential of preventing p38 nuclear translocation in treatment of VILI.
Collapse
Affiliation(s)
- Ning Ding
- Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250031, China.
- Key Laboratory of Critical Rehabilitation Medicine of Shandong Province, Shandong Provincial Third Hospital, Jinan, 250031, China.
| | - Huiqing Li
- Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250031, China
- Key Laboratory of Critical Rehabilitation Medicine of Shandong Province, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Zengzhen Zhang
- Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250031, China
- Key Laboratory of Critical Rehabilitation Medicine of Shandong Province, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Haiyan Jia
- Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250031, China
- Key Laboratory of Critical Rehabilitation Medicine of Shandong Province, Shandong Provincial Third Hospital, Jinan, 250031, China
| |
Collapse
|
7
|
Rodari MM, Cerf-Bensussan N, Parlato M. Dysregulation of the immune response in TGF-β signalopathies. Front Immunol 2022; 13:1066375. [PMID: 36569843 PMCID: PMC9780292 DOI: 10.3389/fimmu.2022.1066375] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family of cytokines exerts pleiotropic functions during embryonic development, tissue homeostasis and repair as well as within the immune system. Single gene defects in individual component of this signaling machinery cause defined Mendelian diseases associated with aberrant activation of TGF-β signaling, ultimately leading to impaired development, immune responses or both. Gene defects that affect members of the TGF-β cytokine family result in more restricted phenotypes, while those affecting downstream components of the signaling machinery induce broader defects. These rare disorders, also known as TGF-β signalopathies, provide the unique opportunity to improve our understanding of the role and the relevance of the TGF-β signaling in the human immune system. Here, we summarize this elaborate signaling pathway, review the diverse clinical presentations and immunological phenotypes observed in these patients and discuss the phenotypic overlap between humans and mice genetically deficient for individual components of the TGF-β signaling cascade.
Collapse
|
8
|
García-García M, Sánchez-Perales S, Jarabo P, Calvo E, Huyton T, Fu L, Ng SC, Sotodosos-Alonso L, Vázquez J, Casas-Tintó S, Görlich D, Echarri A, Del Pozo MA. Mechanical control of nuclear import by Importin-7 is regulated by its dominant cargo YAP. Nat Commun 2022; 13:1174. [PMID: 35246520 PMCID: PMC8897400 DOI: 10.1038/s41467-022-28693-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 01/19/2022] [Indexed: 12/31/2022] Open
Abstract
Mechanical forces regulate multiple essential pathways in the cell. The nuclear translocation of mechanoresponsive transcriptional regulators is an essential step for mechanotransduction. However, how mechanical forces regulate the nuclear import process is not understood. Here, we identify a highly mechanoresponsive nuclear transport receptor (NTR), Importin-7 (Imp7), that drives the nuclear import of YAP, a key regulator of mechanotransduction pathways. Unexpectedly, YAP governs the mechanoresponse of Imp7 by forming a YAP/Imp7 complex that responds to mechanical cues through the Hippo kinases MST1/2. Furthermore, YAP behaves as a dominant cargo of Imp7, restricting the Imp7 binding and the nuclear translocation of other Imp7 cargoes such as Smad3 and Erk2. Thus, the nuclear import process is an additional regulatory layer indirectly regulated by mechanical cues, which activate a preferential Imp7 cargo, YAP, which competes out other cargoes, resulting in signaling crosstalk. The translation of mechanical cues into gene expression changes is dependent on the nuclear import of mechanoresponsive transcriptional regulators. Here the authors identify that Importin-7 drives the nuclear import of one such regulator YAP while YAP then controls Importin-7 response to mechanical cues and restricts Importin-7 binding to other cargoes.
Collapse
Affiliation(s)
- María García-García
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Sara Sánchez-Perales
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Patricia Jarabo
- Instituto Cajal-CSIC, Avda. Doctor Arce, 37, 28002, Madrid, Spain
| | - Enrique Calvo
- Proteomics Unit. Area of Vascular Physiopathology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Trevor Huyton
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Liran Fu
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Sheung Chun Ng
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Jesús Vázquez
- Proteomics Unit. Area of Vascular Physiopathology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
9
|
She Y, Zhang Y, Xiao Z, Yuan G, Yang G. The regulation of Msx1 by BMP4/pSmad1/5 signaling is mediated by importin7 in dental mesenchymal cells. Cells Dev 2022; 169:203763. [PMID: 34995814 DOI: 10.1016/j.cdev.2021.203763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/08/2023]
Abstract
Msx1 is essential for the maintenance of the odontogenic fate of dental mesenchymal cells, and is regulated by BMP/Smad1/5 signaling in a Smad4-independent manner. However, the exact co-factors that assist pSmad1/5 entering the nucleus to regulate Msx1 in dental mesenchymal cells are still unknown. Importin7 (IPO7) is one of the important members of importin β-superfamily, which is mainly responsible for nucleocytoplasmic shuttling of RNAs and proteins, including transcription factors. This study aims to investigate whether IPO7 participates in the nuclear translocation of pSmad1/5 activated by BMP4 to regulate Msx1 expression in mouse dental mesenchymal cells. In the current study, we found that IPO7 was strongly expressed in the mouse dental mesenchymal cells at postnatal day 1 (PN1) both in vitro and in vivo. With BMP4 stimulation, IPO7 showed a translocation from the cytoplasm to the nucleus. Knockdown of IPO7 with siRNA inhibited the nuclear accumulation of pSmad1/5 in response to BMP4 stimulation. Furthermore, the co-immunoprecipitation assay showed pSmad1/5 was a nuclear import cargo of IPO7. Next, knockdown of IPO7 abolished the upregulation of Msx1 induced by BMP4, while overexpression of Smad1 was able to rescue the Msx1 expression. Finally, ChIP and Re-ChIP assay showed IPO7 facilitated the recruitment of pSmad1/5 to the Msx1 promoter. Taken together, our data demonstrated that the regulation of Msx1 by BMP4/pSmad1/5 signaling is mediated by importin7 in mouse dental mesenchymal cells.
Collapse
Affiliation(s)
- Yawei She
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ziqiu Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guobin Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Yang Y, Ye WL, Zhang RN, He XS, Wang JR, Liu YX, Wang Y, Yang XM, Zhang YJ, Gan WJ. The Role of TGF- β Signaling Pathways in Cancer and Its Potential as a Therapeutic Target. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6675208. [PMID: 34335834 PMCID: PMC8321733 DOI: 10.1155/2021/6675208] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway mediates various biological functions, and its dysregulation is closely related to the occurrence of malignant tumors. However, the role of TGF-β signaling in tumorigenesis and development is complex and contradictory. On the one hand, TGF-β signaling can exert antitumor effects by inhibiting proliferation or inducing apoptosis of cancer cells. On the other hand, TGF-β signaling may mediate oncogene effects by promoting metastasis, angiogenesis, and immune escape. This review summarizes the recent findings on molecular mechanisms of TGF-β signaling. Specifically, this review evaluates TGF-β's therapeutic potential as a target by the following perspectives: ligands, receptors, and downstream signaling. We hope this review can trigger new ideas to improve the current clinical strategies to treat tumors related to the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Yun Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Ruo-Nan Zhang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Xiao-Shun He
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Jing-Ru Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yu-Xuan Liu
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Yi Wang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xue-Mei Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yu-Juan Zhang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Soochow University, Suzhou 215124, China
| |
Collapse
|
11
|
Ziegler A, Duclaux-Loras R, Revenu C, Charbit-Henrion F, Begue B, Duroure K, Grimaud L, Guihot AL, Desquiret-Dumas V, Zarhrate M, Cagnard N, Mas E, Breton A, Edouard T, Billon C, Frank M, Colin E, Lenaers G, Henrion D, Lyonnet S, Faivre L, Alembik Y, Philippe A, Moulin B, Reinstein E, Tzur S, Attali R, McGillivray G, White SM, Gallacher L, Kutsche K, Schneeberger P, Girisha KM, Nayak SS, Pais L, Maroofian R, Rad A, Vona B, Karimiani EG, Lekszas C, Haaf T, Martin L, Ruemmele F, Bonneau D, Cerf-Bensussan N, Del Bene F, Parlato M. Bi-allelic variants in IPO8 cause a connective tissue disorder associated with cardiovascular defects, skeletal abnormalities, and immune dysregulation. Am J Hum Genet 2021; 108:1126-1137. [PMID: 34010604 PMCID: PMC8206386 DOI: 10.1016/j.ajhg.2021.04.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulated transforming growth factor TGF-β signaling underlies the pathogenesis of genetic disorders affecting the connective tissue such as Loeys-Dietz syndrome. Here, we report 12 individuals with bi-allelic loss-of-function variants in IPO8 who presented with a syndromic association characterized by cardio-vascular anomalies, joint hyperlaxity, and various degree of dysmorphic features and developmental delay as well as immune dysregulation; the individuals were from nine unrelated families. Importin 8 belongs to the karyopherin family of nuclear transport receptors and was previously shown to mediate TGF-β-dependent SMADs trafficking to the nucleus in vitro. The important in vivo role of IPO8 in pSMAD nuclear translocation was demonstrated by CRISPR/Cas9-mediated inactivation in zebrafish. Consistent with IPO8's role in BMP/TGF-β signaling, ipo8-/- zebrafish presented mild to severe dorso-ventral patterning defects during early embryonic development. Moreover, ipo8-/- zebrafish displayed severe cardiovascular and skeletal defects that mirrored the human phenotype. Our work thus provides evidence that IPO8 plays a critical and non-redundant role in TGF-β signaling during development and reinforces the existing link between TGF-β signaling and connective tissue defects.
Collapse
Affiliation(s)
- Alban Ziegler
- Department of Biochemistry and Molecular Biology, CHU d'Angers, 49000 Angers, France; University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Rémi Duclaux-Loras
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, 75015 Paris, France
| | - Céline Revenu
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, 75012 Paris, France; Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, 75005 Paris, France
| | - Fabienne Charbit-Henrion
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, 75015 Paris, France; Department of Pediatric Gastroenterology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, 75015 Paris, France; Department of Molecular Genetics, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Bernadette Begue
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, 75015 Paris, France
| | - Karine Duroure
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, 75012 Paris, France; Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, 75005 Paris, France
| | - Linda Grimaud
- University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Anne Laure Guihot
- University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Valérie Desquiret-Dumas
- Department of Biochemistry and Molecular Biology, CHU d'Angers, 49000 Angers, France; University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Mohammed Zarhrate
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cité University, 75015 Paris, France
| | - Nicolas Cagnard
- Bioinformatics Core Facility, INSERM-UMR 1163, Imagine Institute, 75015 Paris, France
| | - Emmanuel Mas
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse 31300, France; Centre de Référence des Maladies Rares Digestives, and Pediatric Clinical Research Unit, Toulouse Clinical Investigation Center INSERM U1436, Hôpital des Enfants, CHU de Toulouse, Toulouse 31300, France
| | - Anne Breton
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse 31300, France; Centre de Référence des Maladies Rares Digestives, and Pediatric Clinical Research Unit, Toulouse Clinical Investigation Center INSERM U1436, Hôpital des Enfants, CHU de Toulouse, Toulouse 31300, France
| | - Thomas Edouard
- Reference Centre for Marfan Syndrome and Reference Centre on Rare Bone Diseases, Pediatric Clinical Research Unit, Children's Hospital, Toulouse University Hospital, RESTORE, INSERM UMR1301, 31300 Toulouse, France
| | - Clarisse Billon
- Centre de Génétique, Centre de Référence des Maladies Vasculaires Rares, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Michael Frank
- Centre de Génétique, Centre de Référence des Maladies Vasculaires Rares, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Estelle Colin
- Department of Biochemistry and Molecular Biology, CHU d'Angers, 49000 Angers, France
| | - Guy Lenaers
- University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Daniel Henrion
- University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Stanislas Lyonnet
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France; Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Laurence Faivre
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, 21000 Dijon, France
| | - Yves Alembik
- Département de Génétique Médicale, CHU de Hautepierre, 67200 Strasbourg, France
| | - Anaïs Philippe
- Département de Génétique Médicale, CHU de Hautepierre, 67200 Strasbourg, France
| | - Bruno Moulin
- Nephrology and Transplantation Department, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Eyal Reinstein
- Medical Genetics Institute, Meir Medical Center, Kfar-Saba 4428164, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shay Tzur
- Genomic Research Department, Emedgene Technologies, 67443 Tel Aviv, Israel
| | - Ruben Attali
- Genomic Research Department, Emedgene Technologies, 67443 Tel Aviv, Israel
| | - George McGillivray
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville 3052, Melbourne, VIC, Australia
| | - Susan M White
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville 3052, Melbourne, VIC, Australia
| | - Lyndon Gallacher
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville 3052, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, 3010 Parkville, Melbourne, VIC, Australia
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Pauline Schneeberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Katta M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shalini S Nayak
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Lynn Pais
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Reza Maroofian
- Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Aboulfazl Rad
- Department of Otolaryngology-Head & Neck Surgery, Tübingen Hearing Research Centre, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Barbara Vona
- Department of Otolaryngology-Head & Neck Surgery, Tübingen Hearing Research Centre, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany; Institute of Human Genetics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Ehsan Ghayoor Karimiani
- Molecular and Clinical Sciences Institute, St. George's, University of London, Cranmer Terrace London, SW17 ORE London, UK; Innovative Medical Research Center, Mashhad Branch, Islamic Azdad University, Mashhad 9133736351, Iran
| | - Caroline Lekszas
- Institute of Human Genetics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Ludovic Martin
- University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France; Department of Dermatology, CHU d'Angers, 49000 Angers, France
| | - Frank Ruemmele
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, 75015 Paris, France; Department of Pediatric Gastroenterology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Dominique Bonneau
- Department of Biochemistry and Molecular Biology, CHU d'Angers, 49000 Angers, France; University of Angers, MitoVasc, UMR CNRS 6015, INSERM 1083, 49933 Angers, France
| | - Nadine Cerf-Bensussan
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, 75015 Paris, France
| | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, 75012 Paris, France; Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, 75005 Paris, France.
| | - Marianna Parlato
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, 75015 Paris, France.
| |
Collapse
|
12
|
Park JH, Kim J, Choi SY, Lee B, Lee JE, Park H, Moon JW, Park SH, Lee JM, Lee HS, Oh J. Albumin inhibits the nuclear translocation of Smad3 via interleukin-1beta signaling in hepatic stellate cells. Sci Rep 2021; 11:3196. [PMID: 33542418 PMCID: PMC7862402 DOI: 10.1038/s41598-021-82758-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Activation of quiescent hepatic stellate cells (HSCs) to myofibroblasts plays a key role in liver fibrosis. We had previously shown that albumin and its derivative, R-III (a retinol-binding protein—albumin domain III fusion protein), inhibited HSC activation by sequestering retinoic acid (RA) and that R-III administration reduced carbon tetrachloride (CCl4)-induced liver fibrosis. In this study, we aimed to elucidate the mechanism of action of albumin downstream of RA sequestration. Nuclear factor-κB p65 was evenly distributed in the cytoplasm in activated mouse HSCs, whereas albumin expression or R-III treatment (albumin/R-III) caused the nuclear translocation of p65, probably via RA sequestration, resulting in a dramatic increase in interleukin-1beta (IL-1β) expression. Albumin/R-III in turn induced the phosphorylation of Smad3 at the linker region, inhibiting its nuclear import in an IL-1β-dependent manner. Consistent with the in vitro results, the level of IL-1β mRNA expression was higher in CCl4/R-III-treated livers than in CCl4-treated livers. These findings reveal that albumin/R-III inhibits the transforming growth factor-β-Smad3 signaling as well as the retinoic acid receptor-mediated pathway, which probably contributes to the inhibition of HSC activation, and suggest that R-III may be an anti-fibrotic drug candidate.
Collapse
Affiliation(s)
- Ji Hoon Park
- Protein Drug Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong, 28160, Korea
| | - Janghyun Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - So-Young Choi
- Protein Drug Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong, 28160, Korea
| | - Boram Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Jung-Eun Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Heekyung Park
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Ji Wook Moon
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Sun-Hwa Park
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Jae Min Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Korea
| | - Hong Sik Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Korea
| | - Junseo Oh
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea. .,Department of Biomedical Science, Korea University Graduate School, Seoul, 02841, Korea.
| |
Collapse
|
13
|
Panagiotopoulos AA, Polioudaki C, Ntallis SG, Dellis D, Notas G, Panagiotidis CA, Theodoropoulos PA, Castanas E, Kampa M. The sequence [EKRKI(E/R)(K/L/R/S/T)] is a nuclear localization signal for importin 7 binding (NLS7). Biochim Biophys Acta Gen Subj 2021; 1865:129851. [PMID: 33482249 DOI: 10.1016/j.bbagen.2021.129851] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Nuclear translocation of large proteins is mediated through specific protein carriers, collectively named karyopherins (importins, exportins and adaptor proteins). Cargo proteins are recognized by importins through specific motifs, known as nuclear localization signals (NLS). However, only the NLS recognized by importin α and transportin (M9 NLS) have been identified so far METHODS: An unsupervised in silico approach was used, followed by experimental validation. RESULTS We identified the sequence EKRKI(E/R)(K/L/R/S/T) as an NLS signal for importin 7 recognition. This sequence was validated in the breast cancer cell line T47D, which expresses importin 7. Finally, we verified that importin 7-mediated nuclear protein transport is affected by cargo protein phosphorylation. CONCLUSIONS The NLS sequence for importin 7 was identified and we propose this approach as an identification method of novel specific NLS sequences for β-karyopherin family members. GENERAL SIGNIFICANCE Elucidating the complex relationships of the nuclear transporters and their cargo proteins may help in laying the foundation for the development of novel therapeutics, targeting specific importins, with an immediate translational impact.
Collapse
Affiliation(s)
| | - Chara Polioudaki
- Laboratory of Biochemistry, School of Medicine, University of Crete, 71013, Greece
| | - Sotirios G Ntallis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, 71013, Greece
| | - Christos A Panagiotidis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, 71013, Greece.
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, 71013, Greece.
| |
Collapse
|
14
|
Kawasaki N, Miwa T, Hokari S, Sakurai T, Ohmori K, Miyauchi K, Miyazono K, Koinuma D. Long noncoding RNA NORAD regulates transforming growth factor-β signaling and epithelial-to-mesenchymal transition-like phenotype. Cancer Sci 2018; 109:2211-2220. [PMID: 29722104 PMCID: PMC6029837 DOI: 10.1111/cas.13626] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/16/2018] [Accepted: 04/21/2018] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNAs are involved in a variety of cellular functions. In particular, an increasing number of studies have revealed the functions of long noncoding RNA in various cancers; however, their precise roles and mechanisms of action remain to be elucidated. NORAD, a cytoplasmic long noncoding RNA, is upregulated by irradiation and functions as a potential oncogenic factor by binding and inhibiting Pumilio proteins (PUM1/PUM2). Here, we show that NORAD upregulates transforming growth factor-β (TGF-β) signaling and regulates TGF-β-induced epithelial-to-mesenchymal transition (EMT)-like phenotype, which is a critical step in the progression of lung adenocarcinoma, A549 cells. However, PUM1 does not appear to be involved in this process. We thus focused on importin β1 as a binding partner of NORAD and found that knockdown of NORAD partially inhibits the physical interaction of importin β1 with Smad3, inhibiting the nuclear accumulation of Smad complexes in response to TGF-β. Our findings may provide a new mechanism underlying the function of NORAD in cancer cells.
Collapse
Affiliation(s)
- Natsumi Kawasaki
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiki Miwa
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Hokari
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Respiratory Medicine and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tsubasa Sakurai
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuho Ohmori
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kensuke Miyauchi
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Cutler AA, Jackson JB, Corbett AH, Pavlath GK. Non-equivalence of nuclear import among nuclei in multinucleated skeletal muscle cells. J Cell Sci 2018; 131:jcs.207670. [PMID: 29361530 DOI: 10.1242/jcs.207670] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/21/2017] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle is primarily composed of large myofibers containing thousands of post-mitotic nuclei distributed throughout a common cytoplasm. Protein production and localization in specialized myofiber regions is crucial for muscle function. Myonuclei differ in transcriptional activity and protein accumulation, but how these differences among nuclei sharing a cytoplasm are achieved is unknown. Regulated nuclear import of proteins is one potential mechanism for regulating transcription spatially and temporally in individual myonuclei. The best-characterized nuclear localization signal (NLS) in proteins is the classical NLS (cNLS), but many other NLS motifs exist. We examined cNLS and non-cNLS reporter protein import using multinucleated muscle cells generated in vitro, revealing that cNLS and non-cNLS nuclear import differs among nuclei in the same cell. Investigation of cNLS nuclear import rates in isolated myofibers ex vivo confirmed differences in nuclear import rates among myonuclei. Analyzing nuclear import throughout myogenesis revealed that cNLS and non-cNLS import varies during differentiation. Taken together, our results suggest that both spatial and temporal regulation of nuclear import pathways are important in muscle cell differentiation and protein regionalization in myofibers.
Collapse
Affiliation(s)
- Alicia A Cutler
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA.,Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | | | - Anita H Corbett
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
16
|
Hu X, Kan H, Boye A, Jiang Y, Wu C, Yang Y. Mitogen-activated protein kinase inhibitors reduce the nuclear accumulation of phosphorylated Smads by inhibiting Imp 7 or Imp 8 in HepG2 cells. Oncol Lett 2018; 15:4867-4872. [PMID: 29552126 PMCID: PMC5840705 DOI: 10.3892/ol.2018.7926] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/30/2018] [Indexed: 11/19/2022] Open
Abstract
The transforming growth factor (TGF)-β/Smad signaling pathway is involved in hepatocellular carcinoma development. Smad2 and Smad3 are phosphorylated following TGF-β1 stimulation and subsequently oligomerize with Smad4 to form the Smad2/3/4 complex, which translocates into the nucleus and regulates target genes, including plasminogen activator inhibitor type 1 (PAI1). Importin (Imp)7 and Imp8 are responsible for transporting phosphorylated (p)Smad2/3 and Smad4 into the nucleus. In our previous study, it was demonstrated that mitogen-activated protein kinase (MAPK) inhibitors, including inhibitors of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 could inhibit the transcription of PAI1, but ERK inhibitor had no significant effect on the phosphorylation of Smad2/3, and the formation of Smad2/3/4 complexes, which was different from the effect of JNK or p38 inhibitor. We hypothesized that MAPK inhibitors, particularly ERK inhibitor, reduced the transport of Smads into the nucleus by affecting Imp7 and Imp8. To confirm this hypothesis, HepG2 cells were incubated with different MAPK inhibitors for 5 h and subsequently stimulated with TGF-β1 for 1 h. Next, the intracellular locations of Smads (pSmad2C, pSmad2L, pSmad3C, pSmad3L and Smad4) and Imp7/8 were detected using immunofluorescence staining assays, and the expression of Imp7/8 was investigated using immunoblotting. It was revealed that JNK or p38 inhibitor decreased the phosphorylation of Smad2C, Smad2L and Smad3L, and affected their nuclear accumulation. Although only inhibiting the phosphorylation of Smad2C, ERK inhibitor affected the nuclear accumulation of pSmad2C, pSmad2L, pSmad3C and pSmad3L. The three MAPK inhibitors attenuated the nuclear distribution of Smad4, and the expression and nuclear accumulation of Imp7. ERK and JNK inhibitors attenuated the expression and nuclear accumulation of Imp8. Thus, the results of the present study suggest that MAPK inhibitors, particularly ERK inhibitor, modulate the nuclear accumulation of Smads via the inhibition of Imp 7/8.
Collapse
Affiliation(s)
- Xiangpeng Hu
- Department of Pharmacology, Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China.,Digestive Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Hongwei Kan
- Department of Pharmacology, Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Alex Boye
- Department of Pharmacology, Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yufeng Jiang
- Department of Pharmacology, Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Chao Wu
- Department of Pharmacology, Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan Yang
- Department of Pharmacology, Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
17
|
Xiao X, Senavirathna LK, Gou X, Huang C, Liang Y, Liu L. EZH2 enhances the differentiation of fibroblasts into myofibroblasts in idiopathic pulmonary fibrosis. Physiol Rep 2017; 4:4/17/e12915. [PMID: 27582065 PMCID: PMC5027349 DOI: 10.14814/phy2.12915] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022] Open
Abstract
The accumulation of fibroblasts/myofibroblasts in fibrotic foci is one of the characteristics of idiopathic pulmonary fibrosis (IPF). Enhancer of zeste homolog 2 (EZH2) is the catalytic component of a multiprotein complex, polycomb repressive complex 2, which is involved in the trimethylation of histone H3 at lysine 27. In this study, we investigated the role and mechanisms of EZH2 in the differentiation of fibroblasts into myofibroblasts. We found that EZH2 was upregulated in the lungs of patients with IPF and in mice with bleomycin-induced lung fibrosis. The upregulation of EZH2 occurred in myofibroblasts. The inhibition of EZH2 by its inhibitor 3-deazaneplanocin A (DZNep) or an shRNA reduced the TGF-β1-induced differentiation of human lung fibroblasts into myofibroblasts, as demonstrated by the expression of the myofibroblast markers α-smooth muscle actin and fibronectin, and contractility. DZNep inhibited Smad2/3 nuclear translocation without affecting Smad2/3 phosphorylation. DZNep treatment attenuated bleomycin-induced pulmonary fibrosis in mice. We conclude that EZH2 induces the differentiation of fibroblasts to myofibroblasts by enhancing Smad2/3 nuclear translocation.
Collapse
Affiliation(s)
- Xiao Xiao
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma Department of Physiological Sciences, Lungberg-Kienlen Lung Biology and Toxicology Laboratory, Stillwater, Oklahoma
| | - Lakmini K Senavirathna
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma Department of Physiological Sciences, Lungberg-Kienlen Lung Biology and Toxicology Laboratory, Stillwater, Oklahoma
| | - Xuxu Gou
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma Department of Physiological Sciences, Lungberg-Kienlen Lung Biology and Toxicology Laboratory, Stillwater, Oklahoma
| | - Chaoqun Huang
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma Department of Physiological Sciences, Lungberg-Kienlen Lung Biology and Toxicology Laboratory, Stillwater, Oklahoma
| | - Yurong Liang
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma Department of Physiological Sciences, Lungberg-Kienlen Lung Biology and Toxicology Laboratory, Stillwater, Oklahoma
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma Department of Physiological Sciences, Lungberg-Kienlen Lung Biology and Toxicology Laboratory, Stillwater, Oklahoma
| |
Collapse
|
18
|
Baas R, Sijm A, van Teeffelen HAAM, van Es R, Vos HR, Marc Timmers HT. Quantitative Proteomics of the SMAD (Suppressor of Mothers against Decapentaplegic) Transcription Factor Family Identifies Importin 5 as a Bone Morphogenic Protein Receptor SMAD-specific Importin. J Biol Chem 2016; 291:24121-24132. [PMID: 27703004 DOI: 10.1074/jbc.m116.748582] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/03/2016] [Indexed: 01/11/2023] Open
Abstract
Gene-specific transcription factors (GSTFs) control gene transcription by DNA binding and specific protein complex recruitment, which regulates promoter accessibility for transcription initiation by RNA polymerase II. Mutations in the GSTFs Suppressor of Mothers Against Decapentaplegic 2 (SMAD2) and SMAD4 are frequently associated with colon and rectal carcinomas. These proteins play an important role in bone morphogenic protein (BMP) and transforming growth factor β (TGF-β) signaling pathways controlling cell fate and proliferation. To study the protein interactome of the SMAD protein family we generated a quantitative proteomics pipeline that allows for inducible expression of GFP-tagged SMAD proteins followed by affinity purification and quantitative mass spectrometry analysis. Data are available via ProteomeXchange with identifier PXD004529. The nuclear importin IPO5 was identified as a novel interacting protein of SMAD1. Overexpression of IPO5 in various cell lines specifically increases nuclear localization of BMP receptor-activated SMADs (R-SMADs) confirming a functional relationship between IPO5 and BMP but not TGF-β R-SMADs. Finally, we provide evidence that variation in length of the lysine stretch of the nuclear localization sequence is a determinant for importin specificity.
Collapse
Affiliation(s)
- Roy Baas
- From the Departments of Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine, University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Ayestha Sijm
- From the Departments of Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine, University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Hetty A A M van Teeffelen
- From the Departments of Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine, University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Robert van Es
- From the Departments of Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine, University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Harmjan R Vos
- From the Departments of Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine, University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - H Th Marc Timmers
- From the Departments of Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine, University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
19
|
Abstract
Transforming growth factor β (TGF-β) and related growth factors are secreted pleiotropic factors that play critical roles in embryogenesis and adult tissue homeostasis by regulating cell proliferation, differentiation, death, and migration. The TGF-β family members signal via heteromeric complexes of type I and type II receptors, which activate members of the Smad family of signal transducers. The main attribute of the TGF-β signaling pathway is context-dependence. Depending on the concentration and type of ligand, target tissue, and developmental stage, TGF-β family members transmit distinct signals. Deregulation of TGF-β signaling contributes to developmental defects and human diseases. More than a decade of studies have revealed the framework by which TGF-βs encode a context-dependent signal, which includes various positive and negative modifiers of the principal elements of the signaling pathway, the receptors, and the Smad proteins. In this review, we first introduce some basic components of the TGF-β signaling pathways and their actions, and then discuss posttranslational modifications and modulatory partners that modify the outcome of the signaling and contribute to its context-dependence, including small noncoding RNAs.
Collapse
Affiliation(s)
- Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94143
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
20
|
Quijano JC, Wisotzkey RG, Tran NL, Huang Y, Stinchfield MJ, Haerry TE, Shimmi O, Newfeld SJ. lolal Is an Evolutionarily New Epigenetic Regulator of dpp Transcription during Dorsal-Ventral Axis Formation. Mol Biol Evol 2016; 33:2621-32. [PMID: 27401231 PMCID: PMC5026256 DOI: 10.1093/molbev/msw132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Secreted ligands in the Dpp/BMP family drive dorsal–ventral (D/V) axis formation in all Bilaterian species. However, maternal factors regulating Dpp/BMP transcription in this process are largely unknown. We identified the BTB domain protein longitudinals lacking-like (lolal) as a modifier of decapentaplegic (dpp) mutations. We show that Lolal is evolutionarily related to the Trithorax group of chromatin regulators and that lolal interacts genetically with the epigenetic factor Trithorax-like during Dpp D/V signaling. Maternally driven LolalHA is found in oocytes and translocates to zygotic nuclei prior to the point at which dpp transcription begins. lolal maternal and zygotic mutant embryos display significant reductions in dpp, pMad, and zerknullt expression, but they are never absent. The data suggest that lolal is required to maintain dpp transcription during D/V patterning. Phylogenetic data revealed that lolal is an evolutionarily new gene present only in insects and crustaceans. We conclude that Lolal is the first maternal protein identified with a role in dpp D/V transcriptional maintenance, that Lolal and the epigenetic protein Trithorax-like are essential for Dpp D/V signaling and that the architecture of the Dpp D/V pathway evolved in the arthropod lineage after the separation from vertebrates via the incorporation of new genes such as lolal.
Collapse
Affiliation(s)
| | | | | | - Yunxian Huang
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Theodor E Haerry
- Center for Molecular Biology and Biotechnology, Florida Atlantic University
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
21
|
Xiong J, Hu Z, Wang T, Xu X, Liu J, Wu P, Che X, Li W. RUNX2 controls human IPO8 basal transcription in Saos-2 cells. Mol Med Rep 2016; 14:1418-24. [DOI: 10.3892/mmr.2016.5356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 05/09/2016] [Indexed: 11/06/2022] Open
|
22
|
Abstract
The transforming growth factor-β (TGF-β) family of cytokines figures prominently in regulation of embryonic development and adult tissue homeostasis from Drosophila to mammals. Genetic defects affecting TGF-β signaling underlie developmental disorders and diseases such as cancer in human. Therefore, delineating the molecular mechanism by which TGF-β regulates cell biology is critical for understanding normal biology and disease mechanisms. Forward genetic screens in model organisms and biochemical approaches in mammalian tissue culture were instrumental in initial characterization of the TGF-β signal transduction pathway. With complete sequence information of the genomes and the advent of RNA interference (RNAi) technology, genome-wide RNAi screening emerged as a powerful functional genomics approach to systematically delineate molecular components of signal transduction pathways. Here, we describe a protocol for image-based whole-genome RNAi screening aimed at identifying molecules required for TGF-β signaling into the nucleus. Using this protocol we examined >90 % of annotated Drosophila open reading frames (ORF) individually and successfully uncovered several novel factors serving critical roles in the TGF-β pathway. Thus cell-based high-throughput functional genomics can uncover new mechanistic insights on signaling pathways beyond what the classical genetics had revealed.
Collapse
|
23
|
Van Bortle K, Peterson AJ, Takenaka N, O'Connor MB, Corces VG. CTCF-dependent co-localization of canonical Smad signaling factors at architectural protein binding sites in D. melanogaster. Cell Cycle 2016; 14:2677-87. [PMID: 26125535 DOI: 10.1080/15384101.2015.1053670] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The transforming growth factor β (TGF-β) and bone morphogenetic protein (BMP) pathways transduce extracellular signals into tissue-specific transcriptional responses. During this process, signaling effector Smad proteins translocate into the nucleus to direct changes in transcription, but how and where they localize to DNA remain important questions. We have mapped Drosophila TGF-β signaling factors Mad, dSmad2, Medea, and Schnurri genome-wide in Kc cells and find that numerous sites for these factors overlap with the architectural protein CTCF. Depletion of CTCF by RNAi results in the disappearance of a subset of Smad sites, suggesting Smad proteins localize to CTCF binding sites in a CTCF-dependent manner. Sensitive Smad binding sites are enriched at low occupancy CTCF peaks within topological domains, rather than at the physical domain boundaries where CTCF may function as an insulator. In response to Decapentaplegic, CTCF binding is not significantly altered, whereas Mad, Medea, and Schnurri are redirected from CTCF to non-CTCF binding sites. These results suggest that CTCF participates in the recruitment of Smad proteins to a subset of genomic sites and in the redistribution of these proteins in response to BMP signaling.
Collapse
|
24
|
Importin-7 mediates memory consolidation through regulation of nuclear translocation of training-activated MAPK in Drosophila. Proc Natl Acad Sci U S A 2016; 113:3072-7. [PMID: 26929354 DOI: 10.1073/pnas.1520401113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Translocation of signaling molecules, MAPK in particular, from the cytosol to nucleus represents a universal key element in initiating the gene program that determines memory consolidation. Translocation mechanisms and their behavioral impact, however, remain to be determined. Here, we report that a highly conserved nuclear transporter, Drosophila importin-7 (DIM-7), regulates import of training-activated MAPK for consolidation of long-term memory (LTM). We show that silencing DIM-7 functions results in impaired LTM, whereas overexpression of DIM-7 enhances LTM. This DIM-7-dependent regulation of LTM is confined to a consolidation time window and in mushroom body neurons. Image data show that bidirectional alteration in DIM-7 expression results in proportional changes in the intensity of training-activated MAPK accumulated within the nuclei of mushroom body neurons during LTM consolidation. Such DIM-7-regulated nuclear accumulation of activated MAPK is observed only in the training specified for LTM induction and determines the amplitude, but not the time course, of memory consolidation.
Collapse
|
25
|
Wilkes MC, Repellin CE, Kang JH, Andrianifahanana M, Yin X, Leof EB. Sorting nexin 9 differentiates ligand-activated Smad3 from Smad2 for nuclear import and transforming growth factor β signaling. Mol Biol Cell 2015; 26:3879-91. [PMID: 26337383 PMCID: PMC4626071 DOI: 10.1091/mbc.e15-07-0545] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 08/27/2015] [Indexed: 01/23/2023] Open
Abstract
Sorting nexin 9 (SNX9) is shown to differentiate Smad3 from Smad2 nuclear delivery by mediating the association of phosphorylated Smad3 with importin 8 and the nuclear membrane. While the absence of SNX9 had negligible effects on transforming growth factor β receptor activity or Smad2 signaling, Smad3-dependent targets and phenotypes were inhibited. Transforming growth factor β (TGFβ) is a pleiotropic protein secreted from essentially all cell types and primary tissues. While TGFβ’s actions reflect the activity of a number of signaling networks, the primary mediator of TGFβ responses are the Smad proteins. Following receptor activation, these cytoplasmic proteins form hetero-oligomeric complexes that translocate to the nucleus and affect gene transcription. Here, through biological, biochemical, and immunofluorescence approaches, sorting nexin 9 (SNX9) is identified as being required for Smad3-dependent responses. SNX9 interacts with phosphorylated (p) Smad3 independent of Smad2 or Smad4 and promotes more rapid nuclear delivery than that observed independent of ligand. Although SNX9 does not bind nucleoporins Nup153 or Nup214 or some β importins (Imp7 or Impβ), it mediates the association of pSmad3 with Imp8 and the nuclear membrane. This facilitates nuclear translocation of pSmad3 but not SNX9.
Collapse
Affiliation(s)
- Mark C Wilkes
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Claire E Repellin
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Jeong-Han Kang
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Mahefatiana Andrianifahanana
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Xueqian Yin
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Edward B Leof
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905
| |
Collapse
|
26
|
Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature 2015; 524:361-5. [PMID: 26168401 PMCID: PMC5086585 DOI: 10.1038/nature14587] [Citation(s) in RCA: 640] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/20/2015] [Indexed: 12/11/2022]
Abstract
Activation of cellular stress response pathways to maintain metabolic homeostasis is emerging as a critical growth and survival mechanism in many cancers. The pathogenesis of pancreatic ductal adenocarcinoma (PDA) requires high levels of autophagy, a conserved self-degradative process. However, the regulatory circuits that activate autophagy and reprogram PDA cell metabolism are unknown. Here we show that autophagy induction in PDA occurs as part of a broader transcriptional program that coordinates activation of lysosome biogenesis and function, and nutrient scavenging, mediated by the MiT/TFE family of transcription factors. In human PDA cells, the MiT/TFE proteins--MITF, TFE3 and TFEB--are decoupled from regulatory mechanisms that control their cytoplasmic retention. Increased nuclear import in turn drives the expression of a coherent network of genes that induce high levels of lysosomal catabolic function essential for PDA growth. Unbiased global metabolite profiling reveals that MiT/TFE-dependent autophagy-lysosome activation is specifically required to maintain intracellular amino acid pools. These results identify the MiT/TFE proteins as master regulators of metabolic reprogramming in pancreatic cancer and demonstrate that transcriptional activation of clearance pathways converging on the lysosome is a novel hallmark of aggressive malignancy.
Collapse
|
27
|
Boye A, Wu C, Jiang Y, Wang J, Wu J, Yang X, Yang Y. Compound Astragalus and Salvia miltiorrhiza extracts modulate MAPK-regulated TGF-β/Smad signaling in hepatocellular carcinoma by multi-target mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2015; 169:219-228. [PMID: 25934513 DOI: 10.1016/j.jep.2015.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 04/01/2015] [Accepted: 04/13/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus membranaceus Bunge (Leguminosae) and Salvia miltiorrhiza Bunge (Lamiaceae) are two important Chinese herbs with a long history of extensive ethnobotanical usage in the treatment of liver-related diseases over many centuries. Presently, these two herbs are being used either as a single herbal formulation or a composite formula for the treatment of liver related conditions. In response, recent studies on these two herbs have focused on elucidating their mechanisms of action, particularly with regards to their anti-hepatocarcinogenic effects. Previously, we have reported that Compound Astragalus and Salvia miltiorrhiza extract (CASE), a synergized composite extract from Astragalus membranaceus and Salvia miltiorrhiza ameliorates liver fibrosis and hepatocellular carcinoma (HCC) by modulating the TGF-β/Smad pathway. Meanwhile, MAPK activation and MAPK-dependent linker phosphorylation of Smad2/3 and their preferential nuclear import are crucial for overall oncogenic role of TGF-β/Smad signaling in HCC. To elucidate further, we studied the effect of CASE on the MAPK pathway and how it affects MAPK-dependent regulation of TGF-β/Smad signaling using both cell and animal models of HCC. MATERIALS AND METHODS We used immunofluorescence and western blot techniques to monitor effect of CASE on the activation of the MAPKs (pERK, pJNK and pp38) in TGF-β1-stimulated hepatic stellate cells (HSCs), HepG2 cells and also diethylnitrosamine (DEN)-induced HCC in rats. Also phosphorylation and subcellular distribution of pSmad2/3, Smad4 and Imp7/8 in TGF-β1-stimulated HSC and HepG2 cells were monitored. The expression of pERK, pJNK, pp38 and PAI-1 gene were monitored by using western blot technique. The effect of CASE on domain-specific phosphorylation of Smad2/3 and their subcellular distribution, and the expression of Smad4 and its subcellular distribution in TGF-β1-stimulated HSCs and HepG2 cells were evaluated by using immunofluorescence technique. And the expression of Imp7/8 and their subcellular distribution were assessed by both immunofluorescence and western blot techniques, while PAI-1 gene expression was assessed by western blot RESULTS In vitro, CASE in a concentration-dependent manner increased the expression of pp38 but decreased the expression of pERK and pJNK; however, in vivo, CASE in a dose dependent manner decreased the expression of pERK, pJNK as well as pp38. Also, CASE concentration dependently inhibited pSmad2C/L, pSmad3L, Smad4, Imp7/8 and their nuclear import; it had no effect on pSmad3C in HepG2 cells; significantly decreased PAI-1 gene expression in both in vitro and in vivo. CONCLUSIONS CASE blocked MAPK activation, MAPK-dependent linker phosphorylation of Smad2/3, Smad4 expression, Imp7 expression and their nuclear import leading to significant down-regulation of PAI-1 gene expression; further highlighting the multi-target anti-HCC effect of CASE and its potential drug candidature.
Collapse
Affiliation(s)
- Alex Boye
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Chao Wu
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Yufeng Jiang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Jiyu Wang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Jiajun Wu
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiaochuan Yang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Yan Yang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
28
|
Jiang Y, Wu C, Boye A, Wu J, Wang J, Yang X, Yang Y. MAPK inhibitors modulate Smad2/3/4 complex cyto-nuclear translocation in myofibroblasts via Imp7/8 mediation. Mol Cell Biochem 2015; 406:255-62. [PMID: 25968067 DOI: 10.1007/s11010-015-2443-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/06/2015] [Indexed: 01/06/2023]
Abstract
Mitogen-activated protein kinase (MAPK) pathway-dependent linker phosphorylation of Smad2/3 and subsequent formation of Smad2/3/4 complex and its nuclear translocation are crucial for dysregulated transforming growth factor beta (TGF)-β/Smad signaling in liver fibrosis. Abrogation of this critical step of TGF-β/Smad signaling leading to liver fibrosis could provide new insights for future therapy, but the mechanisms remain incompletely understood. In pursuit, we investigated the subcellular expression and nuclear trafficking of the rate limiting Smad2/3/4 complex in exogenous TGF-β1-stimulated myofibroblasts (MFBs) using three MAPK-specific inhibitors. Our results showed that exogenous TGF-β1 stimulation of MFBs produced both increased protein expression and nuclear translocation of phosphorylated (p)-Smad2C/L, oncogenic pSmad3L, Smad4, importin7/8 (Imp7/8), and plasminogen activator inhibitor (PAI)-1 (Protein and mRNA), while decreased Smad7 protein expression. However, the MAPK-specific inhibitors differentially reversed these observations; for instance, ERK-specific inhibitor blocked the expression and nuclear translocation of pSmad2C/L, while both JNK and p38-specific inhibitors blocked the expression and nuclear translocation of pSmad2C/L and oncogenic pSmad3L. The MAPK-specific inhibitors had no significant effect on the total protein expression of Smad4, but rather significantly blocked its nuclear translocation. All the MAPK-specific inhibitors restored Smad7 expression and also decreased Imp7/8 and PAI-1 (Protein and mRNA) expression. Evidently, the MAPK-specific inhibitors blocked Smad2/3/4 complex formation via restoration of inhibitory Smad7 expression and blockade of Smad3L phosphorylation, while they blocked nuclear translocation of Smad2/3/4 complex through inhibition of Imp7/8 leading to decreased PAI-1 (Protein and mRNA) expression.
Collapse
Affiliation(s)
- Yufeng Jiang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, 230032, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Zeng Z, de Gorter DJJ, Kowalski M, ten Dijke P, Shimmi O. Ter94/VCP is a novel component involved in BMP signaling. PLoS One 2014; 9:e114475. [PMID: 25469707 PMCID: PMC4255028 DOI: 10.1371/journal.pone.0114475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/10/2014] [Indexed: 11/18/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), a subgroup of the transforming growth factor (TGF)-β family, transduce their signal through multiple components downstream of their receptors. Even though the components involved in the BMP signaling pathway have been intensely studied, many molecules mediating BMP signaling remain to be addressed. To identify novel components that participate in BMP signaling, RNA interference (RNAi)-based screening was established by detecting phosphorylated Mad (pMad) in Drosophila S2 cells. Ter94, a member of the family of AAA ATPases, was identified as a novel mediator of BMP signaling, which is required for the phosphorylation of Mad in Drosophila S2 cells. Moreover, the mammalian orthlog of Ter94 valosin-containing protein (VCP) plays a critical role in the BMP-Smad1/5/8 signaling pathway in mammalian cells. Genetic evidence suggests that Ter94 is involved in the dorsal-ventral patterning of the Drosophila early embryo through regulating decapentaplegic (Dpp)/BMP signals. Taken together, our data suggest that Ter94/VCP appears to be an evolutionarily conserved component that regulates BMP-Smad1/5/8 signaling.
Collapse
Affiliation(s)
- Zhao Zeng
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - David J. J. de Gorter
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maria Kowalski
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Peter ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Centre, Leiden, The Netherlands
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
30
|
Gupta GD, Dey G, MG S, Ramalingam B, Shameer K, Thottacherry JJ, Kalappurakkal JM, Howes MT, Chandran R, Das A, Menon S, Parton RG, Sowdhamini R, Thattai M, Mayor S. Population distribution analyses reveal a hierarchy of molecular players underlying parallel endocytic pathways. PLoS One 2014; 9:e100554. [PMID: 24971745 PMCID: PMC4074053 DOI: 10.1371/journal.pone.0100554] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 05/28/2014] [Indexed: 12/11/2022] Open
Abstract
Single-cell-resolved measurements reveal heterogeneous distributions of clathrin-dependent (CD) and -independent (CLIC/GEEC: CG) endocytic activity in Drosophila cell populations. dsRNA-mediated knockdown of core versus peripheral endocytic machinery induces strong changes in the mean, or subtle changes in the shapes of these distributions, respectively. By quantifying these subtle shape changes for 27 single-cell features which report on endocytic activity and cell morphology, we organize 1072 Drosophila genes into a tree-like hierarchy. We find that tree nodes contain gene sets enriched in functional classes and protein complexes, providing a portrait of core and peripheral control of CD and CG endocytosis. For 470 genes we obtain additional features from separate assays and classify them into early- or late-acting genes of the endocytic pathways. Detailed analyses of specific genes at intermediate levels of the tree suggest that Vacuolar ATPase and lysosomal genes involved in vacuolar biogenesis play an evolutionarily conserved role in CG endocytosis.
Collapse
Affiliation(s)
- Gagan D. Gupta
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Gautam Dey
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Swetha MG
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Balaji Ramalingam
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Khader Shameer
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Joseph Jose Thottacherry
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Joseph Mathew Kalappurakkal
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Mark T. Howes
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Ruma Chandran
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Anupam Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Sindhu Menon
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Robert G. Parton
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - R. Sowdhamini
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Mukund Thattai
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS/GKVK Campus, Bangalore, India
| |
Collapse
|
31
|
Kimura M, Imamoto N. Biological significance of the importin-β family-dependent nucleocytoplasmic transport pathways. Traffic 2014; 15:727-48. [PMID: 24766099 DOI: 10.1111/tra.12174] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/22/2014] [Accepted: 04/22/2014] [Indexed: 12/19/2022]
Abstract
Importin-β family proteins (Imp-βs) are nucleocytoplasmic transport receptors (NTRs) that import and export proteins and RNAs through the nuclear pores. The family consists of 14-20 members depending on the biological species, and each member transports a specific group of cargoes. Thus, the Imp-βs mediate multiple, parallel transport pathways that can be regulated separately. In fact, the spatiotemporally differential expressions and the functional regulations of Imp-βs have been reported. Additionally, the biological significance of each pathway has been characterized by linking the function of a member of Imp-βs to a cellular consequence. Connecting these concepts, the regulation of the transport pathways conceivably induces alterations in the cellular physiological states. However, few studies have linked the regulation of an importin-β family NTR to an induced cellular response and the corresponding cargoes, despite the significance of this linkage in comprehending the biological relevance of the transport pathways. This review of recent reports on the regulation and biological functions of the Imp-βs highlights the significance of the transport pathways in physiological contexts and points out the possibility that the identification of yet unknown specific cargoes will reinforce the importance of transport regulation.
Collapse
Affiliation(s)
- Makoto Kimura
- Cellular Dynamics Laboratory, RIKEN, Hirosawa 2-1, Wako, Saitama, 351-0198, Japan
| | | |
Collapse
|
32
|
Twyffels L, Wauquier C, Soin R, Decaestecker C, Gueydan C, Kruys V. A masked PY-NLS in Drosophila TIS11 and its mammalian homolog tristetraprolin. PLoS One 2013; 8:e71686. [PMID: 23951221 PMCID: PMC3739726 DOI: 10.1371/journal.pone.0071686] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 07/07/2013] [Indexed: 12/11/2022] Open
Abstract
Many RNA-binding proteins (RBPs) dynamically shuttle between the nucleus and the cytoplasm, often exerting different functions in each compartment. Therefore, the nucleo-cytoplasmic distribution of RBPs has a strong impact on their activity. Here we describe the localization and the shuttling properties of the tandem zinc finger RBP dTIS11, which is the Drosophila homolog of mammalian TIS11 proteins. Drosophila and mammalian TIS11 proteins act as destabilizing factors in ARE-mediated decay. At equilibrium, dTIS11 is concentrated mainly in the cytoplasm. We show that dTIS11 is a nucleo-cytoplasmic shuttling protein whose nuclear export is mediated by the exportin CRM1 through the recognition of a nuclear export signal (NES) located in a different region comparatively to its mammalian homologs. We also identify a cryptic Transportin-dependent PY nuclear localization signal (PY-NLS) in the tandem zinc finger region of dTIS11 and show that it is conserved across the TIS11 protein family. This NLS partially overlaps the second zinc finger ZnF2. Importantly, mutations disrupting the capacity of the ZnF2 to coordinate a Zinc ion unmask dTIS11 and TTP NLS and promote nuclear import. All together, our results indicate that the nuclear export of TIS11 proteins is mediated by CRM1 through diverging NESs, while their nuclear import mechanism may rely on a highly conserved PY-NLS whose activity is negatively regulated by ZnF2 folding.
Collapse
Affiliation(s)
- Laure Twyffels
- Laboratoire de Biologie Moléculaire du Gène, Faculté des Sciences, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Corinne Wauquier
- Laboratoire de Biologie Moléculaire du Gène, Faculté des Sciences, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Romuald Soin
- Laboratoire de Biologie Moléculaire du Gène, Faculté des Sciences, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Christine Decaestecker
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
- Laboratory of Image Synthesis and Analysis - Ecole Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Cyril Gueydan
- Laboratoire de Biologie Moléculaire du Gène, Faculté des Sciences, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Véronique Kruys
- Laboratoire de Biologie Moléculaire du Gène, Faculté des Sciences, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| |
Collapse
|
33
|
Liu ZC, Geisbrecht ER. "Importin" signaling roles for import proteins: the function of Drosophila importin-7 (DIM-7) in muscle-tendon signaling. Cell Adh Migr 2012; 6:4-12. [PMID: 22647935 DOI: 10.4161/cam.19774] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The formation of a mature myotendinous junction (MTJ) between a muscle and its site of attachment is a highly regulated process that involves myofiber migration, cell-cell signaling, and culminates with the stable adhesion between the adjacent muscle-tendon cells. Improper establishment or maintenance of muscle-tendon attachment sites results in a decrease in force generation during muscle contraction and progressive muscular dystrophies in vertebrate models. Many studies have demonstrated the important role of the integrins and integrin-associated proteins in the formation and maintenance of the MTJ. We recently demonstrated that moleskin (msk), the gene that encodes for Drosophila importin-7 (DIM-7), is required for the proper formation of muscle-tendon adhesion sites in the developing embryo. Further studies demonstrated an enrichment of DIM-7 to the ends of muscles where the muscles attach to their target tendon cells. Genetic analysis supports a model whereby msk is required in the muscle and signals via the secreted epidermal growth factor receptor (Egfr) ligand Vein to regulate tendon cell maturation. These data demonstrate a novel role for the canonical nuclear import protein DIM-7 in establishment of the MTJ.
Collapse
Affiliation(s)
- Ze Cindy Liu
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri, Kansas City, MO, USA
| | | |
Collapse
|
34
|
Abstract
The canonical TGF-β/Smad signaling pathway was delineated in the mid 90s and enriched over the past decade with many findings about its specificity, regulation, networking, and malfunctions in disease. However, a growing understanding of the chromatin status of a critical class of TGF-β target genes - the genes controlling differentiation of embryonic stem cells - recently prompted a reexamination of this pathway and its critical role in the regulation of stem cell differentiation. The new findings reveal master regulators of the pluripotent state set the stage for Smad-mediated activation of master regulators of the next differentiation stage. Furthermore, a novel branch of the TGF-β/Smad pathway has been identified in which a chromatin-reading Smad complex makes the master differentiation genes accessible to canonical Smad complexes for transcriptional activation. These findings provide exciting new insights into the global role of TGF-β signaling in the regulators of stem cell fate.
Collapse
Affiliation(s)
- Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | |
Collapse
|
35
|
Xu P, Liu J, Derynck R. Post-translational regulation of TGF-β receptor and Smad signaling. FEBS Lett 2012; 586:1871-84. [PMID: 22617150 DOI: 10.1016/j.febslet.2012.05.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 05/06/2012] [Accepted: 05/07/2012] [Indexed: 01/17/2023]
Abstract
TGF-β family signaling through Smads is conceptually a simple and linear signaling pathway, driven by sequential phosphorylation, with type II receptors activating type I receptors, which in turn activate R-Smads. Nevertheless, TGF-β family proteins induce highly complex programs of gene expression responses that are extensively regulated, and depend on the physiological context of the cells. Regulation of TGF-β signaling occurs at multiple levels, including TGF-β activation, formation, activation and destruction of functional TGF-β receptor complexes, activation and degradation of Smads, and formation of Smad transcription complexes at regulatory gene sequences that cooperate with a diverse set of DNA binding transcription factors and coregulators. Here we discuss recent insights into the roles of post-translational modifications and molecular interaction networks in the functions of receptors and Smads in TGF-β signal responses. These layers of regulation demonstrate how a simple signaling system can be coopted to exert exquisitely regulated, complex responses.
Collapse
Affiliation(s)
- Pinglong Xu
- Department of Cell and Tissue Biology, Programs in Cell Biology and Developmental Biology, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
36
|
Chatel G, Fahrenkrog B. Dynamics and diverse functions of nuclear pore complex proteins. Nucleus 2012; 3:162-71. [PMID: 22555605 DOI: 10.4161/nucl.19674] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nuclear pore complexes (NPCs) are best known for their central role in controlling the molecular trafficking between the cytoplasm and the nucleus. NPCs are assembled from about 30 different proteins and a growing body of evidence suggests that these nucleoporins are not only acting in the context of NPCs, but also in the nucleoplasm and cytoplasm. In this context it is well accepted that a set of nucleoporins are important regulators of a variety of mitotic processes, including kinetochore assembly, spindle checkpoint control and cytokinesis, whereas others associate with chromatin and administer gene expression. However, the functional importance of nucleoporins go far beyond these roles and this review will provide an overview of the latest insights into the versatility of metazoan nucleoporins with an emphasis on their roles in cell migration, cellular signaling and tissue-specific activities.
Collapse
Affiliation(s)
- Guillaume Chatel
- Institute of Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi, Belgium
| | | |
Collapse
|
37
|
Sundqvist A, Ten Dijke P, van Dam H. Key signaling nodes in mammary gland development and cancer: Smad signal integration in epithelial cell plasticity. Breast Cancer Res 2012; 14:204. [PMID: 22315972 PMCID: PMC3496114 DOI: 10.1186/bcr3066] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Smad proteins are the key intermediates of transforming growth factor-beta (TGF-β) signaling during development and in tissue homeostasis. Pertubations in TGF-β/Smad signaling have been implicated in cancer and other diseases. In the cell nucleus, Smad complexes trigger cell type- and context-specific transcriptional programs, thereby transmitting and integrating signals from a variety of ligands of the TGF-β superfamily and other stimuli in the cell microenvironment. The actual transcriptional and biological outcome of Smad activation critically depends on the genomic integrity and the modification state of genome and chromatin of the cell. The cytoplasmic and nuclear Smads can also modulate the activity of other signal transducers and enzymes such as microRNA-processing factors. In the case of breast cancer, the role of Smads in epithelial plasticity, tumor-stroma interactions, invasion, and metastasis seems of particular importance.
Collapse
Affiliation(s)
- Anders Sundqvist
- Ludwig Institute for Cancer Research, Uppsala University, Box 595, 75124, Uppsala, Sweden
| | | | | |
Collapse
|
38
|
Blahna MT, Hata A. Smad-mediated regulation of microRNA biosynthesis. FEBS Lett 2012; 586:1906-12. [PMID: 22306316 DOI: 10.1016/j.febslet.2012.01.041] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/19/2012] [Indexed: 12/21/2022]
Abstract
microRNAs (miRNAs) are small non-coding RNAs conserved in metazoans. Depletion of miRNAs results in embryonic lethality, suggesting they are essential for embryogenesis. Similarly, pathways induced by growth factors of the transforming growth factor β (TGF-β) superfamily control cell growth, differentiation, and development. Recently Smad proteins, the signal transducers of the TGF-β pathway, were found to regulate miRNA expression, which, in turn, affects expression of numerous proteins. Smads modulate miRNA expression through both transcriptional and post-transcriptional mechanisms illustrating the complexity of gene regulation by TGF-β. In this chapter we summarize the current knowledge of mechanisms underlying Smad-mediated regulation of miRNA biogenesis.
Collapse
Affiliation(s)
- Matthew T Blahna
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
39
|
Mechanism and regulation of nucleocytoplasmic trafficking of smad. Cell Biosci 2011; 1:40. [PMID: 22204445 PMCID: PMC3292837 DOI: 10.1186/2045-3701-1-40] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 12/28/2011] [Indexed: 11/24/2022] Open
Abstract
Smad proteins are the intracellular mediators of transforming growth factor β (TGF-β) signaling. Smads function as transcription factors and their activities require carboxyl-terminal phosphorylation by TGF-β receptor kinases which are embedded in the cell membrane. Therefore, the translocation of activated Smads from the cytoplasm into the nucleus is a rate-limiting step in TGF-β signal transduction into the nucleus. On the other hand, the export of Smads out of the nucleus turns off TGF-β effect. Such spatial control of Smad ensures a tight regulation of TGF-β target genes. Several cross-talk pathways have been shown to affect TGF-β signaling by impairing nuclear translocation of Smad, exemplifying the biological importance of the nuclear transport process. Many laboratories have investigated the underlying molecular mechanism of Smad nucleocytoplasmic translocation, combining genetics, biochemistry and sophisticated live cell imaging approaches. The last few years have witnessed the elucidation of several key players in Smad nuclear transport, most importantly the karyopherins that carry Smads across the nuclear envelope and nuclear pore proteins that facilitate the trans-nuclear envelope movement. The foundation is now set to further elucidate how the nuclear transport process is regulated and exploit such knowledge to manipulate TGF-β signaling. In this review we will discuss the current understanding of the molecular machinery responsible for nuclear import and export of Smads.
Collapse
|
40
|
Neumuüller RA, Perrimon N. Where gene discovery turns into systems biology: genome-scale RNAi screens in Drosophila. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2011; 3:471-8. [PMID: 21197652 PMCID: PMC3560415 DOI: 10.1002/wsbm.127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Systems biology aims to describe the complex interplays between cellular building blocks which, in their concurrence, give rise to the emergent properties observed in cellular behaviors and responses. This approach tries to determine the molecular players and the architectural principles of their interactions within the genetic networks that control certain biological processes. Large-scale loss-of-function screens, applicable in various different model systems, have begun to systematically interrogate entire genomes to identify the genes that contribute to a certain cellular response. In particular, RNA interference (RNAi)-based high-throughput screens have been instrumental in determining the composition of regulatory systems and paired with integrative data analyses have begun to delineate the genetic networks that control cell biological and developmental processes. Through the creation of tools for both, in vitro and in vivo genome-wide RNAi screens, Drosophila melanogaster has emerged as one of the key model organisms in systems biology research and over the last years has massively contributed to and hence shaped this discipline. WIREs Syst Biol Med 2011 3 471-478 DOI: 10.1002/wsbm.127
Collapse
Affiliation(s)
- Ralph A. Neumuüller
- Department of Genetics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Abstract
The level of TGF-β/bone morphogenetic protein (BMP) signaling through Smad is tightly regulated to ensure proper embryonic patterning and homeostasis. Here we show that Smad activation by TGF-β/BMP is blocked by a highly conserved phosphorylation event in the α-helix 1 region of Smad [T312 in Drosophila Smad1 (MAD)]. α-helix 1 phosphorylation reduces Smad interaction with TGF-β/BMP receptor kinase and affects all receptor-activated Smads except Smad3. Tissue culture and transgenic studies in Drosophila further demonstrate that the biological activity of MAD is repressed by T312 phosphorylation in vivo. Through RNAi screening of the kinome, we have identified Misshapen (Msn) and the mammalian orthologs TNIK, MINK1, and MAP4K4 as the kinases responsible for α-helix 1 phosphorylation. Targeted expression of an active form of Msn in the wing imaginal disk disrupted activation of endogenous MAD by Dpp and expression of the Dpp/MAD target gene. Msn kinases belong to the Ste20 kinase family that has been shown to act as MAP kinase kinase kinase kinase (MAP4K). Our findings thus reveal a function of Msn independent of its impact on MAP kinase cascades. This Smad inhibition mechanism by Msn likely has important implications for development and disease.
Collapse
|
42
|
Role of Smads in TGFβ signaling. Cell Tissue Res 2011; 347:21-36. [PMID: 21643690 DOI: 10.1007/s00441-011-1190-x] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 05/10/2011] [Indexed: 02/07/2023]
Abstract
Transforming growth factor-β (TGFβ) is the prototype for a large family of pleiotropic factors that signal via heterotetrameric complexes of type I and type II serine/threonine kinase receptors. Important intracellular mediators of TGFβ signaling are members of the Smad family. Smad2 and 3 are activated by C-terminal receptor-mediated phosphorylation, whereafter they form complexes with Smad4 and are translocated to the nucleus where they, in cooperation with other transcription factors, co-activators and co-repressors, regulate the transcription of specific genes. Smads have key roles in exerting TGFβ-induced programs leading to cell growth arrest and epithelial-mesenchymal transition. The activity and stability of Smad molecules are carefully regulated by a plethora of post-translational modifications, including phosphorylation, ubiquitination, sumoylation, acetylation and poly(ADP)-ribosylation. The Smad function has been shown to be perturbed in certain diseases such as cancer.
Collapse
|
43
|
Chen J, Liu MY, Parish CR, Chong BH, Khachigian L. Nuclear import of early growth response-1 involves importin-7 and the novel nuclear localization signal serine-proline-serine. Int J Biochem Cell Biol 2011; 43:905-12. [DOI: 10.1016/j.biocel.2011.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 03/04/2011] [Accepted: 03/09/2011] [Indexed: 01/12/2023]
|
44
|
Guest ST, Yu J, Liu D, Hines JA, Kashat MA, Finley RL. A protein network-guided screen for cell cycle regulators in Drosophila. BMC SYSTEMS BIOLOGY 2011; 5:65. [PMID: 21548953 PMCID: PMC3113730 DOI: 10.1186/1752-0509-5-65] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 05/06/2011] [Indexed: 11/15/2022]
Abstract
Background Large-scale RNAi-based screens are playing a critical role in defining sets of genes that regulate specific cellular processes. Numerous screens have been completed and in some cases more than one screen has examined the same cellular process, enabling a direct comparison of the genes identified in separate screens. Surprisingly, the overlap observed between the results of similar screens is low, suggesting that RNAi screens have relatively high levels of false positives, false negatives, or both. Results We re-examined genes that were identified in two previous RNAi-based cell cycle screens to identify potential false positives and false negatives. We were able to confirm many of the originally observed phenotypes and to reveal many likely false positives. To identify potential false negatives from the previous screens, we used protein interaction networks to select genes for re-screening. We demonstrate cell cycle phenotypes for a significant number of these genes and show that the protein interaction network is an efficient predictor of new cell cycle regulators. Combining our results with the results of the previous screens identified a group of validated, high-confidence cell cycle/cell survival regulators. Examination of the subset of genes from this group that regulate the G1/S cell cycle transition revealed the presence of multiple members of three structurally related protein complexes: the eukaryotic translation initiation factor 3 (eIF3) complex, the COP9 signalosome, and the proteasome lid. Using a combinatorial RNAi approach, we show that while all three of these complexes are required for Cdk2/Cyclin E activity, the eIF3 complex is specifically required for some other step that limits the G1/S cell cycle transition. Conclusions Our results show that false positives and false negatives each play a significant role in the lack of overlap that is observed between similar large-scale RNAi-based screens. Our results also show that protein network data can be used to minimize false negatives and false positives and to more efficiently identify comprehensive sets of regulators for a process. Finally, our data provides a high confidence set of genes that are likely to play key roles in regulating the cell cycle or cell survival.
Collapse
Affiliation(s)
- Stephen T Guest
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Cytoplasm-to-nucleus translocation of Smad is a fundamental step in transforming growth factor beta (TGF-beta) signal transduction. Here we identify a subset of nucleoporins that, in conjunction with Msk (Drosophila Imp7/8), specifically mediate activation-induced nuclear translocation of MAD (Drosophila Smad1) but not the constitutive import of proteins harboring a classic nuclear localization signal (cNLS) or the spontaneous nuclear import of Medea (Drosophila Smad4). Surprisingly, many of these nucleoporins, including Sec13, Nup75, Nup93, and Nup205, are scaffold nucleoporins considered important for the overall integrity of the nuclear pore complex (NPC) but not known to have cargo-specific functions. We demonstrate that the roles of these nucleoporins in supporting Smad nuclear import are separate from their previously assigned functions in NPC assembly. Furthermore, we uncovered novel pathway-specific functions of Sec13 and Nup93; both Sec13 and Nup93 are able to preferentially interact with the phosphorylated/activated form of MAD, and Nup93 acts to recruit the importin Msk to the nuclear periphery. These findings, together with the observation that Sec13 and Nup93 could interact directly with Msk, suggest their direct involvement in the nuclear import of MAD. Thus, we have delineated the nucleoporin requirement of MAD nuclear import, reflecting a unique trans-NPC mechanism.
Collapse
|
46
|
Higashi-Kovtun ME, Mosca TJ, Dickman DK, Meinertzhagen IA, Schwarz TL. Importin-beta11 regulates synaptic phosphorylated mothers against decapentaplegic, and thereby influences synaptic development and function at the Drosophila neuromuscular junction. J Neurosci 2010; 30:5253-68. [PMID: 20392948 PMCID: PMC2881940 DOI: 10.1523/jneurosci.3739-09.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 02/11/2010] [Accepted: 03/01/2010] [Indexed: 11/21/2022] Open
Abstract
Importin proteins act both at the nuclear pore to promote substrate entry and in the cytosol during signal trafficking. Here, we describe mutations in the Drosophila gene importin-beta11, which has not previously been analyzed genetically. Mutants of importin-beta11 died as late pupae from neuronal defects, and neuronal importin-beta11 was present not only at nuclear pores but also in the cytosol and at synapses. Neurons lacking importin-beta11 were viable and properly differentiated but exhibited discrete defects. Synaptic transmission was defective in adult photoreceptors and at larval neuromuscular junctions (NMJs). Mutant photoreceptor axons formed grossly normal projections and synaptic terminals in the brain, but synaptic arbors on larval muscles were smaller while still containing appropriate synaptic components. Bone morphogenic protein (BMP) signaling was the apparent cause of the observed NMJ defects. Importin-beta11 interacted genetically with the BMP pathway, and at mutant synaptic boutons, a key component of this pathway, phosphorylated mothers against decapentaplegic (pMAD), was reduced. Neuronal expression of an importin-beta11 transgene rescued this phenotype as well as the other observed neuromuscular phenotypes. Despite the loss of synaptic pMAD, pMAD persisted in motor neuron nuclei, suggesting a specific impairment in the local function of pMAD. Restoring levels of pMAD to mutant terminals via expression of constitutively active type I BMP receptors or by reducing retrograde transport in motor neurons also restored synaptic strength and morphology. Thus, importin-beta11 function interacts with the BMP pathway to regulate a pool of pMAD that must be present at the presynapse for its proper development and function.
Collapse
Affiliation(s)
- Misao E. Higashi-Kovtun
- F. M. Kirby Center for Neurobiology, Children's Hospital, Boston, Massachusetts 02115
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Timothy J. Mosca
- F. M. Kirby Center for Neurobiology, Children's Hospital, Boston, Massachusetts 02115
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Dion K. Dickman
- F. M. Kirby Center for Neurobiology, Children's Hospital, Boston, Massachusetts 02115
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Ian A. Meinertzhagen
- Department of Psychology, Life Sciences Centre, Dalhousie University, Halifax, Nova Scotia B3H 4J1, Canada
| | - Thomas L. Schwarz
- F. M. Kirby Center for Neurobiology, Children's Hospital, Boston, Massachusetts 02115
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, and
| |
Collapse
|
47
|
Abstract
Hypophosphatemia leads to rickets and osteomalacia, the latter of which results in decreased biomechanical integrity of bones, accompanied by poor fracture healing. Impaired phosphate-dependent apoptosis of hypertrophic chondrocytes is the molecular basis for rickets. However, the underlying pathophysiology of impaired fracture healing has not been characterized previously. To address the role of phosphate in fracture repair, mice were placed on a phosphate-restricted diet 2 days prior to or 3 days after induction of a mid-diaphyseal femoral fracture to assess the effects of phosphate deficiency on the initial recruitment of mesenchymal stem cells and their subsequent differentiation. Histologic and micro-computed tomographic (microCT) analyses demonstrated that both phosphate restriction models dramatically impaired fracture healing primarily owing to a defect in differentiation along the chondrogenic lineage. Based on Sox9 and Sox5 mRNA levels, neither the initial recruitment of cells to the callus nor their lineage commitment was effected by hypophosphatemia. However, differentiation of these cells was impaired in association with impaired bone morphogenetic protein (BMP) signaling. In vivo ectopic bone-formation assays and in vitro investigations in ST2 stromal cells confirmed that phosphate restriction leads to BMP-2 resistance. Marrow ablation studies demonstrate that hypophosphatemia has different effects on injury-induced intramembranous bone formation compared with endochondral bone formation. Thus phosphate plays an important role in the skeleton that extends beyond mineralized matrix formation and growth plate maturation and is critical for endochondral bone repair.
Collapse
|
48
|
Regulation of pri-miRNA Processing Through Smads. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 700:15-27. [PMID: 21755469 DOI: 10.1007/978-1-4419-7823-3_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
microRNAs (miRNAs) are small (∼22 nucleotides (nt)), noncoding RNAs that play a critical role in diverse biological functions by modulating mRNA stability and translational control. Numerous miRNA profiling studies have indicated that the levels of miRNAs are tightly controlled during developmental stages and various pathophysiological and physiological conditions. Following transcription, the long primary miRNA transcript undergoes a series of coordinated maturation steps to generate the mature miRNA. Signaling pathways that control miRNA biogenesis and the mechanisms of regulation, however, are not well understood. In this chapter, we will discuss the finding that signal transducers of the Transforming Growth Factor β (TGFβ) signaling pathway, the Smads, play a critical regulatory role in the nuclear processing of miRNAs by the RNase III-type protein Drosha.
Collapse
|
49
|
Abstract
RNA interference (RNAi) is an effective tool for genome-scale, high-throughput analysis of gene function. In the past five years, a number of genome-scale RNAi high-throughput screens (HTSs) have been done in both Drosophila and mammalian cultured cells to study diverse biological processes, including signal transduction, cancer biology, and host cell responses to infection. Results from these screens have led to the identification of new components of these processes and, importantly, have also provided insights into the complexity of biological systems, forcing new and innovative approaches to understanding functional networks in cells. Here, we review the main findings that have emerged from RNAi HTS and discuss technical issues that remain to be improved, in particular the verification of RNAi results and validation of their biological relevance. Furthermore, we discuss the importance of multiplexed and integrated experimental data analysis pipelines to RNAi HTS.
Collapse
Affiliation(s)
- Stephanie Mohr
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts 02115
| | - Chris Bakal
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
50
|
Mattila J, Puig O. Insights to transcriptional networks by using high throughput RNAi strategies. Brief Funct Genomics 2009; 9:43-52. [PMID: 19952073 DOI: 10.1093/bfgp/elp046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
RNA interference (RNAi) is a powerful method to unravel the role of a given gene in eukaryotic cells. The development of high throughput assay platforms such as fluorescence plate readers and high throughput microscopy has allowed the design of genome wide RNAi screens to systemically discern members of regulatory networks around various cellular processes. Here we summarize the different strategies employed in RNAi screens to reveal regulators of transcriptional networks. We focus our discussion in experimental approaches designed to uncover regulatory interactions modulating transcription factor activity.
Collapse
Affiliation(s)
- Jaakko Mattila
- Merck Research Laboratories, Department of Molecular Profiling-Research Informatics, Rahway, NJ 07065, USA.
| | | |
Collapse
|