1
|
Levenhagen MMMD, Neves SC, Machado NM, Silva BC, Oliveira RGS, Kassuya CAL, Salvador MJ, Oliveira RJ. The ethanolic extract of Gomphrena celosioides is not carcinogenic and has antigenotoxic effects and chemopreventive Properties. BRAZ J BIOL 2024; 84:e283243. [PMID: 39383365 DOI: 10.1590/1519-6984.283243] [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: 02/13/2024] [Accepted: 07/08/2024] [Indexed: 10/11/2024] Open
Abstract
Gomphrena celosioides, popularly known as perpétua, perpétua brava, bachelor´s button and prostate globe amarahth, is used for the treatment of urinary tract disorders, kidney stones, for skin diseases, infectious diseases, gastrointestinal and respiratory conditions. Rich in phenolic acids and flavonoids, this plant has therefore a potential for use in cancer prevention. Given the above, the present research aimed to evaluate the carcinogenic effect of the ethanolic extract of G. celosioides (EEGc) in an alternative model of Drosophila melanogaster and the genotoxic and antigenotoxic effects in Swiss mice. The larval survival test and the detection of epithelial tumor clones were performed in D. melanogaster. The tested EEGc concentrations were 0.96, 1.92, 3.85 and 7.70 mg/mL. In Swiss mice, the genotoxicity and antigenotoxicity of doses of 100, 1,000 and 2,000 mg/Kg were evaluated. The results showed that EEGc at a concentration of 7.70 mg/mL reduced (p<0.05) larval survival. However, EEGc was not carcinogenic, and the lowest concentration (0.96 mg/mL) prevented (p<0.05) the basal occurrence of epithelial tumors. In mice, EEGc at the highest dose (2,000mg/Kg) increased the frequency of genomic lesions (p<0.05). Yet, none of the doses caused chromosomal lesions (p>0.05). When associated with cyclophosphamide, EEGc was antigenotoxic (p<0.05). The percentages of reduction of genomic damage ranged from 33.39 to 63.23% and of chromosomal damage from 20.00 to 77.19%. In view of the above, it is suggested that EEGc is not carcinogenic, has an antigenotoxic effect and chemopreventive properties.
Collapse
Affiliation(s)
- M M M D Levenhagen
- Universidade Federal de Mato Grosso do Sul - UFMS, Centro de Estudos em Células-Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Faculdade de Medicina, Programa de Pós-graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Campo Grande, MS, Brasil
| | - S C Neves
- Universidade Federal de Mato Grosso do Sul - UFMS, Centro de Estudos em Células-Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Faculdade de Medicina, Programa de Pós-graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Campo Grande, MS, Brasil
| | - N M Machado
- Centro Universitário de Patos de Minas - UNIPAM, Laboratório de Citogenética e Mutagênese - LABCIM, Patos de Minas, MG, Brasil
| | - B C Silva
- Centro Universitário de Patos de Minas - UNIPAM, Laboratório de Citogenética e Mutagênese - LABCIM, Patos de Minas, MG, Brasil
| | - R G S Oliveira
- Centro Universitário de Patos de Minas - UNIPAM, Laboratório de Citogenética e Mutagênese - LABCIM, Patos de Minas, MG, Brasil
| | - C A L Kassuya
- Universidade Federal da Grande Dourados - UFGD, Faculdade de Ciências da Saúde, Dourados, MS, Brasil
| | - M J Salvador
- Universidade Estadual de Campinas - UNICAMP, Instituto de Biologia, Departamento de Biologia Vegetal, Campinas, SP, Brasil
| | - R J Oliveira
- Universidade Federal de Mato Grosso do Sul - UFMS, Centro de Estudos em Células-Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Faculdade de Medicina, Programa de Pós-graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Campo Grande, MS, Brasil
| |
Collapse
|
2
|
Nie L, Fei C, Fan Y, Dang F, Zhao Z, Zhu T, Wu X, Dai T, Balasubramanian A, Pan J, Hu Y, Luo HR, Wei W, Chen J. Consecutive palmitoylation and phosphorylation orchestrates NLRP3 membrane trafficking and inflammasome activation. Mol Cell 2024; 84:3336-3353.e7. [PMID: 39173637 DOI: 10.1016/j.molcel.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/16/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024]
Abstract
NLRP3 inflammasome activation, essential for cytokine secretion and pyroptosis in response to diverse stimuli, is closely associated with various diseases. Upon stimulation, NLRP3 undergoes subcellular membrane trafficking and conformational rearrangements, preparing itself for inflammasome assembly at the microtubule-organizing center (MTOC). Here, we elucidate an orchestrated mechanism underlying these ordered processes using human and murine cells. Specifically, NLRP3 undergoes palmitoylation at two sites by palmitoyl transferase zDHHC1, facilitating its trafficking between subcellular membranes, including the mitochondria, trans-Golgi network (TGN), and endosome. This dynamic trafficking culminates in the localization of NLRP3 to the MTOC, where LATS1/2, pre-recruited to MTOC during priming, phosphorylates NLRP3 to further facilitate its interaction with NIMA-related kinase 7 (NEK7), ultimately leading to full NLRP3 activation. Consistently, Zdhhc1-deficiency mitigated LPS-induced inflammation and conferred protection against mortality in mice. Altogether, our findings provide valuable insights into the regulation of NLRP3 membrane trafficking and inflammasome activation, governed by palmitoylation and phosphorylation events.
Collapse
Affiliation(s)
- Li Nie
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China.
| | - Chenjie Fei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Yizeng Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Fabin Dang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyue Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Tingfang Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Xiangyu Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Ting Dai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Arumugam Balasubramanian
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA 02115, USA
| | - Jing Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Yang Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China
| | - Hongbo R Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 811, Boston, MA 02115, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, P.R. China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, P.R. China.
| |
Collapse
|
3
|
Cappucci U, Proietti M, Casale AM, Schiavo S, Chiavarini S, Accardo S, Manzo S, Piacentini L. Assessing genotoxic effects of plastic leachates in Drosophila melanogaster. CHEMOSPHERE 2024; 361:142440. [PMID: 38821133 DOI: 10.1016/j.chemosphere.2024.142440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
Plastic polymers were largely added with chemical substances to be utilized in the items and product manufacturing. The leachability of these substances is a matter of concern given the wide amount of plastic waste, particularly in terrestrial environments, where soil represents a sink for these novel contaminants and a possible pathway of human health risk. In this study, we integrated genetic, molecular, and behavioral approaches to comparatively evaluate toxicological effects of plastic leachates, virgin and oxodegradable polypropylene (PP) and polyethylene (PE), in Drosophila melanogaster, a novel in vivo model organism for environmental monitoring studies and (eco)toxicological research. The results of this study revealed that while conventional toxicological endpoints such as developmental times and longevity remain largely unaffected, exposure to plastic leachates induces chromosomal abnormalities and transposable element (TE) activation in neural tissues. The combined effects of DNA damage and TE mobilization contribute to genome instability and increase the likelihood of LOH events, thus potentiating tumor growth and metastatic behavior ofRasV12 clones. Collectively, these findings indicate that plastic leachates exert genotoxic effects in Drosophila thus highlighting potential risks associated with leachate-related plastic pollution and their implications for ecosystems and human health.
Collapse
Affiliation(s)
- Ugo Cappucci
- Department of Biology and Biotechnologies "C. Darwin", Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy
| | - Mirena Proietti
- Department of Biology and Biotechnologies "C. Darwin", Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy
| | - Assunta Maria Casale
- Department of Biology and Biotechnologies "C. Darwin", Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy
| | - Simona Schiavo
- ENEA, Department for Sustainability, Division Protection and Enhancement of the Natural Capital, P. le E. Fermi 1, 80055 Portici, Na, Italy
| | - Salvatore Chiavarini
- ENEA, Department for Sustainability, Division Protection and Enhancement of the Natural Capital, P. le E. Fermi 1, 80055 Portici, Na, Italy
| | - Sara Accardo
- ENEA, Department for Sustainability, Division Protection and Enhancement of the Natural Capital, P. le E. Fermi 1, 80055 Portici, Na, Italy
| | - Sonia Manzo
- ENEA, Department for Sustainability, Division Protection and Enhancement of the Natural Capital, P. le E. Fermi 1, 80055 Portici, Na, Italy.
| | - Lucia Piacentini
- Department of Biology and Biotechnologies "C. Darwin", Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
4
|
Expression of Key Factors of the Hippo Signaling Pathway in Yak (Bos grunniens) Mammary Gland. Animals (Basel) 2022; 12:ani12162103. [PMID: 36009693 PMCID: PMC9404922 DOI: 10.3390/ani12162103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The Hippo signaling pathway plays a significant role in regulating the organ development processes of mammals. Our research aimed to investigate the expression and distribution of key members of the Hippo signaling pathway in yak mammary glands during different stages. Using immunohistochemistry, Western blot, and relative quantitative real-time polymerase chain reaction techniques, we found that the protein and mRNA expression levels of MST1, LATS1, YAP1 and TEAD1 in the yak’s mammary gland varies with the growth, lactation, and dry periods. The differential expression in the yak’s mammary gland at different stages strongly suggests that the Hippo signaling pathway plays an important role in regulating the mammary gland development processes under different physiological conditions. Abstract Due to its rich nutritional value, yak milk is an important food source in the alpine pastoral areas. However, yaks have a low milk yield. The Hippo pathway participates in cell proliferation and organ development. We aimed to determine the regulatory mechanism of this pathway in yak mammary cells. A greater understanding of how the expression of its essential genes influence the reproductive cycle could lead to improvements in lactation performance. The expression levels of the key genes MST1, LATS1, YAP1, and TEAD1 were detected by quantitative real-time PCR, Western blotting, and immunohistochemistry during the growth, lactation, and dry periods (GP, LP and DP, respectively). The MST1 and LATS1 mRNA and protein expression level was highest during GP and lowest during LP. The YAP1 and TEAD1 mRNA and protein expression level decreased from GP to LP and DP. MST1 and LATS1 were expressed in the cytoplasm whereas YAP1 and TEAD1 were expressed in the nucleus and cytoplasm, respectively. The differential expression of MST1, LATS1, YAP1, and TEAD1 expression in the yak mammary gland during different developmental stages strongly suggests that they play an important role in the regulation of developmental functions under different physiological conditions.
Collapse
|
5
|
Vitamin B6 Deficiency Promotes Loss of Heterozygosity (LOH) at the Drosophila warts (wts) Locus. Int J Mol Sci 2022; 23:ijms23116087. [PMID: 35682766 PMCID: PMC9181336 DOI: 10.3390/ijms23116087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
The active form of vitamin B6, pyridoxal 5'-phosphate (PLP), is a cofactor for more than 200 enzymes involved in many metabolic pathways. Moreover, PLP has antioxidant properties and quenches the reactive oxygen species (ROS). Accordingly, PLP deficiency causes chromosome aberrations in Drosophila, yeast, and human cells. In this work, we investigated whether PLP depletion can also cause loss of heterozygosity (LOH) of the tumor suppressor warts (wts) in Drosophila. LOH is usually initiated by DNA breakage in heterozygous cells for a tumor suppressor mutation and can contribute to oncogenesis inducing the loss of the wild-type allele. LOH at the wts locus results in epithelial wts homozygous tumors easily detectable on adult fly cuticle. Here, we found that PLP depletion, induced by two PLP inhibitors, promotes LOH of wts locus producing significant frequencies of wts tumors (~7% vs. 2.3%). In addition, we identified the mitotic recombination as a possible mechanism through which PLP deficiency induces LOH. Moreover, LOH of wts locus, induced by PLP inhibitors, was rescued by PLP supplementation. These data further confirm the role of PLP in genome integrity maintenance and indicate that vitamin B6 deficiency may impact on cancer also by promoting LOH.
Collapse
|
6
|
Kim CL, Lim SB, Kim K, Jeong HS, Mo JS. Phosphorylation analysis of the Hippo-YAP pathway using Phos-tag. J Proteomics 2022; 261:104582. [DOI: 10.1016/j.jprot.2022.104582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022]
|
7
|
Brandão DC, Lima PMAP, Martins IC, Cordeiro CS, Cordeiro AO, Vecchi L, Guerra JFC, Orsolin PC, Gazolla MC, Costa DS, da Silva Filho AA, Araújo TG. Arrabidaea chica chloroform extract modulates estrogen and androgen receptors on luminal breast cancer cells. BMC Complement Med Ther 2022; 22:18. [PMID: 35057779 PMCID: PMC8773405 DOI: 10.1186/s12906-022-03506-3] [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: 08/17/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast Cancer (BC) is the most common cancer in women worldwide and, although 70% of patients are responsive to selective Estrogen Receptor (ER) modulators such as Tamoxifen (Tam), patients' survival is comprised by resistance to endocrine therapy. Brazilian flora, especially the Amazon biome, is one of the richest global sources of native species with potentially bioactive compounds. Arrabidaea chica is a plant native to the Amazon that has been used in the treatment of different diseases. However, its action on BC remains unclear. METHODS Herein the biological effects of the chloroform extract of A. chica (CEAC) were evaluated on BC cells and in in vivo model. After confirmation of CEAC antioxidant capacity, cells were treated with CEAC and Tam, alone and with CEAC+Tam. The cell viability was evaluated by MTT and hormone receptor transcripts levels were assessed (ESR1, ESR2 and AR). Finally, anticarcinogenicity of CEAC was recorded in Drosophila melanogaster through Epithelial Tumor Test (ETT). RESULTS The study confirmed the antioxidant activity of CEAC. CEAC was selective for MCF-7, downregulating ESR2 and AR transcripts and upregulating ESR2 expression. The modulatory effects of CEAC on ERs did not differ between cells treated with Tam and with CEAC+Tam. Interestingly, previous treatment with CEAC, followed by treatment with Tam promoted a significant decrease in cell viability. The extract also presented anticarcinogenic effect in in vivo assay. CONCLUSION The bioassays on breast tumor cells demonstrated the antiproliferative activity of the extract, which modulated the expression of hormone receptors and sensitized luminal tumor cells to Tam. These results suggest that CEAC could be a complementary treatment for BC.
Collapse
Affiliation(s)
- Douglas C. Brandão
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
| | - Paula M. A. P. Lima
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
- Laboratory of Cytogenetic and Mutagenesis, University Center of Patos de Minas, Patos de Minas, MG Brazil
| | - Isabella C. Martins
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
| | - Carina S. Cordeiro
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
| | - Antonielle O. Cordeiro
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG Brazil
| | - Lara Vecchi
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG Brazil
| | - Joyce F. C. Guerra
- Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas, MG Brazil
| | - Priscila C. Orsolin
- Laboratory of Cytogenetic and Mutagenesis, University Center of Patos de Minas, Patos de Minas, MG Brazil
| | - Matheus C. Gazolla
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG Brazil
| | - Danilo S. Costa
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG Brazil
| | - Ademar A. da Silva Filho
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG Brazil
| | - Thaise G. Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG Brazil
| |
Collapse
|
8
|
Xiao Y, Dong J. The Hippo Signaling Pathway in Cancer: A Cell Cycle Perspective. Cancers (Basel) 2021; 13:cancers13246214. [PMID: 34944834 PMCID: PMC8699626 DOI: 10.3390/cancers13246214] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/25/2023] Open
Abstract
Simple Summary Cancer is increasingly viewed as a cell cycle disease in that the dysregulation of the cell cycle machinery is a common feature in cancer. The Hippo signaling pathway consists of a core kinase cascade as well as extended regulators, which together control organ size and tissue homeostasis. The aberrant expression of cell cycle regulators and/or Hippo pathway components contributes to cancer development, and for this reason, we specifically focus on delineating the roles of the Hippo pathway in the cell cycle. Improving our understanding of the Hippo pathway from a cell cycle perspective could be used as a powerful weapon in the cancer battlefield. Abstract Cell cycle progression is an elaborate process that requires stringent control for normal cellular function. Defects in cell cycle control, however, contribute to genomic instability and have become a characteristic phenomenon in cancers. Over the years, advancement in the understanding of disrupted cell cycle regulation in tumors has led to the development of powerful anti-cancer drugs. Therefore, an in-depth exploration of cell cycle dysregulation in cancers could provide therapeutic avenues for cancer treatment. The Hippo pathway is an evolutionarily conserved regulator network that controls organ size, and its dysregulation is implicated in various types of cancers. Although the role of the Hippo pathway in oncogenesis has been widely investigated, its role in cell cycle regulation has not been comprehensively scrutinized. Here, we specifically focus on delineating the involvement of the Hippo pathway in cell cycle regulation. To that end, we first compare the structural as well as functional conservation of the core Hippo pathway in yeasts, flies, and mammals. Then, we detail the multi-faceted aspects in which the core components of the mammalian Hippo pathway and their regulators affect the cell cycle, particularly with regard to the regulation of E2F activity, the G1 tetraploidy checkpoint, DNA synthesis, DNA damage checkpoint, centrosome dynamics, and mitosis. Finally, we briefly discuss how a collective understanding of cell cycle regulation and the Hippo pathway could be weaponized in combating cancer.
Collapse
Affiliation(s)
| | - Jixin Dong
- Correspondence: ; Tel.: +402-559-5596; Fax: +402-559-4651
| |
Collapse
|
9
|
Meng F, Xie B, Martin JF. Targeting the Hippo pathway in heart repair. Cardiovasc Res 2021; 118:2402-2414. [PMID: 34528077 DOI: 10.1093/cvr/cvab291] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo pathway is an evolutionarily and functionally conserved signaling pathway that controls organ size by regulating cell proliferation, apoptosis, and differentiation. Emerging evidence has shown that the Hippo pathway plays critical roles in cardiac development, homeostasis, disease, and regeneration. Targeting the Hippo pathway has tremendous potential as a therapeutic strategy for treating intractable cardiovascular diseases such as heart failure. In this review, we summarize the function of the Hippo pathway in the heart. Particularly, we highlight the posttranslational modification of Hippo pathway components, including the core kinases LATS1/2 and their downstream effectors YAP/TAZ, in different contexts, which has provided new insights and avenues in cardiac research.
Collapse
Affiliation(s)
- Fansen Meng
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Bing Xie
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030.,Texas Heart Institute, Houston, Texas, 77030
| |
Collapse
|
10
|
Cellular feedback dynamics and multilevel regulation driven by the hippo pathway. Biochem Soc Trans 2021; 49:1515-1527. [PMID: 34374419 PMCID: PMC8421037 DOI: 10.1042/bst20200253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
The Hippo pathway is a dynamic cellular signalling nexus that regulates differentiation and controls cell proliferation and death. If the Hippo pathway is not precisely regulated, the functionality of the upstream kinase module is impaired, which increases nuclear localisation and activity of the central effectors, the transcriptional co-regulators YAP and TAZ. Pathological YAP and TAZ hyperactivity consequently cause cancer, fibrosis and developmental defects. The Hippo pathway controls an array of fundamental cellular processes, including adhesion, migration, mitosis, polarity and secretion of a range of biologically active components. Recent studies highlight that spatio-temporal regulation of Hippo pathway components are central to precisely controlling its context-dependent dynamic activity. Several levels of feedback are integrated into the Hippo pathway, which is further synergized with interactors outside of the pathway that directly regulate specific Hippo pathway components. Likewise, Hippo core kinases also ‘moonlight’ by phosphorylating multiple substrates beyond the Hippo pathway and thereby integrates further flexibility and robustness in the cellular decision-making process. This topic is still in its infancy but promises to reveal new fundamental insights into the cellular regulation of this therapeutically important pathway. We here highlight recent advances emphasising feedback dynamics and multilevel regulation of the Hippo pathway with a focus on mitosis and cell migration, as well as discuss potential productive future research avenues that might reveal novel insights into the overall dynamics of the pathway.
Collapse
|
11
|
Yoon HS, Fujino K, Liu S, Takano T, Tsugama D. NDR/LATS-family protein kinase genes are indispensable for embryogenesis in Arabidopsis. FEBS Open Bio 2021; 11:2600-2606. [PMID: 34320276 PMCID: PMC8409290 DOI: 10.1002/2211-5463.13257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 11/10/2022] Open
Abstract
NDR/LATS‐family protein kinases are conserved among eukaryotes. These protein kinases in yeast and animals phosphorylate specific targets and regulate the cell cycle. Arabidopsis thaliana has eight NDR/LATS‐family protein kinase genes (NDR1‐8), of which NDR2, NDR4, and NDR5 are involved in regulating pollen development. However, the functions of the other NDR/LATS‐family protein kinase genes in plants are unclear. Here, we show that three putative phosphorylation sites of an Arabidopsis basic leucine zipper transcription factor, VIP1, correspond to NDR/LATS‐family protein kinase phosphorylation motifs and that two of these three sites are phosphorylated by NDR2, NDR3, or NDR8 in vitro. Expression of NDR1‐8 was detected in various tissues. An NDR4 NDR6 NDR7 NDR8 quadruple mutation caused embryonic lethality These results suggest that different NDR/LATS‐family protein kinases in plants have distinct physiological roles.
Collapse
Affiliation(s)
- Hyuk Sung Yoon
- Asian Research Center for Bioresource and Environmental Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Nishitokyo-shi, Japan
| | - Kaien Fujino
- Laboratory of Crop Physiology, Research Faculty of Agriculture, Hokkaido University, Sapporo-shi, Japan
| | - Shenkui Liu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Lin'an, China
| | - Tetsuo Takano
- Asian Research Center for Bioresource and Environmental Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Nishitokyo-shi, Japan
| | - Daisuke Tsugama
- Asian Research Center for Bioresource and Environmental Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Nishitokyo-shi, Japan
| |
Collapse
|
12
|
Ruehle MD, Stemm-Wolf AJ, Pearson CG. Sas4 links basal bodies to cell division via Hippo signaling. J Cell Biol 2021; 219:151794. [PMID: 32435796 PMCID: PMC7401811 DOI: 10.1083/jcb.201906183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 03/10/2020] [Accepted: 05/01/2020] [Indexed: 01/07/2023] Open
Abstract
Basal bodies (BBs) are macromolecular complexes required for the formation and cortical positioning of cilia. Both BB assembly and DNA replication are tightly coordinated with the cell cycle to ensure their accurate segregation and propagation to daughter cells, but the mechanisms ensuring coordination are unclear. The Tetrahymena Sas4/CPAP protein is enriched at assembling BBs, localizing to the core BB structure and to the base of BB-appendage microtubules and striated fiber. Sas4 is necessary for BB assembly and cortical microtubule organization, and Sas4 loss disrupts cell division furrow positioning and DNA segregation. The Hippo signaling pathway is known to regulate cell division furrow position, and Hippo molecules localize to BBs and BB-appendages. We find that Sas4 loss disrupts localization of the Hippo activator, Mob1, suggesting that Sas4 mediates Hippo activity by promoting scaffolds for Mob1 localization to the cell cortex. Thus, Sas4 links BBs with an ancient signaling pathway known to promote the accurate and symmetric segregation of the genome.
Collapse
Affiliation(s)
- Marisa D Ruehle
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Alexander J Stemm-Wolf
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
13
|
Masliantsev K, Karayan-Tapon L, Guichet PO. Hippo Signaling Pathway in Gliomas. Cells 2021; 10:184. [PMID: 33477668 PMCID: PMC7831924 DOI: 10.3390/cells10010184] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
The Hippo signaling pathway is a highly conserved pathway involved in tissue development and regeneration that controls organ size through the regulation of cell proliferation and apoptosis. The core Hippo pathway is composed of a block of kinases, MST1/2 (Mammalian STE20-like protein kinase 1/2) and LATS1/2 (Large tumor suppressor 1/2), which inhibits nuclear translocation of YAP/TAZ (Yes-Associated Protein 1/Transcriptional co-activator with PDZ-binding motif) and its downstream association with the TEAD (TEA domain) family of transcription factors. This pathway was recently shown to be involved in tumorigenesis and metastasis in several cancers such as lung, breast, or colorectal cancers but is still poorly investigated in brain tumors. Gliomas are the most common and the most lethal primary brain tumors representing about 80% of malignant central nervous system neoplasms. Despite intensive clinical protocol, the prognosis for patients remains very poor due to systematic relapse and treatment failure. Growing evidence demonstrating the role of Hippo signaling in cancer biology and the lack of efficient treatments for malignant gliomas support the idea that this pathway could represent a potential target paving the way for alternative therapeutics. Based on recent advances in the Hippo pathway deciphering, the main goal of this review is to highlight the role of this pathway in gliomas by a state-of-the-art synthesis.
Collapse
Affiliation(s)
- Konstantin Masliantsev
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| | - Lucie Karayan-Tapon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| | - Pierre-Olivier Guichet
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| |
Collapse
|
14
|
Vasconcelos MA, Orsolin PC, Oliveira VC, Lima PMAP, Naves MPC, de Morais CR, Nicolau-Júnior N, Bonetti AM, Spanó MA. Modulating effect of vitamin D3 on the mutagenicity and carcinogenicity of doxorubicin in Drosophila melanogaster and in silico studies. Food Chem Toxicol 2020; 143:111549. [PMID: 32640329 PMCID: PMC7335493 DOI: 10.1016/j.fct.2020.111549] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/18/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023]
Abstract
Vitamin D3 (VD3) deficiency increases DNA damage, while supplementation may exert a pro-oxidant activity, prevent viral infections and formation of tumors. The aim of this study was to investigate the mutagenicity and carcinogenicity of VD3 alone or in combination with doxorubicin (DXR) using the Somatic Mutation and Recombination Test and the Epithelial Tumor Test, both in Drosophila melanogaster. For better understanding of the molecular interactions of VD3 and receptors, in silico analysis were performed with molecular docking associated with molecular dynamics. Findings revealed that VD3 alone did not increase the frequency of mutant spots, but reduced the frequency of mutant spots when co-administered with DXR. In addition, VD3 did not alter the recombinogenic effect of DXR in both ST and HB crosses. VD3 alone did not increase the total frequency of tumor, but significantly reduced the total frequency of tumor when co-administered with DXR. Molecular modeling and molecular dynamics between calcitriol and Ecdysone Receptor (EcR) showed a stable interaction, indicating the possibility of signal transduction between VD3 and EcR. In conclusion, under these experimental conditions, VD3 has modulatory effects on the mutagenicity and carcinogenicity induced by DXR in somatic cells of D. melanogaster and exhibited satisfactory interactions with the EcR.
Collapse
Affiliation(s)
- Mirley Alves Vasconcelos
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, Uberlândia, MG, Brazil.
| | - Priscila Capelari Orsolin
- Laboratory of Cytogenetic and Mutagenesis, University Center of Patos de Minas, Patos de Minas, MG, Brazil.
| | - Victor Constante Oliveira
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, Uberlândia, MG, Brazil.
| | | | | | | | - Nilson Nicolau-Júnior
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, Uberlândia, MG, Brazil.
| | - Ana Maria Bonetti
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, Uberlândia, MG, Brazil.
| | - Mário Antônio Spanó
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, Uberlândia, MG, Brazil.
| |
Collapse
|
15
|
Luo SY, Kwok HH, Yang PC, Ip MSM, Minna JD, Lam DCL. Expression of large tumour suppressor (LATS) kinases modulates chemotherapy response in advanced non-small cell lung cancer. Transl Lung Cancer Res 2020; 9:294-305. [PMID: 32420069 PMCID: PMC7225163 DOI: 10.21037/tlcr.2020.03.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background The Hippo signalling pathway plays an important role in regulating organ size and cell proliferation. Down-regulation of large tumour suppressor (LATS) protein homologs LATS1 or LATS2 has been found in lung cancer. LATS1 and LATS2 are the core components of the Hippo signalling pathway. LATS1 and LATS2 share some conserved structural features and exhibit redundant biological functions. The aim of this study was to dissect the interaction between these two homologs. Methods In lung adenocarcinoma (AD) cells, protein expression of LATS1 and LATS2 were determined by western blotting; cell viability and apoptosis were measured by MTT and annexin V staining after treatment with cisplatin; subcellular distributions of LATS proteins were determined by immunofluorescence microscopy; LATS2 expression was modulated by shRNA-mediated knockdown or ectopic expression in cancer cell lines. Results Manipulation of the expression of these two LATS kinases influenced cisplatin response in advanced lung AD cell lines. High LATS2-to-LATS1 ratio in H2023 cells was associated with cisplatin resistance, while low LATS2-to-LATS1 ratio in CL1-0 and CL83 cells was associated with sensitivity to cisplatin. Manipulating the LATS2-to-LATS1 ratio by LATS2 over-expression in CL1-0 and CL83 rendered them resistant to cisplatin treatment, whereas LATS2 knockdown in H2023 alleviated the LATS2-to-LATS1 ratio and sensitized cancer cells to cisplatin exposure. Conclusions Our data suggested that the ratio of expression of LATS kinases played a role in the modulation of cisplatin sensitivity in advanced lung AD, and targeting of LATS proteins as a novel therapeutic strategy for lung AD deserves further investigation.
Collapse
Affiliation(s)
- Susan Yang Luo
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| | - Hoi-Hin Kwok
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Mary Sau-Man Ip
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| | - John Dorrance Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David Chi-Leung Lam
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
de Sousa FA, de Morais CR, Vieira JS, Maranho LS, Machado FL, Pereira S, Barbosa LC, Coelho HE, Campos CF, Bonetti AM. Genotoxicity and carcinogenicity of ivermectin and amoxicillin in vivo systems. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 70:103196. [PMID: 31152944 DOI: 10.1016/j.etap.2019.103196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 04/21/2019] [Accepted: 05/21/2019] [Indexed: 06/09/2023]
Abstract
Antiparasitic substances are chemicals used to control or kill endoparasites and ectoparasites. Based on the premise that Ivermectin (IVM) and Amoxicillin (AMX) are commonly considered in parasitic control in mammals, the present study aimed to evaluate the carcinogenic and genotoxic potential of different concentrations of IVM and AMX through the detection of epithelial tumor test in Drosophila melanogaster. Third-instar larvae descending from the cross between wts/TM3, Sb1 females and mwh/mwh males were treated with different concentrations of IVM (2.9, 5.8, 11.6 and 23.2 x 10-17 mM) or AMX (1.37, 2.74, 5.48 and 10.9 x 10-16mM). The results revealed that IVM increased the frequency of epithelial tumor in D. melanogaster considering all evaluated concentrations, while AMX showed no carcinogenic effect. Furthermore, the Micronucleus (MN) test in Tradescantia pallida was used to evaluate the genotoxic effect of IVM and AMX. T. pallida individuals were exposed for 8 hours at different concentrations of IVM (5.71, 11.42, 22.84 and 45.68 x 10-5mM) or AMX (5.13, 10.26, 20.52 and 41.05 x 10-3mM). Findings showed an increase in the frequency of micronuclei in T. pallida treated with 11.42, 22.84 and 45.68 x 10-5mM of IVM. We conclude that chronic exposure to IVM is directly associated with events resulting from genetic instability (genotoxicity and carcinogenicity). On the other hand, AMX was neither carcinogenic nor genotoxic for D. melanogaster and T. pallida.
Collapse
Affiliation(s)
- Francielle Aparecida de Sousa
- Department of Genetics, University Center of Cerrado Patrocínio, Avenida Líria Terezinha Lassi Capuano, 466, 38747-792, Patrocínio, Minas Gerais, Brazil
| | - Cássio Resende de Morais
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil.
| | - Jéssica Soares Vieira
- Department of Cell Biology, Carmelitana Foundation Mário Palmério, 38500-000, Monte Carmelo, Minas Gerais, Brazil
| | - Lavínia Sales Maranho
- Department of Genetics, University Center of Cerrado Patrocínio, Avenida Líria Terezinha Lassi Capuano, 466, 38747-792, Patrocínio, Minas Gerais, Brazil
| | - Francielli Lara Machado
- Department of Genetics, University Center of Cerrado Patrocínio, Avenida Líria Terezinha Lassi Capuano, 466, 38747-792, Patrocínio, Minas Gerais, Brazil
| | - Samanta Pereira
- Department of Genetics, University Center of Cerrado Patrocínio, Avenida Líria Terezinha Lassi Capuano, 466, 38747-792, Patrocínio, Minas Gerais, Brazil
| | - Lilian Cristina Barbosa
- Department of Genetics, University Center of Cerrado Patrocínio, Avenida Líria Terezinha Lassi Capuano, 466, 38747-792, Patrocínio, Minas Gerais, Brazil
| | - Humberto Eustáquio Coelho
- Department of Animal Pathology, University of Uberaba, Avenida Nenê Sabino, 1801 - Bairro Universitário, 38055-500, Uberaba, Minas Gerais, Brazil
| | - Carlos Fernando Campos
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil
| | - Ana Maria Bonetti
- Institute of Biotechnology, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
17
|
Yeung YT, Guerrero-Castilla A, Cano M, Muñoz MF, Ayala A, Argüelles S. Dysregulation of the Hippo pathway signaling in aging and cancer. Pharmacol Res 2019; 143:151-165. [PMID: 30910741 DOI: 10.1016/j.phrs.2019.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/04/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
Human beings are facing emerging degenerative and cancer diseases, in large part, as a consequence of increased life expectancy. In the near future, researchers will have to put even more effort into fighting these new challenges, one of which will be prevention of cancer while continuing to improve the aging process through this increased life expectancy. In the last few decades, relevance of the Hippo pathway on cancer has become an important study since it is a major regulator of organ size control and proliferation. However, its deregulation can induce tumors throughout the body by regulating cell proliferation, disrupting cell polarity, releasing YAP and TAZ from the Scribble complexes and facilitating survival gene expression via activation of TEAD transcription factors. This pathway is also involved in some of the most important mechanisms that control the aging processes, such as the AMP-activated protein kinase and sirtuin pathways, along with autophagy and oxidative stress response/antioxidant defense. This could be the link between two tightly connected processes that could open a broader range of targeted molecular therapies to fight aging and cancer. Therefore, available knowledge of the processes involved in the Hippo pathway during aging and cancer must necessarily be well understood.
Collapse
Affiliation(s)
- Yiu To Yeung
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | | | - Mercedes Cano
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mario F Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Antonio Ayala
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
18
|
Kwon L, Magee EM, Crayton A, Goss JW. Fission yeast type 2 node proteins Blt1p and Gef2p cooperate to ensure timely completion of cytokinesis. BMC Mol Cell Biol 2019; 20:1. [PMID: 31041892 PMCID: PMC6446504 DOI: 10.1186/s12860-018-0182-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 12/14/2018] [Indexed: 12/01/2022] Open
Abstract
Background The conserved NDR-family kinase Sid2p localizes to the contractile ring during fission yeast cytokinesis to promote ring constriction, septation, and completion of cell division. Previous studies have found that the Type 2 interphase node proteins Blt1p and Gef2p contribute to localization of Sid2p and its regulatory protein Mob1p at the division site. However, their relative contributions and whether they operate in the same or parallel pathways has been unclear. In this study, we quantify the respective roles of Blt1p and Gef2p in Sid2p/Mob1p recruitment and characterize the effect of single and double deletion mutants on contractile ring dynamics and completion of cell division. Results Using quantitative confocal fluorescence microscopy, we measured Sid2p and Mob1p recruitment to the division site in blt1∆, gef2∆, and blt1∆/gef2∆ mutant cells. We observed an equivalent decrease in Sid2p/Mob1p localization for both single and double mutants. Though assembly of the contractile ring is normal in these mutants, the reduction in Sid2p/Mob1p at the division site delayed the onset of contractile ring constriction and completion of division. We quantified localization of Blt1p and Gef2p at the medial cortex throughout the cell cycle and found that Blt1p localization to interphase nodes and the contractile ring is independent of Gef2p. However, Gef2p localization to the contractile ring is decreased in blt1∆ mutants. Conclusions Blt1p and Gef2p work in the same pathway, rather than in parallel, to localize the NDR-family kinase Sid2p and its regulatory partner Mob1p to the division site, thereby promoting timely completion of cell division. Future studies are necessary to understand how additional fission yeast cytokinesis proteins work with these Type 2 interphase node components to promote Sid2p/Mob1p recruitment. Electronic supplementary material The online version of this article (10.1186/s12860-018-0182-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lois Kwon
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA, 02481, USA
| | - Emma M Magee
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA, 02481, USA
| | - Alexis Crayton
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA, 02481, USA
| | - John W Goss
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA, 02481, USA.
| |
Collapse
|
19
|
Kong F, Ran W, Jiang N, Li S, Zhang D, Sun D. Identification and characterization of differentially expressed miRNAs in HepG2 cells under normoxic and hypoxic conditions. RSC Adv 2019; 9:16884-16891. [PMID: 35516357 PMCID: PMC9064406 DOI: 10.1039/c9ra01523j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are important post-transcriptional regulators involved in hypoxia conditions; however, their roles in HepG2 cells remain poorly understood. Our previous study showed that hypoxia treatment modulated gene expression accompanied by with HepG2 cell proliferation arrest and increased cell death. To better understand the mechanism of phenotypic changes of HepG2 under hypoxia conditions; we conducted a comparative RNA sequencing to identify differentially expressed miRNAs between hypoxia treatment and control cells. In total, 165 differentially expressed miRNAs were identified, among which the expression of 114 miRNAs were up-regulated and that of 51 miRNAs were down-regulated in hypoxia treated HepG2 cells. Expression profiles of eleven randomly selected miRNAs were validated by qRT-PCR. Furthermore, 19 367 annotated target genes of differentially expressed miRNAs were predicted by bioinformatics tools. The Gene Ontology analysis indicated that the molecular function of target genes was primarily related to binding and catalytic activity, and that the Kyoto Encyclopedia of Genes and Genomes annotation for target genes were further classified into pathways involved in cellular processes, metabolism, organismal systems, genetic information processing, human disease and environmental information processing. Among the environmental information processing, certain pathways associated with cell proliferation and apoptosis, such as the hippo signalling pathway, wnt signalling pathway, MAPK signalling pathway and Jak-STAT signaling pathways, represented potential factors in the response to hypoxia treatment. In conclusion, the expression profiles of miRNA in HepG2 cells were significantly altered under hypoxia conditions; which were closely related to cell proliferation arrest and apoptosis. Our findings expand our understanding of miRNAs function in regulating cell fate under hypoxia conditions. MicroRNAs (miRNAs) are important post-transcriptional regulators involved in hypoxia conditions; however, their roles in HepG2 cells remain poorly understood.![]()
Collapse
Affiliation(s)
- Fanzhi Kong
- College of Animal Science and Veterinary Medicine
- Heilongjiang Bayi Agricultural University
- Daqing 163319
- P. R. China
| | - Wei Ran
- College of Animal Science and Veterinary Medicine
- Heilongjiang Bayi Agricultural University
- Daqing 163319
- P. R. China
| | - Ning Jiang
- College of Animal Science and Veterinary Medicine
- Heilongjiang Bayi Agricultural University
- Daqing 163319
- P. R. China
| | - Shize Li
- College of Animal Science and Veterinary Medicine
- Heilongjiang Bayi Agricultural University
- Daqing 163319
- P. R. China
| | - Dongjie Zhang
- College of Food Science
- Heilongjiang Bayi Agricultural University
- Daqing 163319
- P. R. China
| | - Dongbo Sun
- College of Animal Science and Veterinary Medicine
- Heilongjiang Bayi Agricultural University
- Daqing 163319
- P. R. China
| |
Collapse
|
20
|
Carvalho Naves MP, de Morais CR, Silva ACA, Dantas NO, Spanó MA, de Rezende AAA. Assessment of mutagenic, recombinogenic and carcinogenic potential of titanium dioxide nanocristals in somatic cells of Drosophila melanogaster. Food Chem Toxicol 2018; 112:273-281. [DOI: 10.1016/j.fct.2017.12.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 01/02/2023]
|
21
|
Oliveira VC, Constante SAR, Polloni L, Orsolin PC, Silva-Oliveira RG, Machado NM, de Oliveira-Júnior RJ, Nepomuceno JC. Protective effect of aspirin against mitomycin C-induced carcinogenicity, assessed by the test for detection of epithelial tumor clones (warts) in Drosophila melanogaster. Drug Chem Toxicol 2017; 41:330-337. [PMID: 29281929 DOI: 10.1080/01480545.2017.1415926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The present study assessed the protective effect of aspirin against carcinogenicity induced by mitomycin C (MMC) by the test for detection of warts/epithelial tumor clones in Drosophila melanogaster. Larvae were treated with different concentrations of aspirin alone (10, 20 or 40 mg/mL) or aspirin in association with MMC. MMC and ultrapure water were employed as the positive and negative control, respectively. Antioxidant activity was determined using the DPPH method. For performing cytotoxicity assay on HeLa cells, the aspirin concentrations used ranged from 200 mmol/L to 3,125 mmol/L. For assessment of apoptosis and necrosis, cells were incubated for 24 h with complete medium in the absence (control group) or presence of aspirin (12.5 mmol/L and 25 mmol/L). The results obtained in the assessment of the possible carcinogenic effects of aspirin at the three concentrations tested indicate no statistically significant increase in tumor frequency compared to the negative control. The anticarcinogenic activity assessment, where the larvae of D. melanogaster were previously induced to tumor formation by MMC and later treated with aspirin, showed a statistically significant reduction in the number of tumors compared to the positive control. Antioxidant activity across the three aspirin concentrations (10, 20 or 40 mg/mL) ranged from 20.81% to 26.5%. It was observed that aspirin reduced growth viability of HeLa cells in a concentration-dependent manner in comparison with the control. These results indicate that aspirin did not induce tumors in Drosophila and reduced MMC-induced carcinogenicity. The antioxidant activity and apoptosis induction appear to be the main mechanisms involved in reducing the frequency of tumors.
Collapse
Affiliation(s)
- Victor Constante Oliveira
- a Genetics and Biochemistry Institute , Federal University of Uberlandia , Uberlandia , Brazil.,b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| | | | - Lorena Polloni
- c Laboratory of Animal Cytogenetics , Genetics and Biochemistry Institute, Federal University of Uberlândia , Uberlandia , Brazil
| | - Priscila Capelari Orsolin
- b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| | | | - Nayane Moreira Machado
- b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| | - Robson José de Oliveira-Júnior
- c Laboratory of Animal Cytogenetics , Genetics and Biochemistry Institute, Federal University of Uberlândia , Uberlandia , Brazil
| | - Júlio César Nepomuceno
- a Genetics and Biochemistry Institute , Federal University of Uberlandia , Uberlandia , Brazil.,b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| |
Collapse
|
22
|
de Morais CR, Carvalho SM, Carvalho Naves MP, Araujo G, de Rezende AAA, Bonetti AM, Spanó MA. Mutagenic, recombinogenic and carcinogenic potential of thiamethoxam insecticide and formulated product in somatic cells of Drosophila melanogaster. CHEMOSPHERE 2017; 187:163-172. [PMID: 28846972 DOI: 10.1016/j.chemosphere.2017.08.108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/18/2017] [Accepted: 08/19/2017] [Indexed: 06/07/2023]
Abstract
Thiamethoxam (TMX) belongs to a class of neuro-active insecticides referred as neonicotinoids, while actara® (AC) is one of the most popular TMX-based products in Brazil. The aim of this study was to evaluate the mutagenic, recombinogenic and carcinogenic potential of TMX and AC insecticides. The mutagenic and recombinogenic effect of TMX and AC were evaluated in vivo by the Somatic Mutation and Recombination Test (SMART) while carcinogenic effects were evaluated through the Test for Detection of Epithelial Tumor Clones (wts test), both in somatic cells of Drosophila melanogaster. In the SMART, third instar larvae from standard (ST) and high bioactivation (HB) crosses were treated with different concentrations of TMX and AC (2.4; 4.8; 9.7 × 10-4 mM and 1.9 × 10-3 mM). The results revealed mutagenic effects at the highest concentrations tested in the HB cross. In the test for the detection of epithelial tumor, third instar larvae resulting from the cross between wts/TM3, Sb1 virgin females and mwh/mwh males were treated with the same concentrations of TMX and AC used in the SMART. No carcinogenic effect was observed at any of the concentrations tested. In this work, the inhibition of the mechanism of repair by homologous recombination was observed in flies exposed to 9.7 × 10-4 and 1.9 × 10-3 mM of AC. In conclusion, TMX and AC demonstrated to be a promutagen in the highest concentrations tested.
Collapse
Affiliation(s)
- Cássio Resende de Morais
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil
| | - Stephan Malfitano Carvalho
- Department of Entomology, Federal University of Lavras, PO Box 3037, 37200-000, Lavras, Minas Gerais, Brazil
| | - Maria Paula Carvalho Naves
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil
| | - Galber Araujo
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil; Department of Molecular Biology, University of Salzburg, 5020, Salzburg, Austria
| | - Alexandre Azenha Alves de Rezende
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil
| | - Ana Maria Bonetti
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil
| | - Mário Antônio Spanó
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38900-402, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
23
|
Abstract
Proper cellular functionality and homeostasis are maintained by the convergent integration of various signaling cascades, which enable cells to respond to internal and external changes. The Dbf2-related kinases LATS1 and LATS2 (LATS) have emerged as central regulators of cell fate, by modulating the functions of numerous oncogenic or tumor suppressive effectors, including the canonical Hippo effectors YAP/TAZ, the Aurora mitotic kinase family, estrogen signaling and the tumor suppressive transcription factor p53. While the basic functions of the LATS kinase module are strongly conserved over evolution, the genomic duplication event leading to the emergence of two closely related kinases in higher organisms has increased the complexity of this signaling network. Here, we review the LATS1 and LATS2 intrinsic features as well as their reported cellular activities, emphasizing unique characteristics of each kinase. While differential activities between the two paralogous kinases have been reported, many converge to similar pathways and outcomes. Interestingly, the regulatory networks controlling the mRNA expression pattern of LATS1 and LATS2 differ strongly, and may contribute to the differences in protein binding partners of each kinase and in the subcellular locations in which each kinase exerts its functions.
Collapse
Affiliation(s)
- Noa Furth
- Department of Molecular Cell Biology, The Weizmann Institute of Science, POB 26, 234 Herzl St., Rehovot 7610001, Israel
| | - Yael Aylon
- Department of Molecular Cell Biology, The Weizmann Institute of Science, POB 26, 234 Herzl St., Rehovot 7610001, Israel
| |
Collapse
|
24
|
Saturnino RS, Machado NM, Lopes JC, Nepomuceno JC. Assessment of the mutagenic, recombinogenic, and carcinogenic potential of amphotericin B in somatic cells of Drosophila melanogaster. Drug Chem Toxicol 2017; 41:9-15. [PMID: 28274136 DOI: 10.1080/01480545.2016.1188302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Amphotericin B (AmB) is an antifungal antibiotic extracted from Streptomyces nodosus. Its fungicidal activity depends primarily on its binding to the sterol group that is present in fungal membranes. In view of the toxicity of this drug, the purpose of this study was to evaluate its mutagenic, carcinogenic, and recombinogenic activity, based on the wing somatic mutation and recombination test (SMART) and the epithelial tumor detection test (wts) applied to Drosophila melanogaster. Larvae were chronically treated with different concentrations of AmB (0.01, 0.02, and 0.04 mg/mL). The results revealed that AmB is a promutagen exhibiting increase in the number of spots on individuals from high bioactivation (HB) cross with a high level of cytochrome P450. The results also indicate that the main genotoxic event induced by AmB is recombinogenicity. Homologous recombination can act as a determinant at different stages of carcinogenesis. For verification of carcinogenic potential of this compound, larvae from the wts/mwh and wts/ORR, flr3 were treated with the same three AmB concentrations used in the SMART assay. The results did not provide evidence that AmB has carcinogenic potential in wts/mwh individuals. However, individuals from wts/ORR, flr3 developed tumors at the highest concentration tested.
Collapse
Affiliation(s)
- Rosiane Soares Saturnino
- a Institute of Genetics and Biochemistry, Federal University of Uberlândia , Uberlândia , Brazil and.,b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| | - Nayane Moreira Machado
- a Institute of Genetics and Biochemistry, Federal University of Uberlândia , Uberlândia , Brazil and.,b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| | - Jeyson Cesary Lopes
- a Institute of Genetics and Biochemistry, Federal University of Uberlândia , Uberlândia , Brazil and.,b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| | - Júlio César Nepomuceno
- a Institute of Genetics and Biochemistry, Federal University of Uberlândia , Uberlândia , Brazil and.,b Laboratory of Cytogenetics and Mutagenesis , University Center of Patos de Minas , Patos de Minas , Brazil
| |
Collapse
|
25
|
de Morais CR, Bonetti AM, Carvalho SM, de Rezende AAA, Araujo GR, Spanó MA. Assessment of the mutagenic, recombinogenic and carcinogenic potential of fipronil insecticide in somatic cells of Drosophila melanogaster. CHEMOSPHERE 2016; 165:342-351. [PMID: 27664524 DOI: 10.1016/j.chemosphere.2016.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/03/2016] [Accepted: 09/06/2016] [Indexed: 06/06/2023]
Abstract
Fipronil (FP) is an insecticide that belongs to the phenylpyrazole chemical family and is used to control pests by blocking GABA receptor at the entrance channel of the chlorine neurons. The aim of this study was to evaluate the mutagenic, recombinogenic and carcinogenic potential of FP. The mutagenic and recombinogenic effects were evaluated using the somatic mutation and recombination test (SMART) on wing cells of Drosophila melanogaster. Third instar larvae from standard (ST) and high bioactivation (HB) crosses were treated with different concentrations of FP (0.3, 0.7, 1.5 or 3.0 × 10-5 mM). The results showed mutagenic effects at all concentrations tested in the HB cross; and all concentrations tested in the ST cross, except at concentration of 0.7 × 10-5 mM. The carcinogenic effect of FP was assayed through the test for detection of epithelial tumor (warts) in D. melanogaster. Third instar larvae from wts/TM3 virgin females mated to mwh/mwh males were treated with different concentrations of FP (0.3, 0.7, 1.5 or 3.0 × 10-5 mM). All these concentrations induced a statistically significant increase in tumor frequency. In conclusion, FP proved to be mutagenic, recombinogenic and carcinogenic in somatic cells of D. melanogaster.
Collapse
Affiliation(s)
- Cássio Resende de Morais
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38400-902, Uberlândia, Minas Gerais, Brazil
| | - Ana Maria Bonetti
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38400-902, Uberlândia, Minas Gerais, Brazil
| | - Stephan Malfitano Carvalho
- Departament of Entomology, Federal University of Lavras, PO Box 3037, 37.200-000, Lavras, Minas Gerais, Brazil
| | - Alexandre Azenha Alves de Rezende
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38400-902, Uberlândia, Minas Gerais, Brazil
| | - Galber Rodrigues Araujo
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38400-902, Uberlândia, Minas Gerais, Brazil
| | - Mário Antônio Spanó
- Institute of Genetics and Biochemistry, Federal University of Uberlândia, Campus Umuarama, 38400-902, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
26
|
Abstract
Centrosome amplification is a common feature of both solid and hematological human malignancies. Extra centrosomes are not merely innocent bystanders in cancer cells, but rather promote tumor progression by disrupting normal cellular architecture and generating chromosome instability. Consequently, centrosome amplification correlates with advanced tumor grade and overall poor clinical prognosis. By contrast, extra centrosomes are adversely tolerated in non-transformed cells and hinder cell proliferation. This suggests that in addition to acquiring extra centrosomes, cancer cells must also adapt to overcome the deleterious consequences associated with them. Here, we review evidence that implicates core components of the Hippo tumor suppressor pathway as having key roles in both the direct and indirect regulation of centrosome number. Intriguingly, functional inactivation of the Hippo pathway, which is common across broad spectrum of human cancers, likely represents one key adaptation that enables cancer cells to tolerate extra centrosomes.
Collapse
|
27
|
Fallahi E, O'Driscoll NA, Matallanas D. The MST/Hippo Pathway and Cell Death: A Non-Canonical Affair. Genes (Basel) 2016; 7:genes7060028. [PMID: 27322327 PMCID: PMC4929427 DOI: 10.3390/genes7060028] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 01/06/2023] Open
Abstract
The MST/Hippo signalling pathway was first described over a decade ago in Drosophila melanogaster and the core of the pathway is evolutionary conserved in mammals. The mammalian MST/Hippo pathway regulates organ size, cell proliferation and cell death. In addition, it has been shown to play a central role in the regulation of cellular homeostasis and it is commonly deregulated in human tumours. The delineation of the canonical pathway resembles the behaviour of the Hippo pathway in the fly where the activation of the core kinases of the pathway prevents the proliferative signal mediated by the key effector of the pathway YAP. Nevertheless, several lines of evidence support the idea that the mammalian MST/Hippo pathway has acquired new features during evolution, including different regulators and effectors, crosstalk with other essential signalling pathways involved in cellular homeostasis and the ability to actively trigger cell death. Here we describe the current knowledge of the mechanisms that mediate MST/Hippo dependent cell death, especially apoptosis. We include evidence for the existence of complex signalling networks where the core proteins of the pathway play a central role in controlling the balance between survival and cell death. Finally, we discuss the possible involvement of these signalling networks in several human diseases such as cancer, diabetes and neurodegenerative disorders.
Collapse
Affiliation(s)
- Emma Fallahi
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland. emma.fallahi---
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland. emma.fallahi---
| | - Niamh A O'Driscoll
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
28
|
Okamoto A, Yabuta N, Mukai S, Torigata K, Nojima H. Phosphorylation of CHO1 by Lats1/2 regulates the centrosomal activation of LIMK1 during cytokinesis. Cell Cycle 2016; 14:1568-82. [PMID: 25786116 DOI: 10.1080/15384101.2015.1026489] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Large tumor suppressor 1 and 2 (Lats1/2) regulate centrosomal integrity, chromosome segregation and cytokinesis. As components of the centralspindlin complex, the kinesin-like protein CHO1 and its splicing variant MKLP1 colocalize with chromosome passenger proteins and GTPases and regulate the formation of the contractile ring and cytokinesis; however, the regulatory mechanisms of CHO1/MKLP1 remain elusive. Here, we show that Lats1/2 phosphorylate Ser716 in the F-actin-interacting region of CHO1, which is absent in MKLP1. Phosphorylated CHO1 localized to the centrosomes and midbody, and the actin polymerization factor LIM-kinase 1 (LIMK1) was identified as its binding partner. Overexpression of constitutively phosphorylated and non-phosphorylated CHO1 altered the mitotic localization and activation of LIMK1 at the centrosomes in HeLa cells, leading to the inhibition of cytokinesis through excessive phosphorylation of Cofilin and mislocalization of Ect2. These results suggest that Lats1/2 stringently control cytokinesis by regulating CHO1 phosphorylation and the mitotic activation of LIMK1 on centrosomes.
Collapse
Affiliation(s)
- Ayumi Okamoto
- a Department of Molecular Genetics ; Research Institute for Microbial Diseases; Osaka University ; Suita City , Osaka , Japan
| | | | | | | | | |
Collapse
|
29
|
Lats1 suppresses centrosome overduplication by modulating the stability of Cdc25B. Sci Rep 2015; 5:16173. [PMID: 26530630 PMCID: PMC4632022 DOI: 10.1038/srep16173] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 10/06/2015] [Indexed: 12/11/2022] Open
Abstract
Numerical aberration of the centrosome results in chromosome missegregation, eventually leading to chromosomal instability, a hallmark of human tumor malignancy. Large tumor suppressors 1 and 2 (Lats1 and Lats2) are central kinases in the Hippo pathway and regulate development and tumorigenesis by coordinating the balance between cell proliferation and apoptosis. Importantly, Lats1 and Lats2 also play pivotal roles in cell cycle checkpoint and mitosis. The Lats proteins localize at centrosomes, but their centrosomal functions remain elusive. Here, we generated Lats1-null knockout (Lats1−/−) mice and established Lats1-null mouse embryonic fibroblasts (MEFs). In Lats1−/− MEFs, centrosomes were markedly overduplicated, leading to severe mitotic defects such as chromosome missegregation and cytokinesis failure. We also found that Lats1 physically interacts with Cdc25B phosphatase that localizes both at the centrosome and in the nucleus and regulates the linkage between the centrosome cycle and mitotic progression. Although Lats1 did not phosphorylate Cdc25B, loss of Lats1 in MEFs caused abnormal accumulation of Cdc25B protein and hyperactivation of Cdk2 toward nucleophosmin (NPM/B23), one of the licensing factors involved in centriole duplication. Taken together, these data suggest that Lats1 regulates Cdc25B protein level and subsequent Cdk2 activity, thereby suppressing centrosome overduplication during interphase.
Collapse
|
30
|
Kawano S, Maruyama J, Nagashima S, Inami K, Qiu W, Iwasa H, Nakagawa K, Ishigami-Yuasa M, Kagechika H, Nishina H, Hata Y. A cell-based screening for TAZ activators identifies ethacridine, a widely used antiseptic and abortifacient, as a compound that promotes dephosphorylation of TAZ and inhibits adipogenesis in C3H10T1/2 cells. J Biochem 2015; 158:413-23. [PMID: 25979969 DOI: 10.1093/jb/mvv051] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 04/27/2015] [Indexed: 12/17/2022] Open
Abstract
Transcriptional co-activator with PSD-95/Dlg-A/ZO-1 (PDZ)-binding motif (TAZ) regulates in cell proliferation and differentiation. In mesenchymal stem cells it promotes osteogenesis and myogenesis, and suppresses adipogenesis. TAZ activators are expected to prevent osteoporosis, obesity and muscle atrophy. TAZ activation induces epithelial-mesenchymal transition, confers stemness to cancer cells and leads to poor clinical prognosis in cancer patients. In this point of view, TAZ inhibitors should contribute to cancer therapy. Thus, TAZ attracts attention as a two-faced drug target. We screened for TAZ modulators by using human lung cancer A549 cells expressing the fluorescent reporter. Through this assay, we obtained TAZ activator candidates. We unexpectedly found that ethacridine, a widely used antiseptic and abortifacient, enhances the interaction of TAZ and protein phosphatases and increases unphosphorylated and nuclear TAZ. Ethacridine inhibits adipogenesis in mesenchymal C3H10T1/2 cells through the activation of TAZ. This finding suggests that ethacridine is a bona fide TAZ activator and supports that our assay is useful to discover TAZ activators.
Collapse
Affiliation(s)
- Shodai Kawano
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Junichi Maruyama
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Shunta Nagashima
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Kazutoshi Inami
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Wenzhe Qiu
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Hiroaki Iwasa
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Kentaro Nakagawa
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | | | - Hiroyuki Kagechika
- Chemical Biology Screening Center, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; and
| | - Yutaka Hata
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| |
Collapse
|
31
|
Saadeldin MK, Shawer H, Mostafa A, Kassem NM, Amleh A, Siam R. New genetic variants of LATS1 detected in urinary bladder and colon cancer. Front Genet 2015; 5:425. [PMID: 25628642 PMCID: PMC4292772 DOI: 10.3389/fgene.2014.00425] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/19/2014] [Indexed: 11/13/2022] Open
Abstract
LATS1, the large tumor suppressor 1 gene, encodes for a serine/threonine kinase protein and is implicated in cell cycle progression. LATS1 is down-regulated in various human cancers, such as breast cancer, and astrocytoma. Point mutations in LATS1 were reported in human sarcomas. Additionally, loss of heterozygosity of LATS1 chromosomal region predisposes to breast, ovarian, and cervical tumors. In the current study, we investigated LATS1 genetic variations including single nucleotide polymorphisms (SNPs), in 28 Egyptian patients with either urinary bladder or colon cancers. The LATS1 gene was amplified and sequenced and the expression of LATS1 at the RNA level was assessed in 12 urinary bladder cancer samples. We report, the identification of a total of 29 variants including previously identified SNPs within LATS1 coding and non-coding sequences. A total of 18 variants were novel. Majority of the novel variants, 13, were mapped to intronic sequences and un-translated regions of the gene. Four of the five novel variants located in the coding region of the gene, represented missense mutations within the serine/threonine kinase catalytic domain. Interestingly, LATS1 RNA steady state levels was lost in urinary bladder cancerous tissue harboring four specific SNPs (16045 + 41736 + 34614 + 56177) positioned in the 5'UTR, intron 6, and two silent mutations within exon 4 and exon 8, respectively. This study identifies novel single-base-sequence alterations in the LATS1 gene. These newly identified variants could potentially be used as novel diagnostic or prognostic tools in cancer.
Collapse
Affiliation(s)
- Mona K Saadeldin
- Biotechnology Department, American University in Cairo New Cairo, Egypt
| | - Heba Shawer
- Biotechnology Department, American University in Cairo New Cairo, Egypt
| | - Ahmed Mostafa
- National Cancer Institute, Cairo University New Cairo, Egypt
| | - Neemat M Kassem
- Clinical Pathology Department, Cairo University New Cairo, Egypt
| | - Asma Amleh
- Biotechnology Department, American University in Cairo New Cairo, Egypt ; Biology Department, American University in Cairo New Cairo, Egypt
| | - Rania Siam
- Biotechnology Department, American University in Cairo New Cairo, Egypt ; Biology Department, American University in Cairo New Cairo, Egypt ; YJ-Science and Technology Research Center, American University in Cairo New Cairo, Egypt
| |
Collapse
|
32
|
Chen BR, Li Y, Eisenstatt JR, Runge KW. Identification of a lifespan extending mutation in the Schizosaccharomyces pombe cyclin gene clg1+ by direct selection of long-lived mutants. PLoS One 2013; 8:e69084. [PMID: 23874875 PMCID: PMC3711543 DOI: 10.1371/journal.pone.0069084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 06/12/2013] [Indexed: 11/30/2022] Open
Abstract
Model organisms such as budding yeast, worms and flies have proven instrumental
in the discovery of genetic determinants of aging, and the fission yeast
Schizosaccharomyces
pombe is a promising new system for these
studies. We devised an approach to directly select for long-lived
S.
pombe mutants from a random DNA insertion
library. Each insertion mutation bears a unique sequence tag called a bar code
that allows one to determine the proportion of an individual mutant in a culture
containing thousands of different mutants. Aging these mutants in culture
allowed identification of a long-lived mutant bearing an insertion mutation in
the cyclin gene clg1+. Clg1p, like
Pas1p, physically associates with the cyclin-dependent kinase Pef1p. We
identified a third Pef1p cyclin, Psl1p, and found that only loss of Clg1p or
Pef1p extended lifespan. Genetic and co-immunoprecipitation results indicate
that Pef1p controls lifespan through the downstream protein kinase Cek1p. While
Pef1p is conserved as Pho85p in Saccharomyces
cerevisiae, and as cdk5 in humans, genome-wide
searches for lifespan regulators in S. cerevisiae have
never identified Pho85p. Thus, the S. pombe system
can be used to identify novel, evolutionarily conserved lifespan extending
mutations, and our results suggest a potential role for mammalian cdk5 as a
lifespan regulator.
Collapse
Affiliation(s)
- Bo-Ruei Chen
- Department of Molecular Genetics, Cleveland Clinic Lerner College of
Medicine at Case Western Reserve University, Cleveland, Ohio,
USA
- Department of Genetics and Genome Sciences, Case Western Reserve
University School of Medicine, Cleveland, Ohio, United States of
America
- Department of Biochemistry, Case Western Reserve University School of
Medicine, Cleveland, Ohio, United States of America
| | - Yanhui Li
- Department of Molecular Genetics, Cleveland Clinic Lerner College of
Medicine at Case Western Reserve University, Cleveland, Ohio,
USA
- Department of Genetics and Genome Sciences, Case Western Reserve
University School of Medicine, Cleveland, Ohio, United States of
America
| | - Jessica R. Eisenstatt
- Department of Molecular Genetics, Cleveland Clinic Lerner College of
Medicine at Case Western Reserve University, Cleveland, Ohio,
USA
- Department of Biochemistry, Case Western Reserve University School of
Medicine, Cleveland, Ohio, United States of America
| | - Kurt W. Runge
- Department of Molecular Genetics, Cleveland Clinic Lerner College of
Medicine at Case Western Reserve University, Cleveland, Ohio,
USA
- Department of Genetics and Genome Sciences, Case Western Reserve
University School of Medicine, Cleveland, Ohio, United States of
America
- * E-mail:
| |
Collapse
|
33
|
Yabuta N, Mukai S, Okamoto A, Okuzaki D, Suzuki H, Torigata K, Yoshida K, Okada N, Miura D, Ito A, Ikawa M, Okabe M, Nojima H. N-terminal truncation of Lats1 causes abnormal cell growth control and chromosomal instability. J Cell Sci 2012; 126:508-20. [PMID: 23230145 DOI: 10.1242/jcs.113431] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The tumor suppressors Lats1 and Lats2 are mediators of the Hippo pathway that regulates tissue growth and proliferation. Their N-terminal non-kinase regions are distinct except for Lats conserved domains 1 and 2 (LCD1 and LCD2), which may be important for Lats1/2-specific functions. Lats1 knockout mice were generated by disrupting the N-terminal region containing LCD1 (Lats1(ΔN/ΔN)). Some Lats1(ΔN/ΔN) mice were born safely and grew normally. However, mouse embryonic fibroblasts (MEFs) from Lats1(ΔN/ΔN) mice displayed mitotic defects, centrosomal overduplication, chromosomal misalignment, multipolar spindle formation, chromosomal bridging and cytokinesis failure. They also showed anchorage-independent growth and continued cell cycles and cell growth, bypassing cell-cell contact inhibition similar to tumor cells. Lats1(ΔN/ΔN) MEFs produced tumors in nude mice after subcutaneous injection, although the tumor growth rate was much slower than that of ordinary cancer cells. Yap, a key transcriptional coactivator of the Hippo pathway, was overexpressed and stably retained in Lats1(ΔN/ΔN) MEFs in a cell density independent manner, and Lats2 mRNA expression was downregulated. In conclusion, N-terminally truncated Lats1 induced Lats2 downregulation and Yap protein accumulation, leading to chromosomal instability and tumorigenesis.
Collapse
Affiliation(s)
- Norikazu Yabuta
- Department of Molecular Genetics, Osaka University, 3-1 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Orsolin PC, Silva-Oliveira RG, Nepomuceno JC. Assessment of the mutagenic, recombinagenic and carcinogenic potential of orlistat in somatic cells of Drosophila melanogaster. Food Chem Toxicol 2012; 50:2598-604. [PMID: 22621838 DOI: 10.1016/j.fct.2012.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 05/02/2012] [Accepted: 05/04/2012] [Indexed: 11/26/2022]
Abstract
In this study the mutagenic, recombinagenic, carcinogenic and anticarcinogenic potential of orlistat was assessed using the somatic mutation and recombination test (SMART) and the epithelial tumor detection test (wts). The experiments were conducted on Drosophila melanogaster. In the assessment using SMART, larvae, descendants from the standard (ST) cross and the high bioactivation (HB) cross, were treated chronically with three orlistat concentrations. The results revealed a recombinagenic effect, associated with orlistat, in the descendants of the HB cross, at all three levels of concentration. Homologous recombination can function as a determinant at different stages of carcinogenesis. For verification, larvae from the wts test, descendants of the wts/TM3 virgin female and mwh/mwh male cross, were treated with the same three orlistat concentrations separately and in association with mitomicin C (0.1mM). The results did not, however, provide evidence that orlistat has carcinogenic potential nor was it associated with the reduction of tumors induced by mitomicin C in D. melanogaster.
Collapse
Affiliation(s)
- P C Orsolin
- Universidade Federal de Uberlândia, Instituto de Genética e Bioquímica, Bloco 2E, Campus Umuarama, Uberlândia, Minas Gerais, Brazil
| | | | | |
Collapse
|
35
|
Rauskolb C, Pan G, Reddy BVVG, Oh H, Irvine KD. Zyxin links fat signaling to the hippo pathway. PLoS Biol 2011; 9:e1000624. [PMID: 21666802 PMCID: PMC3110180 DOI: 10.1371/journal.pbio.1000624] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 04/27/2011] [Indexed: 01/15/2023] Open
Abstract
Using genetic and molecular analyses, the authors identify Zyx as a positive regulator of Hippo signaling and characterize its role within the pathway. The Hippo signaling pathway has a conserved role in growth control and is of fundamental importance during both normal development and oncogenesis. Despite rapid progress in recent years, key steps in the pathway remain poorly understood, in part due to the incomplete identification of components. Through a genetic screen, we identified the Drosophila Zyxin family gene, Zyx102 (Zyx), as a component of the Hippo pathway. Zyx positively regulates the Hippo pathway transcriptional co-activator Yorkie, as its loss reduces Yorkie activity and organ growth. Through epistasis tests, we position the requirement for Zyx within the Fat branch of Hippo signaling, downstream of Fat and Dco, and upstream of the Yorkie kinase Warts, and we find that Zyx is required for the influence of Fat on Warts protein levels. Zyx localizes to the sub-apical membrane, with distinctive peaks of accumulation at intercellular vertices. This partially overlaps the membrane localization of the myosin Dachs, which has similar effects on Fat-Hippo signaling. Co-immunoprecipitation experiments show that Zyx can bind to Dachs and that Dachs stimulates binding of Zyx to Warts. We also extend characterization of the Ajuba LIM protein Jub and determine that although Jub and Zyx share C-terminal LIM domains, they regulate Hippo signaling in distinct ways. Our results identify a role for Zyx in the Hippo pathway and suggest a mechanism for the role of Dachs: because Fat regulates the localization of Dachs to the membrane, where it can overlap with Zyx, we propose that the regulated localization of Dachs influences downstream signaling by modulating Zyx-Warts binding. Mammalian Zyxin proteins have been implicated in linking effects of mechanical strain to cell behavior. Our identification of Zyx as a regulator of Hippo signaling thus also raises the possibility that mechanical strain could be linked to the regulation of gene expression and growth through Hippo signaling. Processes that control cell numbers are essential during normal development, when they are required to generate organs of the correct size, and during cancinogenesis, when they influence tumor growth. The Hippo pathway is an intercellular signaling pathway that relays information about cell-cell contact and cell polarity to a signal transduction pathway that regulates the transcription of genes controlling cell numbers. The role of Hippo signaling in controlling growth is conserved from fruit flies to humans, but many aspects of the Hippo signal transduction pathway remain poorly understood. In this article, we identify Zyx as a previously unknown component of the Hippo pathway in Drosophila, and characterize its role within the pathway. We show that Zyx plays an essential role in a branch of Hippo signaling that involves the transmembrane receptor protein Fat and its target Dachs, which is a myosin family protein. Our results suggest a model in which Fat regulates the localization of Dachs, Dachs subsequently binds Zyx, stimulating its binding with the kinase Warts/Lats, and thereby regulates downstream signaling events. Zyx is conserved in vertebrates and we suggest that vertebrate Zyx proteins might also be involved in the regulation of Hippo signaling and, thereby, organ growth.
Collapse
Affiliation(s)
- Cordelia Rauskolb
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Guohui Pan
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - B. V. V. G. Reddy
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Hyangyee Oh
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kenneth D. Irvine
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
36
|
|
37
|
Abstract
First discovered in Drosophila, the Hippo signaling pathway is a conserved regulator of organ size. Central to this pathway is a kinase cascade leading from the tumor suppressor Hippo (Mst1 and Mst2 in mammals) to the oncoprotein Yki (YAP and TAZ in mammals), a transcriptional coactivator of target genes involved in cell proliferation and survival. Here, I review recent progress in elucidating the molecular mechanism and physiological function of Hippo signaling in Drosophila and mammals. These studies suggest that the core Hippo kinase cascade integrates multiple upstream inputs, enabling dynamic regulation of tissue homeostasis in animal development and physiology.
Collapse
|
38
|
SASAKI HIDEFUMI, HIKOSAKA YU, KAWANO OSAMU, YANO MOTOKI, FUJII YOSHITAKA. Hypermethylation of the large tumor suppressor genes in Japanese lung cancer. Oncol Lett 2010; 1:303-307. [PMID: 22966299 PMCID: PMC3436364 DOI: 10.3892/ol_00000054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Accepted: 09/03/2009] [Indexed: 11/05/2022] Open
Abstract
Large tumor suppressor (LATS) 1 and 2 are tumor suppressor genes implicated in the regulation of the cell cycle. The methylation statuses of the promoter regions of these genes were studied in Japanese lung cancers. The methylation statuses of the promoter regions of LATS1 and LATS2 were investigated by methylation-specific PCR. The findings were compared to clinicopathological features of lung cancer. Methylation-specific PCR showed that the LATS1 promoter region was hypermethylated in 95 out of 119 (79.8%) lung cancers. The methylation status of LATS1 was significantly associated with squamous histology (p=0.0267) and smoking status (never smoker vs. smoker; p=0.0399). LATS1-ummethylated patients harbored more EGFR mutations (p=0.0143). The LATS2 promoter region was hypermethylated in 160 out of 203 (78.8%) lung cancers. However, the methylation status had no association with the clinicopathological characteristics of the lung cancers cases. Both the LATS1 and LATS2 methylation statuses did not correlate with survival of lung cancer patients. Thus, the EGFR methylation status of the LATS genes has limited value in Japanese lung cancers.
Collapse
Affiliation(s)
- HIDEFUMI SASAKI
- Department of Oncology, Immunology and Surgery, Nagoya City University Medical School, Nagoya 467-8601, Japan
| | - YU HIKOSAKA
- Department of Oncology, Immunology and Surgery, Nagoya City University Medical School, Nagoya 467-8601, Japan
| | - OSAMU KAWANO
- Department of Oncology, Immunology and Surgery, Nagoya City University Medical School, Nagoya 467-8601, Japan
| | - MOTOKI YANO
- Department of Oncology, Immunology and Surgery, Nagoya City University Medical School, Nagoya 467-8601, Japan
| | - YOSHITAKA FUJII
- Department of Oncology, Immunology and Surgery, Nagoya City University Medical School, Nagoya 467-8601, Japan
| |
Collapse
|
39
|
The NDR kinase DBF-2 is involved in regulation of mitosis, conidial development, and glycogen metabolism in Neurospora crassa. EUKARYOTIC CELL 2009; 9:502-13. [PMID: 19966031 DOI: 10.1128/ec.00230-09] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Neurospora crassa dbf-2 encodes an NDR (nuclear Dbf2-related) protein kinase, homologous to LATS1, a core component of the Hippo pathway. This pathway plays important roles in restraining cell proliferation and promoting apoptosis in differentiating cells. Here, we demonstrate that DBF-2 is involved in three fundamental processes in a filamentous fungus: cell cycle regulation, glycogen biosynthesis, and conidiation. DBF-2 is predominantly localized to the nucleus, and most (approximately 60%) dbf-2 null mutant nuclei are delayed in mitosis, indicating that DBF-2 activity is required for properly completing the cell cycle. The dbf-2 mutant exhibits reduced basal hyphal extension rates accompanied by a carbon/nitrogen ratio-dependent bursting of hyphal tips, vast glycogen leakage, defects in aerial hypha formation, and impairment of all three asexual conidiation pathways in N. crassa. Our findings also indicate that DBF-2 is essential for sexual reproduction in a filamentous fungus. Defects in other Hippo and glycogen metabolism pathway components (mob-1, ccr-4, mst-1, and gsk-3) share similar phenotypes such as mitotic delay and decreased CDC-2 (cell division cycle 2) protein levels, massive hyphal swellings, hyphal tip bursting, glycogen leakage, and impaired conidiation. We propose that DBF-2 functions as a link between Hippo and glycogen metabolism pathways.
Collapse
|
40
|
Siam R, Harada R, Cadieux C, Battat R, Vadnais C, Nepveu A. Transcriptional activation of the Lats1 tumor suppressor gene in tumors of CUX1 transgenic mice. Mol Cancer 2009; 8:60. [PMID: 19656388 PMCID: PMC2731069 DOI: 10.1186/1476-4598-8-60] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2009] [Accepted: 08/05/2009] [Indexed: 02/01/2023] Open
Abstract
Background Lats1 (large tumor suppressor 1) codes for a serine/threonine kinase that plays a role in the progression through mitosis. Genetic studies demonstrated that the loss of LATS1 in mouse, and of its ortholog wts (warts) in Drosophila, is associated with increased cancer incidence. There are conflicting reports, however, as to whether overexpression of Lats1 inhibits cell proliferation. CUX1 is a transcription factor that exists in different isoforms as a result of proteolytic processing or alternative transcription initiation. Expression of p110 and p75 CUX1 in transgenic mice increases the susceptibility to cancer in various organs and tissues. In tissue culture, p110 CUX1 was shown to accelerate entry into S phase and stimulate cell proliferation. Results Genome-wide location arrays in cell lines of various cell types revealed that Lats1 was a transcriptional target of CUX1. Scanning ChIP analysis confirmed that CUX1 binds to the immediate promoter of Lats1. Expression of Lats1 was reduced in cux1-/- MEFs, whereas it was increased in cells stably or transiently expressing p110 or p75 CUX1. Reporter assays confirmed that the immediate promoter of Lats1 was sufficient to confer transcriptional activation by CUX1. Lats1 was found to be overexpressed in tumors from the mammary gland, uterus and spleen that arise in p110 or p75 CUX1 transgenic mice. In tissue culture, such elevated LATS1 expression did not hinder cell cycle progression in cells overexpressing p110 CUX1. Conclusion While inactivation of Lats1/wts in mouse and Drosophila can increase cancer incidence, results from the present study demonstrate that Lats1 is a transcriptional target of CUX1 that can be overexpressed in tumors of various tissue-types. Interestingly, two other studies documented the overexpression of LATS1 in human cervical cancers and basal-like breast cancers. We conclude that, similarly to other genes involved in mitotic checkpoint, cancer can be associated with either loss-of-function or overexpression of Lats1.
Collapse
Affiliation(s)
- Rania Siam
- Goodman Cancer Center, McGill University, 1160 Pine Avenue West, Room 410, Montreal, Quebec H3A 1A3, Canada.
| | | | | | | | | | | |
Collapse
|
41
|
Reddy BVVG, Irvine KD. The Fat and Warts signaling pathways: new insights into their regulation,mechanism and conservation. Development 2008; 135:2827-38. [DOI: 10.1242/dev.020974] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A cassette of cytoplasmic Drosophila tumor suppressors, including the kinases Hippo and Warts, has recently been linked to the transmembrane tumor suppressor Fat. These proteins act within interconnected signaling pathways, the principal functions of which are to control the growth and polarity of developing tissues. Recent studies have enhanced our understanding of the basis for signal transduction by Fat and Warts pathways, including the identification of a DNA-binding protein at the end of the pathway, have established the conservation of Fat and Warts signaling from flies to mammals,and have given us new insights into their regulation and biological functions.
Collapse
Affiliation(s)
- B. V. V. G. Reddy
- Howard Hughes Medical Institute, Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kenneth D. Irvine
- Howard Hughes Medical Institute, Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
42
|
Simcox A, Mitra S, Truesdell S, Paul L, Chen T, Butchar JP, Justiniano S. Efficient genetic method for establishing Drosophila cell lines unlocks the potential to create lines of specific genotypes. PLoS Genet 2008; 4:e1000142. [PMID: 18670627 PMCID: PMC2474701 DOI: 10.1371/journal.pgen.1000142] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Accepted: 06/24/2008] [Indexed: 12/22/2022] Open
Abstract
Analysis of cells in culture has made substantial contributions to biological research. The versatility and scale of in vitro manipulation and new applications such as high-throughput gene silencing screens ensure the continued importance of cell-culture studies. In comparison to mammalian systems, Drosophila cell culture is underdeveloped, primarily because there is no general genetic method for deriving new cell lines. Here we found expression of the conserved oncogene RasV12 (a constitutively activated form of Ras) profoundly influences the development of primary cultures derived from embryos. The cultures become confluent in about three weeks and can be passaged with great success. The lines have undergone more than 90 population doublings and therefore constitute continuous cell lines. Most lines are composed of spindle-shaped cells of mesodermal type. We tested the use of the method for deriving Drosophila cell lines of a specific genotype by establishing cultures from embryos in which the warts (wts) tumor suppressor gene was targeted. We successfully created several cell lines and found that these differ from controls because they are primarily polyploid. This phenotype likely reflects the known role for the mammalian wts counterparts in the tetraploidy checkpoint. We conclude that expression of RasV12 is a powerful genetic mechanism to promote proliferation in Drosophila primary culture cells and serves as an efficient means to generate continuous cell lines of a given genotype. In Drosophila, the genetic analysis of whole animals has been the focus of the field and has been exceptionally successful. Gene discoveries in flies have led to parallel studies in vertebrates and hence have accelerated the understanding of biology. Furthermore, some 60–70% of human disease genes are conserved in Drosophila, thus making the genetically tractable fly a useful disease model. While the whole-organism approach in Drosophila is powerful, there are studies that can best be conducted in cell lines. In this regard, Drosophila lags far behind mammalian systems, in which creation of cell lines using genetic manipulation is routine. We sought to test whether similar genetic approaches could be used in Drosophila. We discovered a simple genetic method for the rapid production of fly cell lines using an activated oncogene to stimulate proliferation in cultured embryonic cells. The method has immediate application for creating custom cell lines of a given genotype. We provided an example of this by making lines in which a tumor suppressor gene is targeted. Specifically designed cell lines will be extremely valuable for gene discovery using whole-genome RNAi screens and for producing large numbers of cells of a specific genotype for biochemical studies.
Collapse
Affiliation(s)
- Amanda Simcox
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio, United States of America.
| | | | | | | | | | | | | |
Collapse
|
43
|
Zhang J, Smolen GA, Haber DA. Negative regulation of YAP by LATS1 underscores evolutionary conservation of the Drosophila Hippo pathway. Cancer Res 2008; 68:2789-94. [PMID: 18413746 DOI: 10.1158/0008-5472.can-07-6205] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Hippo pathway defines a novel signaling cascade regulating cell proliferation and survival in Drosophila, which involves the negative regulation of the transcriptional coactivator Yorkie by the kinases Hippo and Warts. We have recently shown that the human ortholog of Yorkie, YAP, maps to a minimal amplification locus in mouse and human cancers, and that it mediates dramatic transforming activity in MCF10A primary mammary epithelial cells. Here, we show that LATS proteins (mammalian orthologs of Warts) interact directly with YAP in mammalian cells and that ectopic expression of LATS1, but not LATS2, effectively suppresses the YAP phenotypes. Furthermore, shRNA-mediated knockdown of LATS1 phenocopies YAP overexpression. Because this effect can be suppressed by simultaneous YAP knockdown, it suggests that YAP is the primary target of LATS1 in mammalian cells. Expression profiling of genes induced by ectopic expression of YAP or by knockdown of LATS1 reveals a subset of potential Hippo pathway targets implicated in epithelial-to-mesenchymal transition, suggesting that this is a key feature of YAP signaling in mammalian cells.
Collapse
Affiliation(s)
- Jianmin Zhang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | |
Collapse
|
44
|
The mob as tumor suppressor gene is essential for early development and regulates tissue growth in Drosophila. Genetics 2008; 178:957-65. [PMID: 18245354 DOI: 10.1534/genetics.107.081570] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Studies in Drosophila have defined a new growth inhibitory pathway mediated by Fat (Ft), Merlin (Mer), Expanded (Ex), Hippo (Hpo), Salvador (Sav)/Shar-pei, Warts (Wts)/Large tumor suppressor (Lats), and Mob as tumor suppressor (Mats), which are all evolutionarily conserved in vertebrate animals. We previously found that the Mob family protein Mats functions as a coactivator of Wts kinase. Here we show that mats is essential for early development and is required for proper chromosomal segregation in developing embryos. Mats is expressed at low levels ubiquitously, which is consistent with the role of Mats as a general growth regulator. Like mammalian Mats, Drosophila Mats colocalizes with Wts/Lats kinase and cyclin E proteins at the centrosome. This raises the possibility that Mats may function together with Wts/Lats to regulate cyclin E activity in the centrosome for mitotic control. While Hpo/Wts signaling has been implicated in the control of cyclin E and diap1 expression, we found that it also modulates the expression of cyclin A and cyclin B. Although mats depletion leads to aberrant mitoses, this does not seem to be due to compromised mitotic spindle checkpoint function.
Collapse
|
45
|
Hergovich A, Cornils H, Hemmings BA. Mammalian NDR protein kinases: from regulation to a role in centrosome duplication. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2007; 1784:3-15. [PMID: 17881309 DOI: 10.1016/j.bbapap.2007.07.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Revised: 07/20/2007] [Accepted: 07/24/2007] [Indexed: 02/06/2023]
Abstract
The NDR (nuclear Dbf2-related) family of kinases is highly conserved from yeast to human, and has been classified as a subgroup of the AGC group of protein kinases based on the sequence of the catalytic domain. Like all other members of the AGC class of protein kinases, NDR kinases require the phosphorylation of conserved Ser/Thr residues for activation. Importantly, NDR family members have two unique stretches of primary sequence: an N-terminal regulatory (NTR) domain and an insert of several residues between subdomains VII and VIII of the kinase domain. The kinase domain insert functions as an auto-inhibitory sequence (AIS), while binding of the co-activator MOB (Mps-one binder) proteins to the NTR domain releases NDR kinases from inhibition of autophosphorylation. However, despite such advances in our understanding of the molecular activation mechanism(s) and physiological functions of NDR kinases in yeast and invertebrates, most biological NDR substrates still remain to be identified. Nevertheless, by showing that the centrosomal subpopulation of human NDR1/2 is required for proper centrosome duplication, the first biological role of human NDR1/2 kinases has been defined recently. How far NDR-driven centrosome overduplication could actually contribute to cellular transformation will also be discussed.
Collapse
Affiliation(s)
- Alexander Hergovich
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058, Basel, Switzerland
| | | | | |
Collapse
|
46
|
Abstract
The control of organ (or organism) size is a fundamental aspect of life that has long captured human imagination. What makes an elephant grow a million times larger than a mouse? How do our two hands develop independently of each other yet reach very similar size? How does a liver precisely regenerate its original mass when two-thirds of it is removed? The recent discovery of a novel signaling network in Drosophila, known as the Hippo (Hpo) pathway, might provide an important entry point to these fascinating questions. The Hpo pathway consists of several negative growth regulators acting in a kinase cascade that ultimately phosphorylates and inactivates Yorkie (Yki), a transcriptional coactivator that positively regulates cell growth, survival, and proliferation. Components of the Hpo pathway are highly conserved throughout evolution, suggesting that this pathway may function as a global regulator of tissue homeostasis in all metazoan animals. Here, I provide a historical review of this potent growth-regulatory pathway and highlight outstanding questions that will likely be the focus of future investigation.
Collapse
Affiliation(s)
- Duojia Pan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| |
Collapse
|
47
|
Yabuta N, Okada N, Ito A, Hosomi T, Nishihara S, Sasayama Y, Fujimori A, Okuzaki D, Zhao H, Ikawa M, Okabe M, Nojima H. Lats2 is an essential mitotic regulator required for the coordination of cell division. J Biol Chem 2007; 282:19259-71. [PMID: 17478426 DOI: 10.1074/jbc.m608562200] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Tumor suppressor Lats2 is a member of the conserved Dbf2 kinase family. It localizes to the centrosome and has been implicated in regulation of the cell cycle and apoptosis. However, the in vivo function of this kinase remains unclear. Here, we show that complete disruption of the gene encoding Lats2 in mice causes developmental defects in the nervous system and embryonic lethality. Furthermore, mutant cells derived from total LATS2-knock-out embryos exhibit mitotic defects including centrosome fragmentation and cytokinesis defects, followed by nuclear enlargement and multinucleation. We show that the Mob1 family, a regulator of mitotic exit, associates with Lats2 to induce its activation. We also show that the complete LATS2-knock-out cells exhibit an acceleration of exit from mitosis and marked down-regulation of critical mitotic regulators. These results suggest that Lats2 plays an essential mitotic role in coordinating accurate cytokinesis completion, governing the stabilization of other mitotic regulators.
Collapse
Affiliation(s)
- Norikazu Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Guo C, Tommasi S, Liu L, Yee JK, Dammann R, Pfeifer GP. RASSF1A is part of a complex similar to the Drosophila Hippo/Salvador/Lats tumor-suppressor network. Curr Biol 2007; 17:700-5. [PMID: 17379520 DOI: 10.1016/j.cub.2007.02.055] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Revised: 02/24/2007] [Accepted: 02/26/2007] [Indexed: 12/21/2022]
Abstract
The Ras Association Domain Family 1A (RASSF1A) gene is one of the most frequently silenced genes in human cancer. RASSF1A has been shown to interact with the proapoptotic kinase MST1. Recent work in Drosophila has led to the discovery of a new tumor-suppressor pathway involving the Drosophila MST1 and MST2 ortholog, Hippo, as well as the Lats/Warts serine/threonine kinase and a protein named Salvador (Sav). Little is known about this pathway in mammalian cells. We report that complexes consisting of RASSF1A, MST2, WW45 (the human ortholog of Sav), and LATS1 exist in human cells. MST2 enhances the RASSF1A-WW45 interaction, which requires the C-terminal SARAH domain of both proteins. Components of this complex are localized at centrosomes and spindle poles from interphase to telophase and at the midbody during cytokinesis. Both RASSF1A and WW45 activate MST2 by promoting MST2 autophosphorylation and LATS1 phosphorylation. Mitosis is delayed in Rassf1a(-/-) mouse embryo fibroblasts and frequently results in cytokinesis failure, similar to what has been observed for LATS1-deficient cells. RASSF1A, MST2, or WW45 can rescue this defect. The complex of RASSF1A, MST2, WW45, and LATS1 consists of several tumor suppressors, is conserved in mammalian cells, and appears to be involved in controlling mitotic exit.
Collapse
Affiliation(s)
- Cai Guo
- Division of Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010, USA
| | | | | | | | | | | |
Collapse
|
49
|
Aylon Y, Michael D, Shmueli A, Yabuta N, Nojima H, Oren M. A positive feedback loop between the p53 and Lats2 tumor suppressors prevents tetraploidization. Genes Dev 2007; 20:2687-700. [PMID: 17015431 PMCID: PMC1578695 DOI: 10.1101/gad.1447006] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Damage to the mitotic spindle and centrosome dysfunction can lead to cancer. To prevent this, cells trigger a succession of checkpoint responses, where an initial mitotic delay is followed by slippage without cytokinesis, spawning tetraploid G1 cells that undergo a p53-dependent G1/S arrest. We describe the importance of Lats2 (Large Tumor Suppressor 2) in this checkpoint response. Lats2 binds Mdm2, inhibits its E3 ligase activity, and activates p53. Nocodazole, a microtubule poison that provokes centrosome/mitotic apparatus dysfunction, induces Lats2 translocation from centrosomes to the nucleus and p53 accumulation. In turn, p53 rapidly and selectively up-regulates Lats2 expression in G2/M cells, thereby defining a positive feedback loop. Abrogation of Lats2 promotes accumulation of polyploid cells upon exposure to nocodazole, which can be prevented by direct activation of p53. The Lats2-Mdm2-p53 axis thus constitutes a novel checkpoint pathway critical for the maintenance of proper chromosome number.
Collapse
Affiliation(s)
- Yael Aylon
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | |
Collapse
|
50
|
Kuninaka S, Iida SI, Hara T, Nomura M, Naoe H, Morisaki T, Nitta M, Arima Y, Mimori T, Yonehara S, Saya H. Serine protease Omi/HtrA2 targets WARTS kinase to control cell proliferation. Oncogene 2006; 26:2395-406. [PMID: 17130845 DOI: 10.1038/sj.onc.1210042] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The serine protease Omi/HtrA2 was initially regarded as a proapoptotic molecule that proteolyses several proteins to induce cell death. Recent studies, however, indicate that loss of Omi protease activity increases susceptibility to stress-induced cell death. These complicated findings suggest that the protease activity of Omi is involved not only in apoptosis but also in cellular homeostasis. However, the targets which Omi uses to mediate this novel process are unknown. Previously, we showed that WARTS (WTS)/large tumor-suppressor 1 mitotic kinase interacts with the protein/discs-large protein/zonula (PDZ) domain of Omi and promotes its protease activity. We now report that WTS is a substrate for Omi protease activity, thus it is not only a regulator but also a downstream target of this protease. Interaction with Omi PDZ domain is required for WTS to be proteolysed. When caspase-9-deficient mouse embryonic fibroblasts (MEFs) were treated with staurosporine, WTS was proteolysed by activated endogenous Omi without induction of cell death. Therefore, protease activity of Omi and proteolysis of WTS are not necessarily required for cell death. We found that depletion of Omi from HeLa cells results in accelerated cell proliferation despite no significant change in the duration of mitosis. The depletion of WTS showed the same effect on S phase progression. Therefore, WTS proteolytic fragment(s) generated by Omi may act as an inhibitor of G1/S progression. Our data reveal a role for Omi-mediated processing of WTS in negative regulation of cell cycle progression at interphase, suggesting a novel function of Omi other than apoptosis.
Collapse
Affiliation(s)
- S Kuninaka
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Kumamoto, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|