1
|
Huang Y, He J, Wang Y, Li L, Lin S. Nitrogen source type modulates heat stress response in coral symbiont ( Cladocopium goreaui). Appl Environ Microbiol 2025:e0059124. [PMID: 39772785 DOI: 10.1128/aem.00591-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/09/2024] [Indexed: 01/11/2025] Open
Abstract
Ocean warming due to climate change endangers coral reefs, and regional nitrogen overloading exacerbates the vulnerability of reef-building corals as the dual stress disrupts coral-Symbiodiniaceae mutualism. Different forms of nitrogen may create different interactive effects with thermal stress, but the underlying mechanisms remain elusive. To address the gap, we measured and compared the physiological and transcriptional responses of the Symbiodiniaceae Cladocopium goreaui to heat stress (31°C) when supplied with different types of nitrogen (nitrate, ammonium, or urea). Under heat stress (HS), cell proliferation and photosynthesis of C. goreaui declined, while cell size, lipid storage, and total antioxidant capacity increased, both to varied extents depending on the nitrogen type. Nitrate-cultured cells exhibited the most robust acclimation to HS, as evidenced by the fewest differentially expressed genes (DEGs) and less ROS accumulation, possibly due to activated nitrate reduction and enhanced ascorbate biogenesis. Ammonium-grown cultures exhibited higher algal proliferation and ROS scavenging capacity due to enhanced carotenoid and ascorbate quenching, but potentially reduced host recognizability due to the downregulation of N-glycan biosynthesis genes. Urea utilization led to the greatest ROS accumulation as genes involved in photorespiration, plant respiratory burst oxidase (RBOH), and protein refolding were markedly upregulated, but the greatest cutdown in photosynthate potentially available to corals as evidenced by photoinhibition and selfish lipid storage, indicating detrimental effects of urea overloading. The differential warming nitrogen-type interactive effects documented here has significant implication in coral-Symbiodiniaceae mutualism, which requires further research.IMPORTANCERegional nitrogen pollution exacerbates coral vulnerability to globally rising sea-surface temperature, with different nitrogen types exerting different interactive effects. How this occurs is poorly understood and understudied. This study explored the underlying mechanism by comparing physiological and transcriptional responses of a coral symbiont to heat stress under different nitrogen supplies (nitrate, ammonium, and urea). The results showed some common, significant responses to heat stress as well as some unique, N-source dependent responses. These findings underscore that nitrogen eutrophication is not all the same, the form of nitrogen pollution should be considered in coral conservation, and special attention should be given to urea pollution.
Collapse
Affiliation(s)
- Yulin Huang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiamin He
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yujie Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Ling Li
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Senjie Lin
- Department of Marine Sciences, University of Connecticut, Groton, Connecticut, USA
| |
Collapse
|
2
|
Gu R, Wu T, Fu J, Sun YJ, Sun XX. Advances in the genetic etiology of female infertility. J Assist Reprod Genet 2024; 41:3261-3286. [PMID: 39320554 PMCID: PMC11707141 DOI: 10.1007/s10815-024-03248-w] [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: 04/19/2024] [Accepted: 08/07/2024] [Indexed: 09/26/2024] Open
Abstract
Human reproduction is a complex process involving gamete maturation, fertilization, embryo cleavage and development, blastocyst formation, implantation, and live birth. If any of these processes are abnormal or arrest, reproductive failure will occur. Infertility is a state of reproductive dysfunction caused by various factors. Advances in molecular genetics, including cell and molecular genetics, and high-throughput sequencing technologies, have found that genetic factors are important causes of infertility. Genetic variants have been identified in infertile women or men and can cause gamete maturation arrest, poor quality gametes, fertilization failure, and embryonic developmental arrest during assisted reproduction technology (ART), and thus reduce the clinical success rates of ART. This article reviews clinical studies on repeated in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) failures caused by ovarian dysfunction, oocyte maturation defects, oocyte abnormalities, fertilization disorders, and preimplantation embryonic development arrest due to female genetic etiology, the accumulation of pathogenic genes and gene pathogenic loci, and the functional mechanism and clinical significance of pathogenic genes in gametogenesis and early embryonic development.
Collapse
Affiliation(s)
- Ruihuan Gu
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China
| | - Tianyu Wu
- Institute of Pediatrics, State Key Laboratory of Genetic Engineering, Institutes of BiomedicalSciences, Shanghai Key Laboratory of Medical Epigenetics, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Jing Fu
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China
| | - Yi-Juan Sun
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China.
| | - Xiao-Xi Sun
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China.
| |
Collapse
|
3
|
Lin Y, Kong L, Zhao Y, Zhai F, Zhan Z, Li Y, Jingfei Z, Chunhong Y, Jin X. The oncogenic role of EIF4A3/CDC20 axis in the endometrial cancer. J Mol Med (Berl) 2024; 102:1395-1410. [PMID: 39316093 DOI: 10.1007/s00109-024-02486-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024]
Abstract
Eukaryotic initiation factor 4A-3 (EIF4A3) is a key component of the exon junction complex (EJC) and is extensively involved in RNA splicing, inducing mRNA decay, and regulating the cell cycle and apoptosis. However, the potential role of EIF4A3 in EC has not been comprehensively investigated and remains unknown. Here, we report that the expression level of EIF4A3 is dramatically elevated in endometrial cancer (EC) samples compared with normal EC samples via bioinformatics analysis and immunohistochemistry analysis, and that high expression of EIF4A3 promotes the proliferation, migration, and invasion of EC cells. Mechanistically, we found that high EIF4A3 expression stabilized cell division cyclin 20 (CDC20) mRNA, and high EIF4A3 expression induced pro-carcinogenic effects in EC cells that were efficiently antagonized upon knockdown of CDC20, as well as Apcin, an inhibitor of CDC20. These findings reveal a novel mechanism by which high expression of EIF4A3 induces CDC20 upregulation, thus leading to EC tumorigenesis and metastasis, indicating a potential treatment strategy for EC patients with high EIF4A3 expression using Apcin. KEY MESSAGES: The expression level of EIF4A3 was dramatically elevated in endometrial cancer (EC) samples compared with normal endometrial cancer samples. High EIF4A3 expression stabilized CDC20 mRNA, and high EIF4A3 expression induced pro-carcinogenic effect in EC cells which was efficiently antagonized upon knockdown of CDC20. Apcin, an inhibitor of CDC20, could effectively counteract high expression of EIF4A3 inducing EC tumourigenesis and metastasis, indicating the potential treatment strategy for EC patients with EIF4A3 high expression by using Apcin.
Collapse
Affiliation(s)
- Yan Lin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lili Kong
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yiting Zhao
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Ziqing Zhan
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yuxuan Li
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Zheng Jingfei
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Yan Chunhong
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
4
|
Meng L, Dong R, Mi W, Qin K, Ouyang K, Sun J, Li J. The ubiquitin E3 ligase APC/C Cdc20 mediates mitotic degradation of OGT. J Biol Chem 2024; 300:107448. [PMID: 38844135 PMCID: PMC11261447 DOI: 10.1016/j.jbc.2024.107448] [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: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 07/01/2024] Open
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) is the sole enzyme that catalyzes all O-GlcNAcylation reactions intracellularly. Previous investigations have found that OGT levels oscillate during the cell division process. Specifically, OGT abundance is downregulated during mitosis, but the underlying mechanism is lacking. Here we demonstrate that OGT is ubiquitinated by the ubiquitin E3 ligase, anaphase promoting complex/cyclosome (APC/C)-cell division cycle 20 (Cdc20). We show that APC/CCdc20 interacts with OGT through a conserved destruction box (D-box): Arg-351/Leu-354, the abrogation of which stabilizes OGT. As APC/CCdc20-substrate binding is often preceded by a priming ubiquitination event, we also used mass spectrometry and mapped OGT Lys-352 to be a ubiquitination site, which is a prerequisite for OGT association with APC/C subunits. Interestingly, in The Cancer Genome Atlas, R351C is a uterine carcinoma mutant, suggesting that mutations of the D-box are linked with tumorigenesis. Paradoxically, we found that both R351C and the D-box mutants (R351A/L354A) inhibit uterine carcinoma in mouse xenograft models, probably due to impaired cell division and proliferation. In sum, we propose a model where OGT Lys-352 ubiquitination primes its binding with APC/C, and then APC/CCdc20 partners with OGT through the D-box for its mitotic destruction. Our work not only highlights the key mechanism that regulates OGT during the cell cycle, but also reveals the mutual coordination between glycosylation and the cell division machinery.
Collapse
Affiliation(s)
- Li Meng
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
| | - Rui Dong
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Weixiao Mi
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
| | - Ke Qin
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Jianwei Sun
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China.
| | - Jing Li
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China.
| |
Collapse
|
5
|
Wang J, Zhang T, Tu A, Xie H, Hu H, Chen J, Yang J. Genome-Wide Identification and Analysis of APC E3 Ubiquitin Ligase Genes Family in Triticum aestivum. Genes (Basel) 2024; 15:271. [PMID: 38540330 PMCID: PMC10970508 DOI: 10.3390/genes15030271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 06/15/2024] Open
Abstract
E3 ubiquitin ligases play a pivotal role in ubiquitination, a crucial post-translational modification process. Anaphase-promoting complex (APC), a large cullin-RING E3 ubiquitin ligase, regulates the unidirectional progression of the cell cycle by ubiquitinating specific target proteins and triggering plant immune responses. Several E3 ubiquitin ligases have been identified owing to advancements in sequencing and annotation of the wheat genome. However, the types and functions of APC E3 ubiquitin ligases in wheat have not been reported. This study identified 14 members of the APC gene family in the wheat genome and divided them into three subgroups (CCS52B, CCS52A, and CDC20) to better understand their functions. Promoter sequence analysis revealed the presence of several cis-acting elements related to hormone and stress responses in the APC E3 ubiquitin ligases in wheat. All identified APC E3 ubiquitin ligase family members were highly expressed in the leaves, and the expression of most genes was induced by the application of methyl jasmonate (MeJA). In addition, the APC gene family in wheat may play a role in plant defense mechanisms. This study comprehensively analyzes APC genes in wheat, laying the groundwork for future research on the function of APC genes in response to viral infections and expanding our understanding of wheat immunity mechanisms.
Collapse
Affiliation(s)
- Jinnan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Tianye Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Aizhu Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Haoxin Xie
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Haichao Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Jianping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Jian Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China; (J.W.); (T.Z.); (A.T.); (H.X.); (H.H.)
| |
Collapse
|
6
|
Lamas-Maceiras M, Vizoso-Vázquez Á, Barreiro-Alonso A, Cámara-Quílez M, Cerdán ME. Thanksgiving to Yeast, the HMGB Proteins History from Yeast to Cancer. Microorganisms 2023; 11:microorganisms11040993. [PMID: 37110415 PMCID: PMC10142021 DOI: 10.3390/microorganisms11040993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Yeasts have been a part of human life since ancient times in the fermentation of many natural products used for food. In addition, in the 20th century, they became powerful tools to elucidate the functions of eukaryotic cells as soon as the techniques of molecular biology developed. Our molecular understandings of metabolism, cellular transport, DNA repair, gene expression and regulation, and the cell division cycle have all been obtained through biochemistry and genetic analysis using different yeasts. In this review, we summarize the role that yeasts have had in biological discoveries, the use of yeasts as biological tools, as well as past and on-going research projects on HMGB proteins along the way from yeast to cancer.
Collapse
Affiliation(s)
- Mónica Lamas-Maceiras
- Centro Interdisciplinar de Química y Biología (CICA), As Carballeiras, s/n, Campus de Elviña, Universidade da Coruña, 15071 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), As Xubias de Arriba 84, 15006 A Coruña, Spain
- Facultad de Ciencias, A Fraga, s/n, Campus de A Zapateira, Universidade da Coruña, 15071 A Coruña, Spain
| | - Ángel Vizoso-Vázquez
- Centro Interdisciplinar de Química y Biología (CICA), As Carballeiras, s/n, Campus de Elviña, Universidade da Coruña, 15071 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), As Xubias de Arriba 84, 15006 A Coruña, Spain
- Facultad de Ciencias, A Fraga, s/n, Campus de A Zapateira, Universidade da Coruña, 15071 A Coruña, Spain
| | - Aida Barreiro-Alonso
- Centro Interdisciplinar de Química y Biología (CICA), As Carballeiras, s/n, Campus de Elviña, Universidade da Coruña, 15071 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), As Xubias de Arriba 84, 15006 A Coruña, Spain
- Facultad de Ciencias, A Fraga, s/n, Campus de A Zapateira, Universidade da Coruña, 15071 A Coruña, Spain
| | - María Cámara-Quílez
- Centro Interdisciplinar de Química y Biología (CICA), As Carballeiras, s/n, Campus de Elviña, Universidade da Coruña, 15071 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), As Xubias de Arriba 84, 15006 A Coruña, Spain
- Facultad de Ciencias, A Fraga, s/n, Campus de A Zapateira, Universidade da Coruña, 15071 A Coruña, Spain
| | - María Esperanza Cerdán
- Centro Interdisciplinar de Química y Biología (CICA), As Carballeiras, s/n, Campus de Elviña, Universidade da Coruña, 15071 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), As Xubias de Arriba 84, 15006 A Coruña, Spain
- Facultad de Ciencias, A Fraga, s/n, Campus de A Zapateira, Universidade da Coruña, 15071 A Coruña, Spain
| |
Collapse
|
7
|
Bhattacharjee D, Kaveti S, Jain N. APC/C CDH1 ubiquitinates STAT3 in mitosis. Int J Biochem Cell Biol 2023; 154:106333. [PMID: 36400381 DOI: 10.1016/j.biocel.2022.106333] [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: 06/30/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
STAT3, an oncogene drives tumor growth and is associated with poor prognosis. However, small molecule-based STAT3 inhibitors were unsuccessful in clinics. Recently, STAT3 degraders that ubiquitinate STAT3 were found to elicit long-lasting anti-tumor responses. Thus, triggering STAT3 ubiquitination in cancers is a better strategy than STAT3 inhibition. However, not much is known about the identity of E3-ligases that ubiquitinate STAT3 in cancers. Therefore, to design better therapies to degrade STAT3, we sought to identify E3-ligases that ubiquitinate STAT3 in cancer cells. To answer this question, we determined the cell cycle-dependent ubiquitination of STAT3 in HEK293T cells and examined the link between STAT3 dephosphorylation and ubiquitination. We found that STAT3 is more strongly ubiquitinated in mitosis than in other phases of the cell cycle. We observed that APC/C CDH1 binds and ubiquitinates STAT3 in mitosis. Further, we also found that inhibiting phosphatases decreases STAT3 ubiquitination. We conclude that APC/C CDH1 ubiquitinates STAT3 in mitosis. We suggest that mitosis can be a potential therapeutic window for treating STAT3-activated cancers.
Collapse
Affiliation(s)
- Debanjan Bhattacharjee
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Sreeram Kaveti
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Nishant Jain
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
8
|
Efficient terminal erythroid differentiation requires the APC/C cofactor Cdh1 to limit replicative stress in erythroblasts. Sci Rep 2022; 12:10489. [PMID: 35729193 PMCID: PMC9213546 DOI: 10.1038/s41598-022-14331-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/06/2022] [Indexed: 11/08/2022] Open
Abstract
The APC/C-Cdh1 ubiquitin ligase complex drives proteosomal degradation of cell cycle regulators and other cellular proteins during the G1 phase of the cycle. The complex serves as an important modulator of the G1/S transition and prevents premature entry into S phase, genomic instability, and tumor development. Additionally, mounting evidence supports a role for this complex in cell differentiation, but its relevance in erythropoiesis has not been addressed so far. Here we show, using mouse models of Cdh1 deletion, that APC/C-Cdh1 activity is required for efficient terminal erythroid differentiation during fetal development as well as postnatally. Consistently, Cdh1 ablation leads to mild but persistent anemia from birth to adulthood. Interestingly, loss of Cdh1 seems to affect both, steady-state and stress erythropoiesis. Detailed analysis of Cdh1-deficient erythroid populations revealed accumulation of DNA damage in maturing erythroblasts and signs of delayed G2/M transition. Moreover, through direct assessment of replication dynamics in fetal liver cells, we uncovered slow fork movement and increased origin usage in the absence of Cdh1, strongly suggesting replicative stress to be the underlying cause of DNA lesions and cell cycle delays in erythroblasts devoid of Cdh1. In turn, these alterations would restrain full maturation of erythroblasts into reticulocytes and reduce the output of functional erythrocytes, leading to anemia. Our results further highlight the relevance of APC/C-Cdh1 activity for terminal differentiation and underscore the need for precise control of replication dynamics for efficient supply of red blood cells.
Collapse
|
9
|
Matsumura Y, Osborne TF, Sakai J. Epigenetic and environmental regulation of adipocyte function. J Biochem 2022; 172:9-16. [PMID: 35476139 DOI: 10.1093/jb/mvac033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/26/2022] [Indexed: 11/14/2022] Open
Abstract
Adipocytes play an essential role in the maintenance of whole-body energy homeostasis. White adipocytes regulate energy storage, whereas brown and beige adipocytes regulate energy expenditure and heat production. De novo production of adipocytes (i.e., adipogenesis) and their functions are dynamically controlled by environmental cues. Environmental changes (e.g., temperature, nutrients, hormones, cytokines) are transmitted via intracellular signaling to facilitate short-term responses and long-term adaptation in adipocytes; however, the molecular mechanisms that link the environment and epigenome are poorly understood. Our recent studies have demonstrated that environmental cues dynamically regulate interactions between transcription factors and epigenomic chromatin regulators, which together trigger combinatorial changes in chromatin structure to influence gene expression in adipocytes. Thus, environmental sensing by the concerted action of multiple chromatin-associated protein complexes is a key determinant of the epigenetic regulation of adipocyte functions.
Collapse
Affiliation(s)
- Yoshihiro Matsumura
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Timothy F Osborne
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, and Medicine in the Division of Endocrinology, Diabetes and Metabolism of the Johns Hopkins University School of Medicine, Petersburg, FL, USA
| | - Juro Sakai
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan.,Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| |
Collapse
|
10
|
Chen L, Qing J, Xiao Y, Huang X, Chi Y, Chen Z. TIM-1 promotes proliferation and metastasis, and inhibits apoptosis, in cervical cancer through the PI3K/AKT/p53 pathway. BMC Cancer 2022; 22:370. [PMID: 35392845 PMCID: PMC8991826 DOI: 10.1186/s12885-022-09386-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/07/2022] [Indexed: 12/09/2022] Open
Abstract
Background T-cell immunoglobulin mucin-1 (TIM-1) has been reported to be associated with the biological behavior of several malignant tumors; however, it is not clear whether it has a role in cervical cancer (CC). Methods TIM-1 expression in cervical epithelial tumor tissues and cells was detected by immunohistochemistry or real-time quantitative-PCR and western blotting. CC cells from cell lines expressing low levels of TIM-1 were infected with lentiviral vectors encoding TIM-1. Changes in the malignant behavior of CC cells were assessed by CCK-8, wound healing, Transwell migration and invasion assays, and flow cytometry in vitro; while a xenograft tumor model was established to analyze the effects of TIM-1 on tumor growth in vivo. Changes in the levels of proteins related to the cell cycle, apoptosis, and Epithelial-mesenchymal transition (EMT) were determined by western blotting. Results TIM-1 expression was higher in CC tissues, than in high grade squamous intraepithelial lesion, low grade squamous intraepithelial lesion, or normal cervical tissues, and was also expressed in three CC cell lines. In HeLa and SiHa cells overexpressing TIM-1, proliferation, invasion, and migration increased, while whereas apoptosis was inhibited. Furthermore, TIM-1 downregulated the expression of p53, BAX, and E-cadherin, and increased cyclin D1, Bcl-2, Snail1, N-cadherin, vimentin, MMP-2, and VEGF. PI3K, p-AKT, and mTOR protein levels also increased, while total AKT protein levels remained unchanged. Conclusions Our study indicated that TIM-1 overexpression promoted cell migration and invasion, and inhibited cell apoptosis in CC through modulation of the PI3K/AKT/p53 and PI3K/AKT/mTOR signaling pathways, and may be a candidate diagnostic biomarker of this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09386-7.
Collapse
Affiliation(s)
- Liuyan Chen
- Joint Inspection Center of Precision Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China.,Department of Clinical Laboratory, the first affiliated hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Jilin Qing
- Center for Reproductive Medicine and Genetics, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China
| | - Yangyang Xiao
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, People's Republic of China
| | - Xiaomei Huang
- Joint Inspection Center of Precision Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China.,Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Yanlin Chi
- Department of Clinical Laboratory, the first affiliated hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Zhizhong Chen
- Joint Inspection Center of Precision Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
11
|
Ferguson CJ, Urso O, Bodrug T, Gassaway BM, Watson ER, Prabu JR, Lara-Gonzalez P, Martinez-Chacin RC, Wu DY, Brigatti KW, Puffenberger EG, Taylor CM, Haas-Givler B, Jinks RN, Strauss KA, Desai A, Gabel HW, Gygi SP, Schulman BA, Brown NG, Bonni A. APC7 mediates ubiquitin signaling in constitutive heterochromatin in the developing mammalian brain. Mol Cell 2022; 82:90-105.e13. [PMID: 34942119 PMCID: PMC8741739 DOI: 10.1016/j.molcel.2021.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 10/14/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022]
Abstract
Neurodevelopmental cognitive disorders provide insights into mechanisms of human brain development. Here, we report an intellectual disability syndrome caused by the loss of APC7, a core component of the E3 ubiquitin ligase anaphase promoting complex (APC). In mechanistic studies, we uncover a critical role for APC7 during the recruitment and ubiquitination of APC substrates. In proteomics analyses of the brain from mice harboring the patient-specific APC7 mutation, we identify the chromatin-associated protein Ki-67 as an APC7-dependent substrate of the APC in neurons. Conditional knockout of the APC coactivator protein Cdh1, but not Cdc20, leads to the accumulation of Ki-67 protein in neurons in vivo, suggesting that APC7 is required for the function of Cdh1-APC in the brain. Deregulated neuronal Ki-67 upon APC7 loss localizes predominantly to constitutive heterochromatin. Our findings define an essential function for APC7 and Cdh1-APC in neuronal heterochromatin regulation, with implications for understanding human brain development and disease.
Collapse
Affiliation(s)
- Cole J Ferguson
- Department of Neuroscience, Washington University, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Neuropathology Division, Physician-Scientist Training Program, Washington University, St. Louis, MO 63110, USA
| | - Olivia Urso
- Department of Neuroscience, Washington University, St. Louis, MO 63110, USA
| | - Tatyana Bodrug
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | - Pablo Lara-Gonzalez
- Department of Cellular and Molecular Medicine, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Raquel C Martinez-Chacin
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Dennis Y Wu
- Department of Neuroscience, Washington University, St. Louis, MO 63110, USA
| | | | | | - Cora M Taylor
- Geisinger Autism & Developmental Medicine Institute, Lewisburg, PA 17837, USA
| | - Barbara Haas-Givler
- Geisinger Autism & Developmental Medicine Institute, Lewisburg, PA 17837, USA
| | - Robert N Jinks
- Department of Biology, Franklin and Marshall College, Lancaster, PA 17603, USA
| | | | - Arshad Desai
- Department of Cellular and Molecular Medicine, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Harrison W Gabel
- Department of Neuroscience, Washington University, St. Louis, MO 63110, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard University, Boston, MA 02138, USA
| | | | - Nicholas G Brown
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Azad Bonni
- Department of Neuroscience, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
12
|
Matsumura Y, Ito R, Yajima A, Yamaguchi R, Tanaka T, Kawamura T, Magoori K, Abe Y, Uchida A, Yoneshiro T, Hirakawa H, Zhang J, Arai M, Yang C, Yang G, Takahashi H, Fujihashi H, Nakaki R, Yamamoto S, Ota S, Tsutsumi S, Inoue SI, Kimura H, Wada Y, Kodama T, Inagaki T, Osborne TF, Aburatani H, Node K, Sakai J. Spatiotemporal dynamics of SETD5-containing NCoR-HDAC3 complex determines enhancer activation for adipogenesis. Nat Commun 2021; 12:7045. [PMID: 34857762 PMCID: PMC8639990 DOI: 10.1038/s41467-021-27321-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023] Open
Abstract
Enhancer activation is essential for cell-type specific gene expression during cellular differentiation, however, how enhancers transition from a hypoacetylated "primed" state to a hyperacetylated-active state is incompletely understood. Here, we show SET domain-containing 5 (SETD5) forms a complex with NCoR-HDAC3 co-repressor that prevents histone acetylation of enhancers for two master adipogenic regulatory genes Cebpa and Pparg early during adipogenesis. The loss of SETD5 from the complex is followed by enhancer hyperacetylation. SETD5 protein levels were transiently increased and rapidly degraded prior to enhancer activation providing a mechanism for the loss of SETD5 during the transition. We show that induction of the CDC20 co-activator of the ubiquitin ligase leads to APC/C mediated degradation of SETD5 during the transition and this operates as a molecular switch that facilitates adipogenesis.
Collapse
Affiliation(s)
- Yoshihiro Matsumura
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| | - Ryo Ito
- grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ayumu Yajima
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ,grid.412339.e0000 0001 1172 4459Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Rei Yamaguchi
- grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshiya Tanaka
- grid.26999.3d0000 0001 2151 536XDepartment of Nuclear Receptor Medicine, Laboratories for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Takeshi Kawamura
- grid.26999.3d0000 0001 2151 536XIsotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Kenta Magoori
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yohei Abe
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Aoi Uchida
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Takeshi Yoneshiro
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Hirakawa
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ,grid.265073.50000 0001 1014 9130Department of Physiology and Cell Biology, Tokyo Medical and Dental University (TMDU), Graduate School, Tokyo, Japan
| | - Ji Zhang
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ,grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Makoto Arai
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ,grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chaoran Yang
- grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ge Yang
- grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroki Takahashi
- grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hitomi Fujihashi
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Ryo Nakaki
- grid.26999.3d0000 0001 2151 536XGenome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ,Rhelixa Inc, Tokyo, Japan
| | - Shogo Yamamoto
- grid.26999.3d0000 0001 2151 536XGenome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Satoshi Ota
- grid.26999.3d0000 0001 2151 536XGenome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shuichi Tsutsumi
- grid.26999.3d0000 0001 2151 536XGenome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shin-ichi Inoue
- grid.69566.3a0000 0001 2248 6943Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Kimura
- grid.32197.3e0000 0001 2179 2105Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Youichiro Wada
- grid.26999.3d0000 0001 2151 536XIsotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- grid.26999.3d0000 0001 2151 536XDepartment of Nuclear Receptor Medicine, Laboratories for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Takeshi Inagaki
- grid.26999.3d0000 0001 2151 536XDivision of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ,grid.256642.10000 0000 9269 4097Laboratory of Epigenetics and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Timothy F. Osborne
- grid.21107.350000 0001 2171 9311Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, and Medicine in the Division of Endocrinology, Diabetes and Metabolism of the Johns Hopkins University School of Medicine, Petersburg, FL USA
| | - Hiroyuki Aburatani
- grid.26999.3d0000 0001 2151 536XGenome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Koichi Node
- grid.412339.e0000 0001 1172 4459Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Juro Sakai
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan. .,Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
13
|
Lambhate S, Bhattacharjee D, Jain N. APC/C CDH1 ubiquitinates IDH2 contributing to ROS increase in mitosis. Cell Signal 2021; 86:110087. [PMID: 34271087 DOI: 10.1016/j.cellsig.2021.110087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/15/2022]
Abstract
NADPH is a cofactor used by reactive oxygen species (ROS) scavenging enzymes to block ROS produced in cells. Recently, it was shown that in cancer cells, ROS progressively increases in tune to cell cycle leading to a peak in mitosis. Loss of IDH2 is known to cause severe oxidative stress in cell and mouse models as ROS increases in mitochondria. Therefore, we hypothesized that IDH2, a major NADPH-producing enzyme in mitochondria is ubiquitinated for ROS to increase in mitosis. To test this hypothesis, in cancer cells we examined IDH2 ubiquitination in mitosis and measured the ROS produced. We found that IDH2 is ubiquitinated in mitosis and on inhibiting anaphase-promoting complex/Cyclosome (APC/C) IDH2 was stabilized. Further, we observed that overexpressing APC/C coactivator CDH1 decreased IDH2, whereas depleting CDH1 decreased IDH2 ubiquitination. To understand the link between IDH2 ubiquitination and ROS produced in mitosis, we show that overexpressing mitochondria-targeted-IDH1 decreased ROS by increasing NADPH in IDH2 ubiquitinated cells. We conclude that APC/C CDH1 ubiquitinates IDH2, a major NADPH-producing enzyme in mitochondria contributing to ROS increase in mitosis. Based on our results, we suggest that mitosis can be a therapeutic window in mutant IDH2-linked pathologies.
Collapse
Affiliation(s)
- Surbhi Lambhate
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debanjan Bhattacharjee
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nishant Jain
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
14
|
Pokrass MJ, Ryan KA, Xin T, Pielstick B, Timp W, Greco V, Regot S. Cell-Cycle-Dependent ERK Signaling Dynamics Direct Fate Specification in the Mammalian Preimplantation Embryo. Dev Cell 2020; 55:328-340.e5. [PMID: 33091369 PMCID: PMC7658051 DOI: 10.1016/j.devcel.2020.09.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/12/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Despite the noisy nature of single cells, multicellular organisms robustly generate different cell types from one zygote. This process involves dynamic cross regulation between signaling and gene expression that is difficult to capture with fixed-cell approaches. To study signaling dynamics and fate specification during preimplantation development, we generated a transgenic mouse expressing the ERK kinase translocation reporter and measured ERK activity in single cells of live embryos. Our results show primarily active ERK in both the inner cell mass and trophectoderm cells due to fibroblast growth factor (FGF) signaling. Strikingly, a subset of mitotic events results in a short pulse of ERK inactivity in both daughter cells that correlates with elevated endpoint NANOG levels. Moreover, endogenous tagging of Nanog in embryonic stem cells reveals that ERK inhibition promotes enhanced stabilization of NANOG protein after mitosis. Our data show that cell cycle, signaling, and differentiation are coordinated during preimplantation development.
Collapse
Affiliation(s)
- Michael J Pokrass
- Department Molecular Biology and Genetics, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biochemistry, Cellular, and Molecular Biology Graduate Program, Baltimore, MD, USA
| | - Kathleen A Ryan
- Department Molecular Biology and Genetics, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tianchi Xin
- Genetics Department, Yale School of Medicine, New Haven, CT 06520, USA
| | - Brittany Pielstick
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Baltimore, MD, USA; Biomedical Engineering Department, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Winston Timp
- Biomedical Engineering Department, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Valentina Greco
- Genetics Department, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sergi Regot
- Department Molecular Biology and Genetics, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Isc10, an Inhibitor That Links the Anaphase-Promoting Complex to a Meiosis-Specific Mitogen-Activated Protein Kinase. Mol Cell Biol 2020; 40:MCB.00097-20. [PMID: 32423992 DOI: 10.1128/mcb.00097-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/13/2020] [Indexed: 11/20/2022] Open
Abstract
Smk1 is a meiosis-specific mitogen-activated protein kinase (MAPK) in yeast that controls spore differentiation. It is activated by a MAPK binding protein, Ssp2, upon completion of the meiotic divisions. The activation of Smk1 by Ssp2 is positively regulated by a meiosis-specific coactivator of the anaphase promoting complex (APC/C) E3 ubiquitin ligase, Ama1. Here, we identify Isc10 as an inhibitor that links APC/CAma1 to Smk1 activation. Isc10 and Smk1 form an inhibited complex during meiosis I (MI). Ssp2 is produced later in the program, and it forms a ternary complex with Isc10 and Smk1 during MII that is poised for activation. Upon completion of MII, Isc10 is ubiquitylated and degraded in an AMA1-dependent manner, thereby triggering the activation of Smk1 by Ssp2. Mutations that caused Ssp2 to be produced before MII, or isc10Δ mutations, modestly reduced the efficiency of spore differentiation whereas spores were nearly absent in the double mutant. These findings define a pathway that couples spore differentiation to the G0-like phase of the cell cycle.
Collapse
|
16
|
Chen A, Bai L, Zhong K, Shu X, Wang A, Xiao Y, Zhang K, Shen C. APC2
CDH1
negatively regulates agrin signaling by promoting the ubiquitination and proteolytic degradation of DOK7. FASEB J 2020; 34:12009-12023. [PMID: 32687671 DOI: 10.1096/fj.202000485r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/28/2023]
Affiliation(s)
- Aizhong Chen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Lei Bai
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Keke Zhong
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Xiaoqiu Shu
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Ailian Wang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Yatao Xiao
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Kejing Zhang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Chengyong Shen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
- Department of Neurobiology Key Laboratory of Medical Neurobiology of Zhejiang Province School of Medicine Zhejiang University Zhejiang China
| |
Collapse
|
17
|
Kimata Y, Leturcq M, Aradhya R. Emerging roles of metazoan cell cycle regulators as coordinators of the cell cycle and differentiation. FEBS Lett 2020; 594:2061-2083. [PMID: 32383482 DOI: 10.1002/1873-3468.13805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023]
Abstract
In multicellular organisms, cell proliferation must be tightly coordinated with other developmental processes to form functional tissues and organs. Despite significant advances in our understanding of how the cell cycle is controlled by conserved cell-cycle regulators (CCRs), how the cell cycle is coordinated with cell differentiation in metazoan organisms and how CCRs contribute to this process remain poorly understood. Here, we review the emerging roles of metazoan CCRs as intracellular proliferation-differentiation coordinators in multicellular organisms. We illustrate how major CCRs regulate cellular events that are required for cell fate acquisition and subsequent differentiation. To this end, CCRs employ diverse mechanisms, some of which are separable from those underpinning the conventional cell-cycle-regulatory functions of CCRs. By controlling cell-type-specific specification/differentiation processes alongside the progression of the cell cycle, CCRs enable spatiotemporal coupling between differentiation and cell proliferation in various developmental contexts in vivo. We discuss the significance and implications of this underappreciated role of metazoan CCRs for development, disease and evolution.
Collapse
Affiliation(s)
- Yuu Kimata
- School of Life Science and Technology, ShanghaiTech University, China
| | - Maïté Leturcq
- School of Life Science and Technology, ShanghaiTech University, China
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| |
Collapse
|
18
|
A Cdh1-FoxM1-Apc axis controls muscle development and regeneration. Cell Death Dis 2020; 11:180. [PMID: 32152291 PMCID: PMC7062904 DOI: 10.1038/s41419-020-2375-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/18/2022]
Abstract
Forkhead box M1 (FoxM1) transcriptional factor has a principal role in regulating cell proliferation, self-renewal, and tumorigenesis. However, whether FoxM1 regulates endogenous muscle development and regeneration remains unclear. Here we found that loss of FoxM1 in muscle satellite cells (SCs) resulted in muscle atrophy and defective muscle regeneration. FoxM1 functioned as a direct transcription activator of adenomatous polyposis coli (Apc), preventing hyperactivation of wnt/β-catenin signaling during muscle regeneration. FoxM1 overexpression in SCs promoted myogenesis but impaired muscle regeneration as a result of spontaneous activation and exhaustion of SCs by transcriptional regulation of Cyclin B1 (Ccnb1). The E3 ubiquitin ligase Cdh1 (also termed Fzr1) was required for FoxM1 ubiquitylation and subsequent degradation. Loss of Cdh1 promoted quiescent SCs to enter into the cell cycle and the SC pool was depleted by serial muscle injuries. Haploinsufficiency of FoxM1 ameliorated muscle regeneration of Cdh1 knock-out mice. These data demonstrate that the Cdh1–FoxM1–Apc axis functions as a key regulator of muscle development and regeneration.
Collapse
|
19
|
Zhu X, Luo C, Lin K, Bu F, Ye F, Huang C, Luo H, Huang J, Zhu Z. Overexpression of DJ-1 enhances colorectal cancer cell proliferation through the cyclin-D1/MDM2-p53 signaling pathway. Biosci Trends 2020; 14:83-95. [PMID: 32132307 DOI: 10.5582/bst.2019.01272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Emerging evidence indicates that DJ-1 is highly expressed in different cancers. It modulates cancer progression, including cell proliferation, cell apoptosis, invasion, and metastasis. However, its role in colorectal cancer (CRC) remains poorly defined. The current study noted increased DJ-1 expression in CRC tumor tissue and found that its expression was closely related to clinical-pathological features. Similarly, DJ-1 increased in CRC cells (SW480, HT-29, Caco-2, LoVo, HCT116, and SW620), and especially in SW480 and HCT116 cells. Functional analyses indicated that overexpression of DJ-1 promoted CRC cell invasion, migration, and proliferation in vitro and in vivo. Mechanistic studies indicated that DJ-1 increased in CRC cell lines, activated specific protein cyclin-D1, and modulated the MDM2/p53 signaling pathway by regulating the levels of the downstream factors Bax, Caspase-3, and Bcl-2, which are related to the cell cycle and apoptosis. Conversely, knockdown of DJ-1 upregulated p53 expression by disrupting the interaction between p53 and MDM2 and inhibiting CRC cell proliferation, revealing the pro-oncogenic mechanism of DJ-1 in CRC. In conclusion, the current findings provide compelling evidence that DJ-1 might be a promoter of CRC cell invasion, proliferation, and migration via the cyclin-D1/MDM2-p53 signaling pathway. Findings also suggest its potential role as a postoperative adjuvant therapy for patients with CRC.
Collapse
Affiliation(s)
- Xiaojian Zhu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Chen Luo
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Kang Lin
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Fanqin Bu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Fan Ye
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Chao Huang
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Huang
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Zhengming Zhu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
20
|
Abstract
Bacteria participate in a wide diversity of symbiotic associations with eukaryotic hosts that require precise interactions for bacterial recognition and persistence. Most commonly, host-associated bacteria interfere with host gene expression to modulate the immune response to the infection. However, many of these bacteria also interfere with host cellular differentiation pathways to create a hospitable niche, resulting in the formation of novel cell types, tissues, and organs. In both of these situations, bacterial symbionts must interact with eukaryotic regulatory pathways. Here, we detail what is known about how bacterial symbionts, from pathogens to mutualists, control host cellular differentiation across the central dogma, from epigenetic chromatin modifications, to transcription and mRNA processing, to translation and protein modifications. We identify four main trends from this survey. First, mechanisms for controlling host gene expression appear to evolve from symbionts co-opting cross-talk between host signaling pathways. Second, symbiont regulatory capacity is constrained by the processes that drive reductive genome evolution in host-associated bacteria. Third, the regulatory mechanisms symbionts exhibit correlate with the cost/benefit nature of the association. And, fourth, symbiont mechanisms for interacting with host genetic regulatory elements are not bound by native bacterial capabilities. Using this knowledge, we explore how the ubiquitous intracellular Wolbachia symbiont of arthropods and nematodes may modulate host cellular differentiation to manipulate host reproduction. Our survey of the literature on how infection alters gene expression in Wolbachia and its hosts revealed that, despite their intermediate-sized genomes, different strains appear capable of a wide diversity of regulatory manipulations. Given this and Wolbachia's diversity of phenotypes and eukaryotic-like proteins, we expect that many symbiont-induced host differentiation mechanisms will be discovered in this system.
Collapse
Affiliation(s)
- Shelbi L Russell
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, CA, USA.
| | | |
Collapse
|
21
|
Rodríguez C, Sánchez-Morán I, Álvarez S, Tirado P, Fernández-Mayoralas DM, Calleja-Pérez B, Almeida Á, Fernández-Jaén A. A novel human Cdh1 mutation impairs anaphase promoting complex/cyclosome activity resulting in microcephaly, psychomotor retardation, and epilepsy. J Neurochem 2019; 151:103-115. [PMID: 31318984 PMCID: PMC6851713 DOI: 10.1111/jnc.14828] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 01/24/2023]
Abstract
The Fizzy-related protein 1 (Fzr1) gene encodes Cdh1 protein, a coactivator of the E3 ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C). Previously, we found that genetic ablation of Fzr1 promotes the death of neural progenitor cells leading to neurogenesis impairment and microcephaly in mouse. To ascertain the possible translation of these findings in humans, we searched for mutations in the Fzr1 gene in 390 whole exomes sequenced in trio in individuals showing neurodevelopmental disorders compatible with a genetic origin. We found a novel missense (p.Asp187Gly) Fzr1 gene mutation (c.560A>G) in a heterozygous state in a 4-year-old boy, born from non-consanguineous Spanish parents, who presents with severe antenatal microcephaly, psychomotor retardation, and refractory epilepsy. Cdh1 protein levels in leucocytes isolated from the patient were significantly lower than those found in his parents. Expression of the Asp187Gly mutant form of Cdh1 in human embryonic kidney 293T cells produced less Cdh1 protein and APC/C activity, resulting in altered cell cycle distribution when compared with cells expressing wild-type Cdh1. Furthermore, ectopic expression of the Asp187Gly mutant form of Cdh1 in cortical progenitor cells in primary culture failed to abolish the enlargement of the replicative phase caused by knockout of endogenous Cdh1. These results indicate that the loss of function of APC/C-Cdh1 caused by Cdh1 Asp187Gly mutation is a new cause of prenatal microcephaly, psychomotor retardation, and severe epilepsy. Read the Editorial Highlight for this article on page 8. Cover Image for this issue: doi: 10.1111/jnc.14524.
Collapse
Affiliation(s)
- Cristina Rodríguez
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, CSIC, Universidad de Salamanca, Salamanca, Spain.,Instituto de Biología Funcional y Genómica, CSIC, Universidad de Salamanca, Salamanca, Spain
| | - Irene Sánchez-Morán
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, CSIC, Universidad de Salamanca, Salamanca, Spain.,Instituto de Biología Funcional y Genómica, CSIC, Universidad de Salamanca, Salamanca, Spain
| | | | - Pilar Tirado
- Departamento de Neuropediatría, Hospital Universitario La Paz, Madrid, Spain
| | - Daniel M Fernández-Mayoralas
- Departamento de Neurología Infantil, Hospital Universitario Quirónsalud, Universidad Europea de Madrid, Madrid, Spain
| | - Beatriz Calleja-Pérez
- Centro de Salud Doctor Cirajas, Servicio de Atención Primaria de Salud, Madrid, Spain
| | - Ángeles Almeida
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, CSIC, Universidad de Salamanca, Salamanca, Spain.,Instituto de Biología Funcional y Genómica, CSIC, Universidad de Salamanca, Salamanca, Spain
| | - Alberto Fernández-Jaén
- Departamento de Neurología Infantil, Hospital Universitario Quirónsalud, Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|