1
|
Jiang H, Qin H, Yang Q, Huang L, Liang X, Wang C, Moro A, Xu S, Wei Q. Effective delivery of miR-150-5p with nucleus pulposus cell-specific nanoparticles attenuates intervertebral disc degeneration. J Nanobiotechnology 2024; 22:292. [PMID: 38802882 PMCID: PMC11129471 DOI: 10.1186/s12951-024-02561-x] [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: 02/15/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The use of gene therapy to deliver microRNAs (miRNAs) has gradually translated to preclinical application for the treatment of intervertebral disc degeneration (IDD). However, the effects of miRNAs are hindered by the short half-life time and the poor cellular uptake, owing to the lack of efficient delivery systems. Here, we investigated nucleus pulposus cell (NPC) specific aptamer-decorated polymeric nanoparticles that can load miR-150-5p for IDD treatment. METHODS The role of miR-150-5p during disc development and degeneration was examined by miR-150-5p knockout (KO) mice. Histological analysis was undertaken in disc specimens. The functional mechanism of miR-150-5p in IDD development was investigated by qRT-PCR assay, Western blot, coimmunoprecipitation and immunofluorescence. NPC specific aptamer-decorated nanoparticles was designed, and its penetration, stability and safety were evaluated. IDD progression was assessed by radiological analysis including X-ray and MRI, after the annulus fibrosus needle puncture surgery with miR-150-5p manipulation by intradiscal injection of nanoparticles. The investigations into the interaction between aptamer and receptor were conducted using mass spectrometry, molecular docking and molecular dynamics simulations. RESULTS We investigated NPC-specific aptamer-decorated polymeric nanoparticles that can bind to miR-150-5p for IDD treatment. Furthermore, we detected that nanoparticle-loaded miR-150-5p inhibitors alleviated NPC senescence in vitro, and the effects of the nanoparticles were sustained for more than 3 months in vivo. The microenvironment of NPCs improves the endo/lysosomal escape of miRNAs, greatly inhibiting the secretion of senescence-associated factors and the subsequent degeneration of NPCs. Importantly, nanoparticles delivering miR-150-5p inhibitors attenuated needle puncture-induced IDD in mouse models by targeting FBXW11 and inhibiting TAK1 ubiquitination, resulting in the downregulation of NF-kB signaling pathway activity. CONCLUSIONS NPC-targeting nanoparticles delivering miR-150-5p show favorable therapeutic efficacy and safety and may constitute a promising treatment for IDD.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Hongyu Qin
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qinghua Yang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Longao Huang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao Liang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Congyang Wang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Abu Moro
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Sheng Xu
- Research Centre for Regenerative Medicine, Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qingjun Wei
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
2
|
Yang Q, Cao Q, Yu Y, Lai X, Feng J, Li X, Jiang Y, Sun Y, Zhou ZW, Li X. Epigenetic and transcriptional landscapes during cerebral cortex development in a microcephaly mouse model. J Genet Genomics 2024; 51:419-432. [PMID: 37923173 DOI: 10.1016/j.jgg.2023.10.006] [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: 07/17/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
The cerebral cortex is a pivotal structure integral to advanced brain functions within the mammalian central nervous system. DNA methylation and hydroxymethylation play important roles in regulating cerebral cortex development. However, it remains unclear whether abnormal cerebral cortex development, such as microcephaly, could rescale the epigenetic landscape, potentially contributing to dysregulated gene expression during brain development. In this study, we characterize and compare the DNA methylome/hydroxymethylome and transcriptome profiles of the cerebral cortex across several developmental stages in wild-type (WT) mice and Mcph1 knockout (Mcph1-del) mice with severe microcephaly. Intriguingly, we discover a global reduction of 5'-hydroxymethylcytosine (5hmC) level, primarily in TET1-binding regions, in Mcph1-del mice compared to WT mice during juvenile and adult stages. Notably, genes exhibiting diminished 5hmC levels and concurrently decreased expression are essential for neurodevelopment and brain functions. Additionally, genes displaying a delayed accumulation of 5hmC in Mcph1-del mice are significantly associated with the establishment and maintenance of the nervous system during the adult stage. These findings reveal that aberrant cerebral cortex development in the early stages profoundly alters the epigenetic regulation program, which provides unique insights into the molecular mechanisms underpinning diseases related to cerebral cortex development.
Collapse
Affiliation(s)
- Qing Yang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Qiang Cao
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yue Yu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xianxin Lai
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jiahao Feng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xinjie Li
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yinan Jiang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yazhou Sun
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhong-Wei Zhou
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Xin Li
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
3
|
Oluwole OG. The analyses of human MCPH1 DNA repair machinery and genetic variations. Open Med (Wars) 2024; 19:20240917. [PMID: 38463519 PMCID: PMC10921449 DOI: 10.1515/med-2024-0917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 10/29/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Causal mutations in the MCPH1 gene have been associated with disorders like microcephaly, and recently congenital hearing impairment. This study examined the MCPH1 DNA repair machinery and identified genetic variations of interest in gnomAD database to discuss the biological roles and effects of rare variants in MCPH1-related diseases. Notably, MCPH1 coordinates two of the seven known mechanisms of DNA repair which confirmed its roles in neurogenesis and chromatin condensation. A pathogenic missense variant in MCPH1 p.Gly753Arg, and two pathogenic frameshifts MCPH1 p.Asn189LysfsTer15 and p.Cys624Ter identified in this study, already had entries in ClinVar and were associated with microcephaly. A pathogenic frameshift in MCPH1 p.Val10SerfsTer5 with a loss-of-function flag and a pathogenic stop gained p.Ser571Ter variants with ultra-rare allele frequency (MAF ≤ 0.001) were identified but have not been linked to any phenotype. The predicted pathogenic ultra-rare variants identified in this study, warranty phenotypic discovery, and also positioned these variants or nearby deleterious variants candidate for screening in MCPH1-associated rare diseases.
Collapse
Affiliation(s)
- Oluwafemi G Oluwole
- Biomedical Research Centre, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
- Non-communicable Diseases Department, Institute of Primate Research, Nairobi, Kenya
| |
Collapse
|
4
|
Zhang S, You X, Zheng Y, Shen Y, Xiong X, Sun Y. The UBE2C/CDH1/DEPTOR axis is an oncogene and tumor suppressor cascade in lung cancer cells. J Clin Invest 2023; 133:162434. [PMID: 36548081 PMCID: PMC9927933 DOI: 10.1172/jci162434] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Ubiquitin-conjugating enzyme E2C (UBE2C) mediates ubiquitylation chain formation via the K11 linkage. While previous in vitro studies showed that UBE2C plays a growth-promoting role in cancer cell lines, the underlying mechanism remains elusive. Still unknown is whether and how UBE2C plays a promoting role in vivo. Here we report that UBE2C was indeed essential for growth and survival of lung cancer cells harboring Kras mutations, and UBE2C was required for KrasG12D-induced lung tumorigenesis, since Ube2c deletion significantly inhibited tumor formation and extended the lifespan of mice. Mechanistically, KrasG12D induced expression of UBE2C, which coupled with APC/CCDH1 E3 ligase to promote ubiquitylation and degradation of DEPTOR, leading to activation of mTORC signaling. Importantly, DEPTOR levels fluctuated during cell cycle progression in a manner dependent on UBE2C and CDH1, indicating their physiological connection. Finally, Deptor deletion fully rescued the tumor inhibitory effect of Ube2c deletion in the KrasG12D lung tumor model, indicating a causal role of Deptor. Taken together, our study shows that the UBE2C/CDH1/DEPTOR axis forms an oncogene and tumor suppressor cascade that regulates cell cycle progression and autophagy and validates UBE2C an attractive target for lung cancer associated with Kras mutations.
Collapse
Affiliation(s)
- Shizhen Zhang
- Cancer Institute and.,Department of Breast Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital, and
| | - Xiahong You
- Cancer Institute and.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yawen Zheng
- Cancer Institute and.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanwen Shen
- Cancer Institute and.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiufang Xiong
- Cancer Institute and.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Sun
- Cancer Institute and.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China.,Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Downregulation of CDC25C in NPCs Disturbed Cortical Neurogenesis. Int J Mol Sci 2023; 24:ijms24021505. [PMID: 36675024 PMCID: PMC9863197 DOI: 10.3390/ijms24021505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Cell division regulators play a vital role in neural progenitor cell (NPC) proliferation and differentiation. Cell division cycle 25C (CDC25C) is a member of the CDC25 family of phosphatases which positively regulate cell division by activating cyclin-dependent protein kinases (CDKs). However, mice with the Cdc25c gene knocked out were shown to be viable and lacked the apparent phenotype due to genetic compensation by Cdc25a and/or Cdc25b. Here, we investigate the function of Cdc25c in developing rat brains by knocking down Cdc25c in NPCs using in utero electroporation. Our results indicate that Cdc25c plays an essential role in maintaining the proliferative state of NPCs during cortical development. The knockdown of Cdc25c causes early cell cycle exit and the premature differentiation of NPCs. Our study uncovers a novel role of CDC25C in NPC division and cell fate determination. In addition, our study presents a functional approach to studying the role of genes, which elicit genetic compensation with knockout, in cortical neurogenesis by knocking down in vivo.
Collapse
|
6
|
De la Cruz G, Nikolaishvili-Feinberg N, Williams SE, Gershon TR. Processing Neonatal Mouse Brains for Immunohistochemical Analysis: As Required for Including Spindle Orientation Analysis and Analysis of DNA Damage and Apoptosis. Methods Mol Biol 2023; 2583:49-54. [PMID: 36418724 DOI: 10.1007/978-1-0716-2752-5_5] [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] [Indexed: 06/16/2023]
Abstract
Analyzing sections of neonatal mouse brain using immunohistochemistry can inform microcephaly pathogenesis, but obtaining and staining high-quality sections can be challenging. The neonatal brain shows less structural integrity than the adult brain. As a result, embedding technique must be optimized to allow sections without cracks or other anatomic disruptions. Moreover, paraffin embedding, which maximized tissue preservation, can reduce antigenicity of proteins in the embedded tissues. We describe an optimized embedding technique and antigen recovery technique that allows successful sectioning and immunohistochemical staining.
Collapse
Affiliation(s)
- Gabriela De la Cruz
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Translational Pathology Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nana Nikolaishvili-Feinberg
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Translational Pathology Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott E Williams
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Timothy R Gershon
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Wang Y, Zong W, Sun W, Chen C, Wang ZQ, Li T. The Central Domain of MCPH1 Controls Development of the Cerebral Cortex and Gonads in Mice. Cells 2022; 11:cells11172715. [PMID: 36078123 PMCID: PMC9455054 DOI: 10.3390/cells11172715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
MCPH1 is the first gene identified to be responsible for the human autosomal recessive disorder primary microcephaly (MCPH). Mutations in the N-terminal and central domains of MCPH1 are strongly associated with microcephaly in human patients. A recent study showed that the central domain of MCPH1, which is mainly encoded by exon 8, interacts with E3 ligase βTrCP2 and regulates the G2/M transition of the cell cycle. In order to investigate the biological functions of MCPH1’s central domain, we constructed a mouse model that lacked the central domain of MCPH1 by deleting its exon 8 (designated as Mcph1-Δe8). Mcph1-Δe8 mice exhibited a reduced brain size and thinner cortex, likely caused by a compromised self-renewal capacity and premature differentiation of Mcph1-Δe8 neuroprogenitors during corticogenesis. Furthermore, Mcph1-Δe8 mice were sterile because of a loss of germ cells in the testis and ovary. The embryonic fibroblasts of Mcph1-Δe8 mice exhibited premature chromosome condensation (PCC). All of these findings indicate that Mcph1-Δe8 mice are reminiscent of MCPH1 complete knockout mice and Mcph1-ΔBR1 mice. Our study demonstrates that the central domain of MCPH1 represses microcephaly, and is essential for gonad development in mammals.
Collapse
Affiliation(s)
- Yaru Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
| | - Wen Zong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
| | - Wenli Sun
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
| | - Chengyan Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
| | - Zhao-Qi Wang
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), 07745 Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, 07743 Jena, Germany
- Correspondence: (Z.-Q.W.); (T.L.); Tel.: +49-3641-656415 (Z.-Q.W.); +86-532-5863-2368 (T.L.); Fax: +49-3641-656413 (Z.-Q.W.)
| | - Tangliang Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (Z.-Q.W.); (T.L.); Tel.: +49-3641-656415 (Z.-Q.W.); +86-532-5863-2368 (T.L.); Fax: +49-3641-656413 (Z.-Q.W.)
| |
Collapse
|
8
|
Tischer T, Yang J, Barford D. The APC/C targets the Cep152-Cep63 complex at the centrosome to regulate mitotic spindle assembly. J Cell Sci 2022; 135:jcs259273. [PMID: 34878135 PMCID: PMC8917351 DOI: 10.1242/jcs.259273] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/25/2021] [Indexed: 11/20/2022] Open
Abstract
The control of protein abundance is a fundamental regulatory mechanism during mitosis. The anaphase-promoting complex/cyclosome (APC/C) is the main protein ubiquitin ligase responsible for the temporal regulation of mitotic progression. It has been proposed that the APC/C might fulfil other functions, including assembly of the mitotic spindle. Here, we show that the APC/C localizes to centrosomes, the organizers of the eukaryotic microtubule cytoskeleton, specifically during mitosis. Recruitment of the APC/C to spindle poles requires the centrosomal protein Cep152, and we identified Cep152 as both an APC/C interaction partner and an APC/C substrate. Previous studies have shown that Cep152 forms a complex with Cep57 and Cep63. The APC/C-mediated ubiquitylation of Cep152 at the centrosome releases Cep57 from this inhibitory complex and enables its interaction with pericentrin, a critical step in promoting microtubule nucleation. Thus, our study extends the function of the APC/C from being a regulator of mitosis to also acting as a positive governor of spindle assembly. The APC/C thereby integrates control of these two important processes in a temporal manner.
Collapse
Affiliation(s)
- Thomas Tischer
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | | | - David Barford
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
9
|
Kristofova M, Ori A, Wang ZQ. Multifaceted Microcephaly-Related Gene MCPH1. Cells 2022; 11:cells11020275. [PMID: 35053391 PMCID: PMC8774270 DOI: 10.3390/cells11020275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
MCPH1, or BRIT1, is often mutated in human primary microcephaly type 1, a neurodevelopmental disorder characterized by a smaller brain size at birth, due to its dysfunction in regulating the proliferation and self-renewal of neuroprogenitor cells. In the last 20 years or so, genetic and cellular studies have identified MCPH1 as a multifaceted protein in various cellular functions, including DNA damage signaling and repair, the regulation of chromosome condensation, cell-cycle progression, centrosome activity and the metabolism. Yet, genetic and animal model studies have revealed an unpredicted essential function of MPCH1 in gonad development and tumorigenesis, although the underlying mechanism remains elusive. These studies have begun to shed light on the role of MPCH1 in controlling various pathobiological processes of the disorder. Here, we summarize the biological functions of MCPH1, and lessons learnt from cellular and mouse models of MCPH1.
Collapse
Affiliation(s)
- Martina Kristofova
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Alessandro Ori
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Zhao-Qi Wang
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Bachstrasse 18k, 07743 Jena, Germany
- Correspondence: ; Tel.: +49-3641-656415; Fax: +49-3641-656335
| |
Collapse
|
10
|
Gallenga CE, Lonardi M, Pacetti S, Violanti SS, Tassinari P, Di Virgilio F, Tognon M, Perri P. Molecular Mechanisms Related to Oxidative Stress in Retinitis Pigmentosa. Antioxidants (Basel) 2021; 10:antiox10060848. [PMID: 34073310 PMCID: PMC8229325 DOI: 10.3390/antiox10060848] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022] Open
Abstract
Retinitis pigmentosa (RP) is an inherited retinopathy. Nevertheless, non-genetic biological factors play a central role in its pathogenesis and progression, including inflammation, autophagy and oxidative stress. The retina is particularly affected by oxidative stress due to its high metabolic rate and oxygen consumption as well as photosensitizer molecules inside the photoreceptors being constantly subjected to light/oxidative stress, which induces accumulation of ROS in RPE, caused by damaged photoreceptor’s daily recycling. Oxidative DNA damage is a key regulator of microglial activation and photoreceptor degeneration in RP, as well as mutations in endogenous antioxidant pathways involved in DNA repair, oxidative stress protection and activation of antioxidant enzymes (MUTYH, CERKL and GLO1 genes, respectively). Moreover, exposure to oxidative stress alters the expression of micro-RNA (miRNAs) and of long non-codingRNA (lncRNAs), which might be implicated in RP etiopathogenesis and progression, modifying gene expression and cellular response to oxidative stress. The upregulation of the P2X7 receptor (P2X7R) also seems to be involved, causing pro-inflammatory cytokines and ROS release by macrophages and microglia, contributing to neuroinflammatory and neurodegenerative progression in RP. The multiple pathways analysed demonstrate that oxidative microglial activation may trigger the vicious cycle of non-resolved neuroinflammation and degeneration, suggesting that microglia may be a key therapy target of oxidative stress in RP.
Collapse
Affiliation(s)
- Carla Enrica Gallenga
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (C.E.G.); (F.D.V.); (M.T.)
| | - Maria Lonardi
- Department of Specialized Surgery, Section of Ophthalmology, Sant’Anna University Hospital, 44121 Ferrara, Italy; (M.L.); (S.P.); (P.T.)
| | - Sofia Pacetti
- Department of Specialized Surgery, Section of Ophthalmology, Sant’Anna University Hospital, 44121 Ferrara, Italy; (M.L.); (S.P.); (P.T.)
| | - Sara Silvia Violanti
- Department of Head and Neck, Section of Ophthalmology, San Paolo Hospital, 17100 Savona, Italy;
| | - Paolo Tassinari
- Department of Specialized Surgery, Section of Ophthalmology, Sant’Anna University Hospital, 44121 Ferrara, Italy; (M.L.); (S.P.); (P.T.)
| | - Francesco Di Virgilio
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (C.E.G.); (F.D.V.); (M.T.)
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (C.E.G.); (F.D.V.); (M.T.)
| | - Paolo Perri
- Department of Neuroscience and Rehabilitation, Section of Ophthalmology, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| |
Collapse
|
11
|
Zhi Y, Zhou X, Yu J, Yuan L, Zhang H, Ng DCH, Xu Z, Xu D. Pathophysiological Significance of WDR62 and JNK Signaling in Human Diseases. Front Cell Dev Biol 2021; 9:640753. [PMID: 33937237 PMCID: PMC8086514 DOI: 10.3389/fcell.2021.640753] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/29/2021] [Indexed: 12/31/2022] Open
Abstract
The c-Jun N-terminal kinase (JNK) is highly evolutionarily conserved and plays important roles in a broad range of physiological and pathological processes. The WD40-repeat protein 62 (WDR62) is a scaffold protein that recruits different components of the JNK signaling pathway to regulate several human diseases including neurological disorders, infertility, and tumorigenesis. Recent studies revealed that WDR62 regulates the process of neural stem cell mitosis and germ cell meiosis through JNK signaling. In this review we summarize the roles of WDR62 and JNK signaling in neuronal and non-neuronal contexts and discuss how JNK-dependent signaling regulates both processes. WDR62 is involved in various human disorders via JNK signaling regulation, and may represent a promising therapeutic strategy for the treatment of related diseases.
Collapse
Affiliation(s)
- Yiqiang Zhi
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - Xiaokun Zhou
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - Jurui Yu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - Ling Yuan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Hongsheng Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Dominic C H Ng
- Faculty of Medicine, School of Biomedical Science, University of Queensland, St. Lucia, QLD, Australia
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Dan Xu
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| |
Collapse
|
12
|
Marx C, Schaarschmidt MU, Kirkpatrick J, Marx-Blümel L, Halilovic M, Westermann M, Hoelzer D, Meyer FB, Geng Y, Buder K, Schadwinkel HM, Siniuk K, Becker S, Thierbach R, Beck JF, Sonnemann J, Wang ZQ. Cooperative treatment effectiveness of ATR and HSP90 inhibition in Ewing's sarcoma cells. Cell Biosci 2021; 11:57. [PMID: 33743824 PMCID: PMC7981928 DOI: 10.1186/s13578-021-00571-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/12/2021] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Ewing's sarcoma is an aggressive childhood malignancy whose outcome has not substantially improved over the last two decades. In this study, combination treatments of the HSP90 inhibitor AUY922 with either the ATR inhibitor VE821 or the ATM inhibitor KU55933 were investigated for their effectiveness in Ewing's sarcoma cells. METHODS Effects were determined in p53 wild-type and p53 null Ewing's sarcoma cell lines by flow cytometric analyses of cell death, mitochondrial depolarization and cell-cycle distribution as well as fluorescence and transmission electron microscopy. They were molecularly characterized by gene and protein expression profiling, and by quantitative whole proteome analysis. RESULTS AUY922 alone induced DNA damage, apoptosis and ER stress, while reducing the abundance of DNA repair proteins. The combination of AUY922 with VE821 led to strong apoptosis induction independent of the cellular p53 status, yet based on different molecular mechanisms. p53 wild-type cells activated pro-apoptotic gene transcription and underwent mitochondria-mediated apoptosis, while p53 null cells accumulated higher levels of DNA damage, ER stress and autophagy, eventually leading to apoptosis. Impaired PI3K/AKT/mTOR signaling further contributed to the antineoplastic combination effects of AUY922 and VE821. In contrast, the combination of AUY922 with KU55933 did not produce a cooperative effect. CONCLUSION Our study reveals that HSP90 and ATR inhibitor combination treatment may be an effective therapeutic approach for Ewing's sarcoma irrespective of the p53 status.
Collapse
Affiliation(s)
- Christian Marx
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Marc U Schaarschmidt
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany.,Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Joanna Kirkpatrick
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Francis Crick Institute, London, UK
| | - Lisa Marx-Blümel
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany.,Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Melisa Halilovic
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - Doerte Hoelzer
- Department of Human Nutrition, Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany.,Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Felix B Meyer
- Department of Human Nutrition, Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany
| | - Yibo Geng
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Katrin Buder
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hauke M Schadwinkel
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany.,Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Kanstantsin Siniuk
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Sabine Becker
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany.,Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - René Thierbach
- Department of Human Nutrition, Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany
| | - James F Beck
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany
| | - Jürgen Sonnemann
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany. .,Research Center Lobeda, Jena University Hospital, Jena, Germany. .,Klinik Für Kinder- Und Jugendmedizin, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany.
| | - Zhao-Qi Wang
- Leibniz Institute On Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Faculty of Biology and Pharmacy, Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
13
|
Liu X, Schneble-Löhnert N, Kristofova M, Qing X, Labisch J, Hofmann S, Ehrenberg S, Sannai M, Jörß T, Ori A, Godmann M, Wang ZQ. The N-terminal BRCT domain determines MCPH1 function in brain development and fertility. Cell Death Dis 2021; 12:143. [PMID: 33542216 PMCID: PMC7862653 DOI: 10.1038/s41419-021-03406-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 11/27/2022]
Abstract
MCPH1 is a causal gene for the neurodevelopmental disorder, human primary microcephaly (MCPH1, OMIM251200). Most pathogenic mutations are located in the N-terminal region of the gene, which encodes a BRCT domain, suggesting an important function of this domain in brain size determination. To investigate the specific function of the N-terminal BRCT domain in vivo, we generated a mouse model lacking the N’-BRCT domain of MCPH1 (referred as Mcph1-ΔBR1). These mutant mice are viable, but exhibit reduced brain size, with a thinner cortex due to a reduction of neuroprogenitor populations and premature neurogenic differentiation. Mcph1-ΔBR1 mice (both male and female) are infertile; however, almost all female mutants develop ovary tumours. Mcph1-ΔBR1 MEF cells exhibit a defect in DNA damage response and DNA repair, and show the premature chromosome condensation (PCC) phenotype, a hallmark of MCPH1 patient cells and also Mcph1 knockout cells. In comparison with Mcph1 complete knockout mice, Mcph1-ΔBR1 mice faithfully reproduce all phenotypes, indicating an essential role of the N-terminal BRCT domain for the physiological function of MCPH1 in the control of brain size and gonad development as well as in multiple cellular processes.
Collapse
Affiliation(s)
- Xiaoqian Liu
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Nadine Schneble-Löhnert
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Martina Kristofova
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Xiaobing Qing
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Jan Labisch
- Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich-Schiller-University of Jena, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Susanne Hofmann
- Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich-Schiller-University of Jena, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Sandra Ehrenberg
- Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich-Schiller-University of Jena, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Mara Sannai
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Tjard Jörß
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Maren Godmann
- Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich-Schiller-University of Jena, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany. .,Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Beutenbergstrasse 11, 07745, Jena, Germany.
| |
Collapse
|
14
|
Stracker TH, Morrison CG, Gergely F. Molecular causes of primary microcephaly and related diseases: a report from the UNIA Workshop. Chromosoma 2020; 129:115-120. [PMID: 32424716 DOI: 10.1007/s00412-020-00737-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022]
Abstract
The International University of Andalucía (UNIA) Current Trends in Biomedicine Workshop on Molecular Causes of Primary Microcephaly and Related Diseases took place in Baeza, Spain, November 18-20, 2019. This meeting brought together scientists from Europe, the USA and China to discuss recent advances in our molecular and genetic understanding of a group of rare neurodevelopmental diseases characterised by primary microcephaly, a condition in which head circumference is smaller than normal at birth. Microcephaly can be caused by inherited mutations that affect key cellular processes, or environmental exposure to radiation or other toxins. It can also result from viral infection, as exemplified by the recent Zika virus outbreak in South America. Here we summarise a number of the scientific advances presented and topics discussed at the meeting.
Collapse
Affiliation(s)
- Travis H Stracker
- Institute for Research in Biomedicine (IRB Barcelona) and Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.
| | - Ciaran G Morrison
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Biosciences Building, Dangan, Galway, H91 TK33, Ireland
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| |
Collapse
|
15
|
Donato L, Scimone C, Alibrandi S, Nicocia G, Rinaldi C, Sidoti A, D’Angelo R. Discovery of GLO1 New Related Genes and Pathways by RNA-Seq on A2E-Stressed Retinal Epithelial Cells Could Improve Knowledge on Retinitis Pigmentosa. Antioxidants (Basel) 2020; 9:416. [PMID: 32413970 PMCID: PMC7278727 DOI: 10.3390/antiox9050416] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/02/2020] [Accepted: 05/10/2020] [Indexed: 12/15/2022] Open
Abstract
Endogenous antioxidants protect cells from reactive oxygen species (ROS)-related deleterious effects, and an imbalance in the oxidant/antioxidant systems generates oxidative stress. Glyoxalase 1 (GLO1) is a ubiquitous cellular enzyme involved in detoxification of methylglyoxal (MG), a cytotoxic byproduct of glycolysis whose excess can produce oxidative stress. In retinitis pigmentosa, one of the most diffuse cause of blindness, oxidative damage leads to photoreceptor death. To clarify the role of GLO1 in retinitis pigmentosa onset and progression, we treated human retinal pigment epithelium cells by the oxidant agent A2E. Transcriptome profiles between treated and untreated cells were performed by RNA-Seq, considering two time points (3 and 6 h), after the basal one. The exposure to A2E highlighted significant expression differences and splicing events in 370 GLO1 first-neighbor genes, and 23 of them emerged from pathway clustered analysis as main candidates to be associated with retinitis pigmentosa. Such a hypothesis was corroborated by the involvement of previously analyzed genes in specific cellular activities related to oxidative stress, such as glyoxylate and dicarboxylate metabolism, glycolysis, axo-dendritic transport, lipoprotein activity and metabolism, SUMOylation and retrograde transport at the trans-Golgi network. Our findings could be the starting point to explore unclear molecular mechanisms involved in retinitis pigmentosa etiopathogenesis.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (S.A.); (C.R.); (R.D.)
- Department of Biomolecular strategies, genetics and avant-garde therapies, I.E.ME.S.T., 90139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (S.A.); (C.R.); (R.D.)
- Department of Biomolecular strategies, genetics and avant-garde therapies, I.E.ME.S.T., 90139 Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (S.A.); (C.R.); (R.D.)
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98125 Messina, Italy
| | - Giacomo Nicocia
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (S.A.); (C.R.); (R.D.)
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (S.A.); (C.R.); (R.D.)
- Department of Biomolecular strategies, genetics and avant-garde therapies, I.E.ME.S.T., 90139 Palermo, Italy
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (S.A.); (C.R.); (R.D.)
- Department of Biomolecular strategies, genetics and avant-garde therapies, I.E.ME.S.T., 90139 Palermo, Italy
| |
Collapse
|
16
|
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: 6] [Impact Index Per Article: 1.2] [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
|
17
|
Journiac N, Gilabert-Juan J, Cipriani S, Benit P, Liu X, Jacquier S, Faivre V, Delahaye-Duriez A, Csaba Z, Hourcade T, Melinte E, Lebon S, Violle-Poirsier C, Oury JF, Adle-Biassette H, Wang ZQ, Mani S, Rustin P, Gressens P, Nardelli J. Cell Metabolic Alterations due to Mcph1 Mutation in Microcephaly. Cell Rep 2020; 31:107506. [DOI: 10.1016/j.celrep.2020.03.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/21/2019] [Accepted: 03/21/2020] [Indexed: 12/13/2022] Open
|
18
|
Kimata Y. APC/C Ubiquitin Ligase: Coupling Cellular Differentiation to G1/G0 Phase in Multicellular Systems. Trends Cell Biol 2019; 29:591-603. [DOI: 10.1016/j.tcb.2019.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/27/2022]
|
19
|
Meyer SK, Dunn M, Vidler DS, Porter A, Blain PG, Jowsey PA. Phosphorylation of MCPH1 isoforms during mitosis followed by isoform-specific degradation by APC/C-CDH1. FASEB J 2019; 33:2796-2808. [PMID: 30303738 PMCID: PMC6338662 DOI: 10.1096/fj.201801353r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/17/2018] [Indexed: 11/11/2022]
Abstract
Microcephalin-1 (MCPH1) exists as 2 isoforms that regulate cyclin-dependent kinase-1 activation and chromosome condensation during mitosis, with MCPH1 mutations causing primary microcephaly. MCPH1 is also a tumor suppressor protein, with roles in DNA damage repair/checkpoints. Despite these important roles, there is little information on the cellular regulation of MCPH1. We show that both MCPH1 isoforms are phosphorylated in a cyclin-dependent kinase-1-dependent manner in mitosis and identify several novel phosphorylation sites. Upon mitotic exit, MCPH1 isoforms were degraded by the anaphase-promoting complex/cyclosome-CDH1 E3 ligase complex. Anaphase-promoting complex/cyclosome-CDH1 target proteins generally have D-Box or KEN-Box degron sequences. We found that MCPH1 isoforms are degraded independently, with the long isoform degradation being D-Box dependent, whereas the short isoform was KEN-Box dependent. Our research identifies several novel mechanisms regulating MCPH1 and also highlights important issues with several commercial MCPH1 antibodies, with potential relevance to previously published data.-Meyer, S. K., Dunn, M., Vidler, D. S., Porter, A., Blain, P. G., Jowsey, P. A. Phosphorylation of MCPH1 isoforms during mitosis followed by isoform-specific degradation by APC/C-CDH1.
Collapse
Affiliation(s)
- Stephanie K. Meyer
- National Intitute for Health Research (NIHR) Health Protection Research Unit for Chemical and Radiation Threats and Hazards, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| | - Michael Dunn
- National Intitute for Health Research (NIHR) Health Protection Research Unit for Chemical and Radiation Threats and Hazards, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| | - Daniel S. Vidler
- National Intitute for Health Research (NIHR) Health Protection Research Unit for Chemical and Radiation Threats and Hazards, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| | - Andrew Porter
- Newcastle University Protein and Proteome Analysis, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Peter G. Blain
- National Intitute for Health Research (NIHR) Health Protection Research Unit for Chemical and Radiation Threats and Hazards, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| | - Paul A. Jowsey
- National Intitute for Health Research (NIHR) Health Protection Research Unit for Chemical and Radiation Threats and Hazards, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; and
| |
Collapse
|
20
|
Wang D, Xu X, Wu Y, Lin Y, Gao M, Hu P, Chen D, Lu X, Chen Z, Wang H, Huang C. SMIP004: A compound with antidepressant-like activities in mouse models. Eur J Pharmacol 2019; 843:260-267. [DOI: 10.1016/j.ejphar.2018.11.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/23/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022]
|
21
|
Sarkar S, Alam N, Mandal SS, Chatterjee K, Ghosh S, Roychoudhury S, Panda CK. Differential transmission of the molecular signature of RBSP3, LIMD1 and CDC25A in basal/ parabasal versus spinous of normal epithelium during head and neck tumorigenesis: A mechanistic study. PLoS One 2018; 13:e0195937. [PMID: 29672635 PMCID: PMC5909606 DOI: 10.1371/journal.pone.0195937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a global disease and mortality burden, necessitating the elucidation of its molecular progression for effective disease management. The study aims to understand the molecular profile of three candidate cell cycle regulatory genes, RBSP3, LIMD1 and CDC25A in the basal/ parabasal versus spinous layer of normal oral epithelium and during head and neck tumorigenesis. Immunohistochemical expression and promoter methylation was used to determine the molecular signature in normal oral epithelium. The mechanism of alteration transmission of this profile during tumorigenesis was then explored through additional deletion and mutation in HPV/ tobacco etiological groups, followed byclinico-pathological correlation. In basal/parabasal layer, the molecular signature of the genes was low protein expression/ high promoter methylation of RBSP3, high expression/ low methylation of LIMD1 and high expression of CDC25A. Dysplastic epithelium maintained the signature of RBSP3 through high methylation/ additional deletion with loss of the signatures of LIMD1 and CDC25A via deletion/ additional methylation. Similarly, maintenance and / or loss of signature in invasive tumors was by recurrent deletion/ methylation. Thus, differential patterns of alteration of the genes might be pre-requisite for the development of dysplastic and invasive lesions. Etiological factors played a key role in promoting genetic alterations and determining prognosis. Tobacco negative HNSCC patients had significantly lower alterations of LIMD1 and CDC25A, along with better survival among tobacco negative/ HPV positive patients. Our data suggests the necessity for perturbation of normal molecular profile of RBSP3, LIMD1 and CDC25A in conjunction with etiological factors for head and neck tumorigenesis, implying their diagnostic and prognostic significance.
Collapse
Affiliation(s)
- Shreya Sarkar
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Syam Sundar Mandal
- Department of Epidemiology and Biostatistics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Kabita Chatterjee
- Department of Oral and Maxillofacial Pathology, Buddha Institute of Dental Sciences and Hospital, Patna, India
| | - Supratim Ghosh
- Department of Oral and Maxillofacial Pathology, Burdwan Dental College and Hospital, Burdwan, India
| | - Susanta Roychoudhury
- Basic Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
- * E-mail:
| |
Collapse
|
22
|
Liu X, Zong W, Li T, Wang Y, Xu X, Zhou ZW, Wang ZQ. The E3 ubiquitin ligase APC/C Cdh1 degrades MCPH1 after MCPH1-βTrCP2-Cdc25A-mediated mitotic entry to ensure neurogenesis. EMBO J 2017; 36:3666-3681. [PMID: 29150431 DOI: 10.15252/embj.201694443] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/20/2022] Open
Abstract
Mutations of microcephalin (MCPH1) can cause the neurodevelopmental disorder primary microcephaly type 1. We previously showed that MCPH1 deletion in neural stem cells results in early mitotic entry that distracts cell division mode, leading to exhaustion of the progenitor pool. Here, we show that MCPH1 interacts with and promotes the E3 ligase βTrCP2 to degrade Cdc25A independent of DNA damage. Overexpression of βTrCP2 or the knockdown of Cdc25A remedies the high mitotic index and rescues the premature differentiation of Mcph1-deficient neuroprogenitors in vivo MCPH1 itself is degraded by APC/CCdh1, but not APC/CCdc20, in late mitosis and G1 phase. Forced MCPH1 expression causes cell death, underlining the importance of MCPH1 turnover after mitosis. Ectopic expression of Cdh1 leads to premature differentiation of neuroprogenitors, mimicking differentiation defects of Mcph1-knockout neuroprogenitors. The homeostasis of MCPH1 in association with the ubiquitin-proteasome system ensures mitotic entry independent of cell cycle checkpoint. This study provides a mechanistic understanding of how MCPH1 controls neural stem cell fate and brain development.
Collapse
Affiliation(s)
- Xiaoqian Liu
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Wen Zong
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Tangliang Li
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Yujun Wang
- Division of Biology, City of Hope National Medical Center/Beckman Research Institute, Duarte, CA, USA
| | - Xingzhi Xu
- Beijing Key Laboratory of DNA Damage Response, College of Life Sciences, Capital Normal University, Beijing, China.,Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Zhong-Wei Zhou
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany .,Faculty of Biology and Pharmacy, Friedrich-Schiller University of Jena, Jena, Germany
| |
Collapse
|