1
|
Wozna‐Wysocka M, Jazurek‐Ciesiolka M, Przybyl L, Wronka D, Misiorek JO, Suszynska‐Zajczyk J, Figura G, Ciesiolka A, Sobieszczanska P, Zeller A, Niemira M, Switonski PM, Fiszer A. Insights into RNA-mediated pathology in new mouse models of Huntington's disease. FASEB J 2024; 38:e70182. [PMID: 39604147 PMCID: PMC11602643 DOI: 10.1096/fj.202401465r] [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: 06/28/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Huntington's disease (HD) is a neurodegenerative polyglutamine (polyQ) disease resulting from the expansion of CAG repeats located in the ORF of the huntingtin gene (HTT). The extent to which mutant mRNA-driven disruptions contribute to HD pathogenesis, particularly in comparison to the dominant mechanisms related to the gain-of-function effects of the mutant polyQ protein, is still debatable. To evaluate this contribution in vivo, we generated two mouse models through a knock-in strategy at the Rosa26 locus. These models expressed distinct variants of human mutant HTT cDNA fragment: a translated variant (HD/100Q model, serving as a reference) and a nontranslated variant (HD/100CAG model). The cohorts of animals were subjected to a broad spectrum of molecular, behavioral, and cognitive analysis for 21 months. Behavioral testing revealed alterations in both models, with the HD/100Q model exhibiting late disease phenotype. The rotarod, static rod, and open-field tests showed some motor deficits in HD/100CAG and HD/100Q model mice during the light phase, while ActiMot indicated hyperkinesis during the dark phase. Both models also exhibited certain gene deregulations in the striatum that are related to disrupted pathways and phenotype alterations observed in HD. In conclusion, we provide in vivo evidence for a minor contributory role of mutant RNA in HD pathogenesis. The separated effects resulting from the presence of mutant RNA in the HD/100CAG model led to less severe but, to some extent, similar types of impairments as in the HD/100Q model. Increased anxiety was one of the most substantial effects caused by mutant HTT RNA.
Collapse
Affiliation(s)
| | | | - Lukasz Przybyl
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | - Dorota Wronka
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | | | | | - Grzegorz Figura
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Present address:
Department of Bioenergetics, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznanPoland
| | - Adam Ciesiolka
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Present address:
Department of Gene Expression, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznanPoland
| | | | - Anna Zeller
- Genomics and Epigenomics Laboratory, Clinical Research CentreMedical University of BialystokBialystokPoland
| | - Magdalena Niemira
- Genomics and Epigenomics Laboratory, Clinical Research CentreMedical University of BialystokBialystokPoland
| | | | - Agnieszka Fiszer
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| |
Collapse
|
2
|
Abstract
Evidence implicating Eph receptor tyrosine kinases and their ephrin ligands (that together make up the 'Eph system') in cancer development and progression has been accumulating since the discovery of the first Eph receptor approximately 35 years ago. Advances in the past decade and a half have considerably increased the understanding of Eph receptor-ephrin signalling mechanisms in cancer and have uncovered intriguing new roles in cancer progression and drug resistance. This Review focuses mainly on these more recent developments. I provide an update on the different mechanisms of Eph receptor-ephrin-mediated cell-cell communication and cell autonomous signalling, as well as on the interplay of the Eph system with other signalling systems. I further discuss recent advances in elucidating how the Eph system controls tumour expansion, invasiveness and metastasis, supports cancer stem cells, and drives therapy resistance. In addition to functioning within cancer cells, the Eph system also mediates the reciprocal communication between cancer cells and cells of the tumour microenvironment. The involvement of the Eph system in tumour angiogenesis is well established, but recent findings also demonstrate roles in immune cells, cancer-associated fibroblasts and the extracellular matrix. Lastly, I discuss strategies under evaluation for therapeutic targeting of Eph receptors-ephrins in cancer and conclude with an outlook on promising future research directions.
Collapse
Affiliation(s)
- Elena B Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
3
|
Cai C, Zhang M, Liu L, Zhang H, Guo Y, Lan T, Xu Y, Ma P, Li S. ADAM10-cleaved ephrin-A5 contributes to prostate cancer metastasis. Cell Death Dis 2022; 13:453. [PMID: 35551177 PMCID: PMC9098485 DOI: 10.1038/s41419-022-04893-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/27/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022]
Abstract
A disintegrin and metalloprotease-10(ADAM10) promotes the metastasis of prostate cancer (PCa), but the specific mechanism is indistinct. Herein, DU145 cell lines with stable overexpression and knockdown of ADAM10 were constructed. We found that ectopic expression of ADAM10 not only significantly facilitated cell proliferation, migration, invasion, and inhibited apoptosis, but also could specifically hydrolyze ephrin-A5 and release the ephrin-A5 soluble ectodomain into extracellular media in vitro. These effects were reversed by ADAM10 depletion or treatment of GI254023X. Meanwhile, the co-location and physical interaction among EphA3, ephrin-A5, and ADAM10 were observed in PCa cells using immunofluorescence and immunoprecipitation techniques. Interestingly, overexpression of EphA3 exerted opposite effects in DU145 (ephrin-A5 + ) cells and PC-3 (ephrin-A5 ± ) cells. In addition, the pro-tumor function of EphA3 was reversed by the treatment with the exogenous ephrin-A5-Fc, which increased the phosphorylation level of EphA3 in PC-3 (ephrin-A5 ± ) cells. In nude mice, ADAM10 accelerated growth of the primary tumor, decreased the level of ephrin-A5 in the tumor tissue, but increased the level of ephrin-A5 in the peripheral blood, accompanied with an increase in the expression of CD31 and VEGF (vascular endothelial growth factor) in the tissue. What is more, the serum ephrin-A5 content of patients with metastatic PCa was significantly higher than that of the non-metastatic group (P < 0.05). The receiver operating characteristic curve(ROC) showed that the area under the curve(AUC) of serum ephrin-A5 as a marker of PCa metastasis was 0.843, with a sensitivity of 93.5% and a specificity of 75%. It is concluded that ADAM10-mediated ephrin-A5 shedding promotes PCa metastasis via transforming the role of EphA3 from ligand-dependent tumor suppressor to ligand-independent promoter, and ephrin-A5 in the blood can be used as a new biomarker for PCa metastasis.
Collapse
Affiliation(s)
- Chenchen Cai
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.452207.60000 0004 1758 0558Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, 221009 China
| | - Miaomiao Zhang
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Lei Liu
- grid.417303.20000 0000 9927 0537Department of Physiology, Xuzhou Medical University, Xuzhou, 221004 PR China
| | - Haoliang Zhang
- grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Yi Guo
- grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Ting Lan
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China
| | - Yinhai Xu
- grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Ping Ma
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Shibao Li
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| |
Collapse
|
4
|
Cirnaru MD, Song S, Tshilenge KT, Corwin C, Mleczko J, Galicia Aguirre C, Benlhabib H, Bendl J, Apontes P, Fullard J, Creus-Muncunill J, Reyahi A, Nik AM, Carlsson P, Roussos P, Mooney SD, Ellerby LM, Ehrlich ME. Unbiased identification of novel transcription factors in striatal compartmentation and striosome maturation. eLife 2021; 10:e65979. [PMID: 34609283 PMCID: PMC8492065 DOI: 10.7554/elife.65979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
Many diseases are linked to dysregulation of the striatum. Striatal function depends on neuronal compartmentation into striosomes and matrix. Striatal projection neurons are GABAergic medium spiny neurons (MSNs), subtyped by selective expression of receptors, neuropeptides, and other gene families. Neurogenesis of the striosome and matrix occurs in separate waves, but the factors regulating compartmentation and neuronal differentiation are largely unidentified. We performed RNA- and ATAC-seq on sorted striosome and matrix cells at postnatal day 3, using the Nr4a1-EGFP striosome reporter mouse. Focusing on the striosome, we validated the localization and/or role of Irx1, Foxf2, Olig2, and Stat1/2 in the developing striosome and the in vivo enhancer function of a striosome-specific open chromatin region 4.4 Kb downstream of Olig2. These data provide novel tools to dissect and manipulate the networks regulating MSN compartmentation and differentiation, including in human iPSC-derived striatal neurons for disease modeling and drug discovery.
Collapse
Affiliation(s)
- Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Sicheng Song
- Department of Biomedical Informatics and Medical Education, University of WashingtonSeattleUnited States
| | | | - Chuhyon Corwin
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Justyna Mleczko
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | | | - Houda Benlhabib
- Department of Biomedical Informatics and Medical Education, University of WashingtonSeattleUnited States
| | - Jaroslav Bendl
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Pasha Apontes
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - John Fullard
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Azadeh Reyahi
- Department of Chemistry and Molecular Biology, University of GothenburgGothenburgSweden
| | - Ali M Nik
- Department of Chemistry and Molecular Biology, University of GothenburgGothenburgSweden
| | - Peter Carlsson
- Department of Chemistry and Molecular Biology, University of GothenburgGothenburgSweden
| | - Panos Roussos
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Mental Illness Research, Education, and Clinical Center (VISN 2 South)BronxUnited States
| | - Sean D Mooney
- Department of Biomedical Informatics and Medical Education, University of WashingtonSeattleUnited States
| | | | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
5
|
Baudet S, Bécret J, Nicol X. Approaches to Manipulate Ephrin-A:EphA Forward Signaling Pathway. Pharmaceuticals (Basel) 2020; 13:ph13070140. [PMID: 32629797 PMCID: PMC7407804 DOI: 10.3390/ph13070140] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/19/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin-producing hepatocellular carcinoma A (EphA) receptors and their ephrin-A ligands are key players of developmental events shaping the mature organism. Their expression is mostly restricted to stem cell niches in adults but is reactivated in pathological conditions including lesions in the heart, lung, or nervous system. They are also often misregulated in tumors. A wide range of molecular tools enabling the manipulation of the ephrin-A:EphA system are available, ranging from small molecules to peptides and genetically-encoded strategies. Their mechanism is either direct, targeting EphA receptors, or indirect through the modification of intracellular downstream pathways. Approaches enabling manipulation of ephrin-A:EphA forward signaling for the dissection of its signaling cascade, the investigation of its physiological roles or the development of therapeutic strategies are summarized here.
Collapse
|
6
|
Zfp422 promotes skeletal muscle differentiation by regulating EphA7 to induce appropriate myoblast apoptosis. Cell Death Differ 2019; 27:1644-1659. [PMID: 31685980 PMCID: PMC7206035 DOI: 10.1038/s41418-019-0448-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022] Open
Abstract
Zinc finger protein 422 (Zfp422) is a widely expressed zinc finger protein that serves as a transcriptional factor to regulate downstream gene expression, but until now, little is known about its roles in myogenesis. We found here that Zfp422 plays a critical role in skeletal muscle development and regeneration. It highly expresses in mouse skeletal muscle during embryonic development. Specific knockout of Zfp422 in skeletal muscle impaired embryonic muscle formation. Satellite cell-specific Zfp422 deletion severely inhibited muscle regeneration. Myoblast differentiation and myotube formation were suppressed in Zfp422-deleted C2C12 cells, isolated primary myoblasts, and satellite cells. Chromatin Immunoprecipitation Sequencing (ChIP-Seq) revealed that Zfp422 regulated ephrin type-A receptor 7 (EphA7) expression by binding an upstream 169-bp DNA sequence, which was proved to be an enhancer of EphA7. Knocking EphA7 down in C2C12 cells or deleting Zfp422 in myoblasts will inhibit cell apoptosis which is required for myoblast differentiation. These results indicate that Zfp422 is essential for skeletal muscle differentiation and fusion, through regulating EphA7 expression to maintain proper apoptosis.
Collapse
|
7
|
Robbins CJ, Bou-Dargham MJ, Sanchez K, Rosen MC, Sang QXA. Decoding Somatic Driver Gene Mutations and Affected Signaling Pathways in Human Medulloblastoma Subgroups. J Cancer 2018; 9:4596-4610. [PMID: 30588243 PMCID: PMC6299398 DOI: 10.7150/jca.27993] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/08/2018] [Indexed: 01/02/2023] Open
Abstract
Medulloblastoma is the most common malignant pediatric brain tumor. Prior studies have concentrated their efforts studying the four molecular subgroups: SHH, Wnt, group 3, and group 4. SHH and Wnt are driven by their canonical pathways. Groups 3 and 4 are highly metastatic and associated with aberrations in epigenetic regulators. Recent developments in the field have revealed that these subgroups are not as homogenous as previously believed. The objective of this study is to investigate the involvement of somatic driver gene mutations in these medulloblastoma subgroups. We obtained medulloblastoma data from the Catalogue of Somatic Mutations in Cancer (COSMIC), which contains distinct samples that were not previously studied in a large cohort. We identified somatic driver gene mutations and the signaling pathways affected by these driver genes for medulloblastoma subgroups using bioinformatics tools. We have revealed novel infrequent drivers in these subgroups that contribute to our understanding of tumor heterogeneity in medulloblastoma. Normally SHH signaling is activated in the SHH subgroup, however, we determined gain-of-function mutations in ubiquitin ligase (CUL1) that inhibit Gli-mediated transcription. This suggests a potential hindrance in SHH signaling for some patients. For group 3, gain-of-function in the inhibitor of proinflammatory cytokines (HIVEP3) suggests an immunosuppressive phenotype and thus a more hostile tumor microenvironment. Surprisingly, group 4 tumors possess mutations that may prompt the activation of Wnt signaling through gain-of-function mutations in MUC16 and PCDH9. These infrequent mutations detected in this study could be due to subclonal or spatially restricted alterations. The investigation of aberrant driver gene mutations can lead to the identification of new drug targets and a greater understanding of human medulloblastoma heterogeneity.
Collapse
Affiliation(s)
- Charles J Robbins
- Department of Chemistry & Biochemistry, Institute of Molecular Biophysics, Florida State University
| | - Mayassa J Bou-Dargham
- Department of Chemistry & Biochemistry, Institute of Molecular Biophysics, Florida State University
| | - Kevin Sanchez
- Department of Chemistry & Biochemistry, Institute of Molecular Biophysics, Florida State University
| | - Matthew C Rosen
- Department of Chemistry & Biochemistry, Institute of Molecular Biophysics, Florida State University
| | - Qing-Xiang Amy Sang
- Department of Chemistry & Biochemistry, Institute of Molecular Biophysics, Florida State University
| |
Collapse
|
8
|
Li S, Zhai J, Liu J, Di F, Sun Y, Li W, Chen ZJ, Du Y. Erythropoietin-producing hepatocellular A7 triggering ovulation indicates a potential beneficial role for polycystic ovary syndrome. EBioMedicine 2018; 36:539-552. [PMID: 30292674 PMCID: PMC6197718 DOI: 10.1016/j.ebiom.2018.09.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
Background The ovulatory dysfunction mechanisms underlying polycystic ovary syndrome (PCOS) are not completely understood. And the roles of EPHA7 and EPHA7-regulated pathway factors in the pathogenesis of anovulation remain to be elucidated. Methods We used human granulosa cells (hGCs) of PCOS and non-PCOS patients to measure EPHA7 and other target gene expressions. We performed in vitro experiments in KGN cells to verify the molecular mechanisms. Additionally, we conducted in vivo loss- and gain-of-function studies using EPHA7 shRNA lentivirus and recombinant EPHA7-Fc protein injection to identify the ovulation effects of EPHA7. Findings EPHA7 functions as a critically positive upstream factor for the expression of ERK1/2-mediated C/EBPβ. This protein, in turn, induced the expression of KLF4 and then ADAMTS1. Moreover, decreased abundance of EPHA7 was positively correlated with that of its downstream factors in hGCs of PCOS patients. Additionally, a 1-week functional EPHA7 shRNA lentivirus in rat ovaries contributed to decreased numbers of retrieved oocytes, and a 3-week functional lentivirus led to menstrual disorders and morphological polycystic changes in rat ovaries. More importantly, we found that EPHA7 triggered ovulation in rats, and it improved polycystic ovarian changes induced by DHEA in PCOS rats. Interpretation Our findings demonstrate a new role of EPHA7 in PCOS, suggesting that EPHA7 is an effective target for the development of innovative medicines to induce ovulation. Fund National Key Research and Development Program of China, National Natural Science Foundation, Shanghai Municipal Education Commission--Gaofeng Clinical Medicine, and Shanghai Commission of Science and Technology.
Collapse
Affiliation(s)
- Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Junyu Zhai
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jiansheng Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Fangfang Di
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Weiping Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| |
Collapse
|
9
|
|
10
|
Ligand-dependent EphA7 signaling inhibits prostate tumor growth and progression. Cell Death Dis 2017; 8:e3122. [PMID: 29022918 PMCID: PMC5682672 DOI: 10.1038/cddis.2017.507] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/08/2017] [Accepted: 08/17/2017] [Indexed: 01/02/2023]
Abstract
The downregulation of receptor tyrosine kinase EphA7 is frequent in epithelial cancers and linked to tumor progression. However, the detailed mechanism of EphA7-mediated prostate tumor progression remains elusive. To test the role of EphA7 receptor in prostate cancer (PCa) progression directly, we generated EphA7 receptor variants that were either lacking the cytoplasmic domain or carrying a point mutation that inhibits its phosphorylation by site-directed mutagenesis. Overexpression of wild-type (WT) EphA7 in PCa cells resulted in decreased tumor volume and increased tumor apoptosis in primary tumors. In addition, ectopic expression of WT EphA7 both can delay PCa cell proliferation and could inhibit PCa cell migration and invasion. This protein can also induce PCa cell apoptosis that correlated with increasing the protein expression levels of Bax, elevating the caspase-3 activities, reducing the protein expression levels of Bcl-2 and facilitating the dephosphorylation of Akt, which is further increased by the stimulation of ephrinA5-Fc. However, expression of these EphA7 mutants in PCa cells has no effect in vivo and in vitro. The expression of EphA7 and ephrinA5 was significantly decreased in PCa specimens compared with BPH tissues or paired normal tissues. Moreover, the phosphorylation of EphA7 was positively related with ephrinA5 expression in human prostate tissues. In sum, receptor phosphorylation of EphA7, at least in part, suppress PCa tumor malignancy through targeting PI3K/Akt signaling pathways.
Collapse
|
11
|
Guan Y, Liang G, Martin GB, Guan LL. Functional changes in mRNA expression and alternative pre-mRNA splicing associated with the effects of nutrition on apoptosis and spermatogenesis in the adult testis. BMC Genomics 2017; 18:64. [PMID: 28068922 PMCID: PMC5223305 DOI: 10.1186/s12864-016-3385-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 12/07/2016] [Indexed: 01/02/2023] Open
Abstract
Background The effects of nutrition on testis mass in the sexually mature male have long been known, however, the cellular and molecular processes of the testis response to nutrition was not fully understood. Methods We tested whether the defects in spermatogenesis and increases in germ cell apoptosis in the testis that are induced by under-nutrition are associated with changes in mRNA expression and pre-mRNA alternative splicing using groups of 8 male sheep fed for a 10% increase or 10% decrease in body mass over 65 days. Results We identified 2,243 mRNAs, including TP53 and Claudin 11, that were differentially expressed in testis from underfed and well-fed sheep (FDR < 0.1), and found that their expression changed in parallel with variations in germ cell numbers, testis size, and spermatogenesis. Furthermore, pairs of 269 mRNAs and 48 miRNAs were identified on the basis of target prediction. The regulatory effect of miRNAs on mRNA expression, in combination with functional analysis, suggests that these miRNAs are involved in abnormal reproductive morphology, apoptosis and male infertility. Nutrition did not affect the total number of alternative splicing events, but affected 206 alternative splicing events. A total of 159 genes, including CREM, SPATA6, and DDX4, were differentially spliced between dietary treatments, with functions related to RNA splicing and spermatogenesis. In addition, three gene modules were positively correlated with spermatogenesis-related phenotypic traits and negatively related to apoptosis-related phenotypic traits. Among these gene modules, seven (CFLAR, PTPRC, F2R, MAP3K1, EPHA7, APP, BCAP31) were also differentially expressed between nutritional treatments, indicating their potential as markers of spermatogenesis or apoptosis. Conclusions Our findings on significant changes in mRNAs and pre-mRNA alternative splicing under-nutrition suggest that they may partly explain the disruption of spermatogenesis and the increase germ cell apoptosis. However, more research is required to verify their causal effects in regulating spermatogenesis and germ cell apoptosis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3385-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yongjuan Guan
- UWA Institute of Agriculture and School of Animal Biology, University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia.,, Present address: 304 Rosenthal, 3800 Spruce Street, Philadelphia, PA, 19104, USA
| | - Guanxiang Liang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Graeme B Martin
- UWA Institute of Agriculture and School of Animal Biology, University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia.
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada.
| |
Collapse
|
12
|
Johnson C, Segovia B, Kandpal RP. EPHA7 and EPHA10 Physically Interact and Differentially Co-localize in Normal Breast and Breast Carcinoma Cell Lines, and the Co-localization Pattern Is Altered in EPHB6-expressing MDA-MB-231 Cells. Cancer Genomics Proteomics 2016; 13:359-368. [PMID: 27566654 PMCID: PMC5070625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/27/2016] [Indexed: 06/06/2023] Open
Abstract
Erythropoietin-producing hepatocellular carcinoma cell (EPH) receptors comprise the most abundant receptor tyrosine kinase family characterized to date in mammals including humans. These proteins are involved in axon guidance, tissue organization, vascular development and the intricate process of various diseases including cancer. These diverse functions of EPH receptors are attributed, in part, to their abilities for heterodimerization. While the interacting partners of kinase-deficient EPHB6 receptor have been characterized, the interaction of the kinase-dead EPHA10 with any other receptor has not been identified. By using co-immunoprecipitation, we demonstrated physical interaction between kinase-deficient EPHA10 with kinase-sufficient EPHA7 receptor. Immunocytochemical analyses have revealed that these two receptors co-localize on the cell surface, and soluble portions of the receptors exist as a complex in the cytoplasm as well as the nuclei. While EPHA7 and EPHA10 co-localize similarly on the membrane in MCF10A and MCF7 cells, they were differentially co-localized in MDA-MB-231 cells stably transfected with empty pcDNA vector (MDA-MB-231-PC) or an expression construct of EPHB6 (MDA-MB-231-B6). The full-length isoforms of these receptors were co-localized on the cell surface, and the soluble forms were present as a complex in the cytoplasm as well as the nucleus in MDA-MB-231-PC cells. MDA-MB-231-B6 cells, on the other hand, were distinguished by the absence of any signal in the nuclei. Our results represent the first demonstration of physical interaction between EPHA10 and EPHA7 and their cellular co-localization. Furthermore, these observations also suggest gene-regulatory functions of the complex of the soluble forms of these receptors in breast carcinoma cells of differential invasiveness.
Collapse
Affiliation(s)
- Candace Johnson
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, U.S.A
| | - Briana Segovia
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, U.S.A
| | - Raj P Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, U.S.A.
| |
Collapse
|
13
|
Amegandjin CA, Jammow W, Laforest S, Riad M, Baharnoori M, Badeaux F, DesGroseillers L, Murai KK, Pasquale EB, Drolet G, Doucet G. Regional expression and ultrastructural localization of EphA7 in the hippocampus and cerebellum of adult rat. J Comp Neurol 2016; 524:2462-78. [PMID: 26780036 DOI: 10.1002/cne.23962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/18/2015] [Accepted: 01/04/2016] [Indexed: 11/06/2022]
Abstract
EphA7 is expressed in the adult central nervous system (CNS), where its roles are yet poorly defined. We mapped its distribution using in situ hybridization (ISH) and immunohistochemistry (IHC) combined with light (LM) and electron microscopy (EM) in adult rat and mouse brain. The strongest ISH signal was in the hippocampal pyramidal and granule cell layers. Moderate levels were detected in habenula, striatum, amygdala, the cingulate, piriform and entorhinal cortex, and in cerebellum, notably the Purkinje cell layer. The IHC signal distribution was consistent with ISH results, with transport of the protein to processes, as exemplified in the hippocampal neuropil layers and weakly stained pyramidal cell layers. In contrast, in the cerebellum, the Purkinje cell bodies were the most strongly immunolabeled elements. EM localized the cell surface-expression of EphA7 essentially in postsynaptic densities (PSDs) of dendritic spines and shafts, and on some astrocytic leaflets, in both hippocampus and cerebellum. Perikaryal and dendritic labeling was mostly intracellular, associated with the synthetic and trafficking machineries. Immunopositive vesicles were also observed in axons and axon terminals. Quantitative analysis in EM showed significant differences in the frequency of labeled elements between regions. Notably, labeled dendrites were ∼3-5 times less frequent in cerebellum than in hippocampus, but they were individually endowed with ∼10-40 times higher frequencies of PSDs, on their shafts and spines. The cell surface localization of EphA7, being preferentially in PSDs, and in perisynaptic astrocytic leaflets, provides morphologic evidence that EphA7 plays key roles in adult CNS synaptic maintenance, plasticity, or function. J. Comp. Neurol. 524:2462-2478, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Clara A Amegandjin
- Département de neurosciences and Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, Montréal, QC, Canada
| | - Wafaa Jammow
- Département de neurosciences and Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, Montréal, QC, Canada
| | - Sylvie Laforest
- Centre hospitalier de l'Université Laval (CHUL), Québec, QC, Canada
| | - Mustapha Riad
- Département de neurosciences and Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, Montréal, QC, Canada
| | - Moogeh Baharnoori
- Département de neurosciences and Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, Montréal, QC, Canada
| | - Frédérique Badeaux
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Luc DesGroseillers
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Keith K Murai
- Department of Neurology and Neurosurgery, McGill University, and Center for Research in Neuroscience, Montréal, QC, Canada
| | - Elena B Pasquale
- Sanford-Burnham Medical Research Institute, La Jolla, California, and Pathology Department, University of California, San Diego, La Jolla, California, USA
| | - Guy Drolet
- Centre hospitalier de l'Université Laval (CHUL), Québec, QC, Canada
| | - Guy Doucet
- Département de neurosciences and Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
14
|
Lee H, Park S, Kang YS, Park S. EphA receptors form a complex with caspase-8 to induce apoptotic cell death. Mol Cells 2015; 38:349-55. [PMID: 25855521 PMCID: PMC4400310 DOI: 10.14348/molcells.2015.2279] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/24/2014] [Accepted: 12/26/2014] [Indexed: 12/02/2022] Open
Abstract
EphA7 has been implicated in the regulation of apoptotic cell death in neural epithelial cells. In this report, we provide evidence that EphA7 interacts with caspase-8 to induce apoptotic cell signaling. First, a pull-down assay using biotinylated ephrinA5-Fc showed that EphA7 coprecipitated with wild type caspase-8 or catalytically inactive caspase-8 mutant. Second, co-transfection of EphA7 with caspase-8 significantly increased the number of cleaved caspase-3 positive apoptotic cells under an experimental condition where transfection of EphA7 or caspase-8 alone did not affect cell viability or apoptosis. EphA4 also had a causative role in inducing apoptotic cell death with caspase-8, whereas EphA8 did not. Third, caspase-8 catalytic activity was essential for the apoptotic signaling cascade, whereas tyrosine kinase activity of the EphA4 receptor was not. Interestingly, we found that kinaseinactive EphA4 was well co-localized at the plasma membrane with catalytically inactive caspase-8, suggesting that an interaction between these mutant proteins was more stable. Finally, we observed that the extracellular region of the EphA7 receptor was critical for interacting with caspase-8, whereas the cytoplasmic region of EphA7 was not. Therefore, we propose that Eph receptors physically associate with a transmembrane protein to form an apoptotic signaling complex and that this unidentified receptorlike protein acts as a biochemical linker between the Eph receptor and caspase-8.
Collapse
Affiliation(s)
- Haeryung Lee
- Department of Biological Science, Sookmyung Women’s University, Seoul 140-742,
Korea
| | - Sunjung Park
- Department of Biological Science, Sookmyung Women’s University, Seoul 140-742,
Korea
| | - Young-Sook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Sookmyung Women’s University, Seoul 140-742,
Korea
| | - Soochul Park
- Department of Biological Science, Sookmyung Women’s University, Seoul 140-742,
Korea
| |
Collapse
|
15
|
Li Y, Liu DX, Li MY, Qin XX, Fang WG, Zhao WD, Chen YH. Ephrin-A3 and ephrin-A4 contribute to microglia-induced angiogenesis in brain endothelial cells. Anat Rec (Hoboken) 2014; 297:1908-18. [PMID: 25070915 DOI: 10.1002/ar.22998] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 06/05/2014] [Indexed: 12/14/2022]
Abstract
The association of microglia with brain vasculature during development and the reduced brain vascular complexity in microglia-deficient mice suggest the role of microglia in cerebrovascular angiogenesis. However, the underlying molecular mechanism remains unclear. Here, using an in vitro angiogenesis model, we found the culture supernatant of BV2 microglial cells significantly enhanced capillary-like tube formation and migration of brain microvascular endothelial cells (BMECs). The expression of angiogenic factors, ephrin-A3 and ephrin-A4, were specifically upregulated in BMECs exposed to BV2-derived culture supernatant. Knockdown of ephrin-A3 and ephrin-A4 in BMECs by siRNA significantly attenuated the enhanced angiogenesis and migration of BMECs induced by BV2 supernatant. Our further results indicated that the ability of BV2 supernatant to promote endothelial angiogenesis was caused by the soluble tumor necrosis factor α (TNF-α) released from BV2 microglial cells. Moreover, the upregulations of ephrin-A3 and ephrin-A4 in BMECs in response to BV2 supernatant were effectively abolished by neutralization antibody against TNF-α and TNF receptor 1, respectively. The present study provides evidence that microglia upregulates endothelial ephrin-A3 and ephrin-A4 to facilitate in vitro angiogenesis of brain endothelial cells, which is mediated by microglia-released TNF-α.
Collapse
Affiliation(s)
- Ying Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, 110001, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110001, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Park S. Brain-Region Specific Apoptosis Triggered by Eph/ephrin Signaling. Exp Neurobiol 2013; 22:143-8. [PMID: 24167409 PMCID: PMC3807001 DOI: 10.5607/en.2013.22.3.143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 09/11/2013] [Accepted: 09/11/2013] [Indexed: 11/19/2022] Open
Abstract
Eph receptors and their ligands, ephrins, are abundantly expressed in neuroepithelial cells of the early embryonic brain. Overstimulation of Eph signaling in vivo increases apoptotic cell death of neuroepithelial cells, whereas null mutation of the Eph gene leads to the development of a larger brain during embryogenesis. Thus, it appears that Eph-ephrin signaling plays a role in regulating apoptotic cell death of neuroepithelial cells, thereby influencing brain size during embryonic development. Interestingly, Eph-ephrin signaling is bi-directional, with forward signaling from ephrin- to Eph-expressing cells and reverse signaling from Eph- to ephrin-expressing cells. However, it is not clear whether this forward or reverse signaling plays a role in regulating the size of the neuroepithelial cell population during early brain development. Also, Eph receptors and their corresponding ligands are mutually exclusive in their expression domains, and they encounter each other only at interfaces between their expression domains. This expression pattern may be a critical mechanism for preventing overstimulation of Eph-ephrin signaling. Nevertheless, Eph receptors are co-expressed with their corresponding ligands in certain brain regions. Recently, two studies demonstrated that brain region-specific apoptosis may be triggered by the overlapping expression of Eph and ephrin, a theme that will be explored in this mini-review.
Collapse
Affiliation(s)
- Soochul Park
- Department of Biological Science, Sookmyung Women's University, Seoul 140-742, Korea
| |
Collapse
|
17
|
Kim Y, Park E, Noh H, Park S. Expression of EphA8-Fc in transgenic mouse embryos induces apoptosis of neural epithelial cells during brain development. Dev Neurobiol 2013; 73:702-12. [PMID: 23696555 DOI: 10.1002/dneu.22092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/04/2013] [Accepted: 05/13/2013] [Indexed: 12/17/2022]
Abstract
EphAs and ephrin-As are expressed in multiple regions of the developing brain and have been implicated in regulating brain size. Here, we report the identification of a novel mechanism in which reverse signaling through ephrin-As controls neural epithelial cell number in the developing brain. Ectopic expression of EphA8-Fc in transgenic embryos induced apoptosis of neural epithelial cells, which was accompanied by a dramatic decrease in brain size. The number of ephrin-A5-expressing cells was significantly reduced in the brain region where EphA8-Fc was ectopically expressed. Furthermore, in vitro culture of the dissociated neuroepithelial cells revealed that EphA8-Fc enhanced apoptotic cell death of the ephrinA5-expressing cells in a caspase-dependent manner. Thus, our results suggest that reverse signaling through ephrin-As is biochemically linked with caspase-dependent proapoptotic signaling during early brain development.
Collapse
Affiliation(s)
- Yujin Kim
- Department of Biological Science, Sookmyung Women's University, Yongsan-gu, Seoul 140-742, Korea
| | | | | | | |
Collapse
|