1
|
Qi C, Bujaroski RS, Baell J, Zheng X. Kinases in cerebral cavernous malformations: Pathogenesis and therapeutic targets. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119488. [PMID: 37209718 DOI: 10.1016/j.bbamcr.2023.119488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
Cerebral cavernous malformations (CCMs) are low-flow, hemorrhagic vascular lesions of the central nervous system of genetic origin, which can cause stroke-like symptoms and seizures. From the identification of CCM1, CCM2 and CCM3 as genes related to disease progression, molecular and cellular mechanisms for CCM pathogenesis have been established and the search for potential drugs to target CCM has begun. Broadly speaking, kinases are the major group signaling in CCM pathogenesis. These include the MEKK3/MEK5/ERK5 cascade, Rho/Rock signaling, CCM3/GCKIII signaling, PI3K/mTOR signaling, and others. Since the discovery of Rho/Rock in CCM pathogenesis, inhibitors for Rho signaling and subsequently other components in CCM signaling were discovered and applied in preclinical and clinical trials to ameliorate CCM progression. This review discusses the general aspects of CCM disease, kinase-mediated signaling in CCM pathogenesis and the current state of potential treatment options for CCM. It is suggested that kinase target drug development in the context of CCM might facilitate and meet the unmet requirement - a non-surgical option for CCM disease.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, China
| | - Richard Sean Bujaroski
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Australian Translational Medicinal Chemistry Facility (ATMCF), Monash University, Parkville, Victoria, Australia
| | - Jonathan Baell
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, China.
| |
Collapse
|
2
|
Wu S, Wang J, Liu J, Zhu H, Li R, Wan X, Lei J, Li Y, You C, Hu F, Zhang S, Zhao K, Shu K, Lei T. Programmed cell death 10 increased blood-brain barrier permeability through HMGB1/TLR4 mediated downregulation of endothelial ZO-1 in glioblastoma. Cell Signal 2023; 107:110683. [PMID: 37075875 DOI: 10.1016/j.cellsig.2023.110683] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 04/21/2023]
Abstract
Dysfunction of blood brain barrier (BBB) contributes to the development of peritumoral edema (PTE) and GBM progression. Programmed cell death 10 (PDCD10) exerts various influence on cancers, especially in glioblastoma (GBM). We previously found that PDCD10 expression was positively correlated with PTE extent in GBM. Thus, the present study aims to investigate the emerging role of PDCD10 in regulating BBB permeability in GBM. Here we found that in vitro indirect co-culture of ECs with Pdcd10-overexpressed GL261 cells resulted in a significant increase of FITC-Dextran (MW, 4000) leakage by reducing endothelial zonula occluden-1 (ZO-1) and Claudin-5 expression in ECs respectively. Overexpression of Pdcd10 in GBM cells (GL261) triggered an increase of soluble high mobility group box 1 (HMGB1) release, which in turn activated endothelial toll like receptor 4 (TLR4) and downstream NF-κB, Erk1/2 and Akt signaling in ECs through a paracrine manner. Moreover, Pdcd10-overexpressed GL261 cells facilitated a formation of abnormal vasculature and increased the BBB permeability in vivo. Our present study demonstrates that upregulation of PDCD10 in GBM triggered HMGB1/TLR4 signaling in ECs and significantly decreased endothelial ZO-1 expression, which in turn dominantly increased BBB permeability and contributed to tumor progression in GBM.
Collapse
Affiliation(s)
- Sisi Wu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Junwen Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Jingdian Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Ran Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Xueyan Wan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Jin Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Yu Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Feng Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China.
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| |
Collapse
|
3
|
The Dual Role of PDCD10 in Cancers: A Promising Therapeutic Target. Cancers (Basel) 2022; 14:cancers14235986. [PMID: 36497468 PMCID: PMC9740655 DOI: 10.3390/cancers14235986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Programmed cell death 10 (PDCD10) was initially considered as a protein associated with apoptosis. However, recent studies showed that PDCD10 is actually an adaptor protein. By interacting with multiple molecules, PDCD10 participates in various physiological processes, such as cell survival, migration, cell differentiation, vesicle trafficking, cellular senescence, neurovascular development, and gonadogenesis. Moreover, over the past few decades, accumulating evidence has demonstrated that the aberrant expression or mutation of PDCD10 is extremely common in various pathological processes, especially in cancers. The dysfunction of PDCD10 has been strongly implicated in oncogenesis and tumor progression. However, the updated data seem to indicate that PDCD10 has a dual role (either pro- or anti-tumor effects) in various cancer types, depending on cell/tissue specificity with different cellular interactors. In this review, we aimed to summarize the knowledge of the dual role of PDCD10 in cancers with a special focus on its cellular function and potential molecular mechanism. With these efforts, we hoped to provide new insight into the future development and application of PDCD10 as a clinical therapeutic target in cancers.
Collapse
|
4
|
Sun Y, Zhao Z, Zhang H, Li J, Chen J, Luan X, Min W, He Y. The interaction of lead exposure and CCM3 defect plays an important role in regulating angiogenesis through eNOS/NO pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 79:103407. [PMID: 32512318 DOI: 10.1016/j.etap.2020.103407] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/14/2020] [Accepted: 05/03/2020] [Indexed: 06/11/2023]
Abstract
In this study, we aimed to explore the role of nitric oxide (NO) in regulating angiogenesis in cerebral cavernous malformations 3 gene (CCM3)-deficient mice exposed to lead during vascular development; further, we aimed to identify and study the potential mechanism involved as well. Angiogenesis was detected by whole mount immunofluorescent staining of retinal vessels in WT and CCM3+/- mice. Brain microvascular endothelial cells (BMECs) isolated from WT and CCM3+/- mice, primary HUVECs, and immortalized HUVECs (imHUVECs) (CCM3+/+ and CCM3-/-) were used and treated with lead acetate (PbAc). RT-PCR and Western blotting were used to detect the mRNA and protein expression of iNOS, eNOS, and VEGF genes. The results showed that both lead exposure and CCM3 gene deficiency adversely affected endothelial cell function, causing abnormal angiogenesis and vascular remodeling. The mRNA expression of eNOS and iNOS was significantly different in WT and CCM3+/- BMECs (0.04 ± 0.001 vs. 0.016 ± 0.002; 0.26 ± 0.002 vs. 0.306 ± 0.002, respectively), and the expression of eNOS and iNOS in imHUVECs (CCM3+/+ and CCM3-/-) also increased after PbAc exposure. In conclusion, CCM3 gene-deficient mice were more susceptible to abnormal vascular development after low-level lead exposure, probably due to the release of NO.
Collapse
Affiliation(s)
- Yi Sun
- Department of Health Toxicology, Sun Yat-sen University School of Public Health, Guangzhou, Guangdong, 510080, China; Department of Environmental Health and Occupational Medicine, Guilin Medical University School of Public Health, Guilin, Guangxi, 541004, China
| | - Zhiqiang Zhao
- Department of Health Toxicology, Sun Yat-sen University School of Public Health, Guangzhou, Guangdong, 510080, China
| | - Haifeng Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Jiong Li
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jingli Chen
- Department of Health Toxicology, Sun Yat-sen University School of Public Health, Guangzhou, Guangdong, 510080, China
| | - Xiaoyi Luan
- Department of Environmental Health and Occupational Medicine, Guilin Medical University School of Public Health, Guilin, Guangxi, 541004, China
| | - Wang Min
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yun He
- Department of Health Toxicology, Sun Yat-sen University School of Public Health, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
5
|
Endothelial cell clonal expansion in the development of cerebral cavernous malformations. Nat Commun 2019; 10:2761. [PMID: 31235698 PMCID: PMC6591323 DOI: 10.1038/s41467-019-10707-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 05/29/2019] [Indexed: 12/22/2022] Open
Abstract
Cerebral cavernous malformation (CCM) is a neurovascular familial or sporadic disease that is characterised by capillary-venous cavernomas, and is due to loss-of-function mutations to any one of three CCM genes. Familial CCM follows a two-hit mechanism similar to that of tumour suppressor genes, while in sporadic cavernomas only a small fraction of endothelial cells shows mutated CCM genes. We reported that in mouse models and in human patients, endothelial cells lining the lesions have different features from the surrounding endothelium, as they express mesenchymal/stem-cell markers. Here we show that cavernomas originate from clonal expansion of few Ccm3-null endothelial cells that express mesenchymal/stem-cell markers. These cells then attract surrounding wild-type endothelial cells, inducing them to express mesenchymal/stem-cell markers and to contribute to cavernoma growth. These characteristics of Ccm3-null cells are reminiscent of the tumour-initiating cells that are responsible for tumour growth. Our data support the concept that CCM has benign tumour characteristics. Cerebral cavernous malformation is a vascular disease characterized by capillary-venous cavernomas in the central nervous system. Here the authors show that cavernomas display benign tumor characteristics and originate from the clonal expansion of mutated endothelial progenitors which can attract surrounding wild-type cells, inducing their mesenchymal transition and leading to growth of the cavernoma.
Collapse
|
6
|
Shi Y, Song Y, Liu P, Li P. YKL-40 can promote angiogenesis in sporadic cerebral cavernous malformation (CCM). J Clin Neurosci 2019; 64:220-226. [PMID: 30948312 DOI: 10.1016/j.jocn.2019.03.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/26/2019] [Accepted: 03/21/2019] [Indexed: 12/19/2022]
Abstract
The factors affecting the formation of sporadic CCMs remain unclear. A cDNA microarray was used to identify characteristic gene expression patterns in sporadic CCMs. Transcription level of YKL-40 was confirmed by reverse transcription-polymerase chain reaction (RT-PCR). The location and expression were revealed by immunochemistry, immunofluorescence staining and level of YKL-40 was quantified by Western blotting. Alterations to endothelial function following the up or down regulation of gene expression was assessed by Transwell assays, cell counting kit-8 assays and capillary-like tube formation assays in human brain microvascular endothelial cells (HBMECs) in vitro. We generated a murine model by stereotaxically injecting HBMECs with expressing amounts of YKL-40 into the brain. cDNA microarray and RT-PCR results revealed that the transcription level of YKL-40 was ≥140-fold higher in sporadic CCMs in healthy controls. Histological staining revealed excessive YKL-40 expression in the CCM endothelium. Western blotting results analysis showed that YKL-40 protein expression was significantly higher in CCM endothelium (P < 0.05). YKL-40 over-expressing HBMECs showed increased cell proliferation, migration and tube formation ability compared with the control group, whereas downregulating of YKL-40 inhibited the proliferation, migration of HBMECs and capillary-like tube formation (P < 0.05). In animals, increased of YKL-40 was associated with abnormal vascular lesions that were similar to CCMs. YKL-40 is over-expressed in the CCM endothelium and acts as an angiogenic factor that promotes the pathogenesis of sporadic CCMs. YKL-40 may therefore represent a potential therapeutic target in the treatment of sporadic CCM.
Collapse
Affiliation(s)
- Yuan Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Wulumiqi Rd., Shanghai 200040, PR China.
| | - Yaying Song
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Rd. No.2, Shanghai 200025, PR China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Wulumiqi Rd., Shanghai 200040, PR China.
| | - Peiliang Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Wulumiqi Rd., Shanghai 200040, PR China.
| |
Collapse
|
7
|
Scimone C, Donato L, Marino S, Alafaci C, D’Angelo R, Sidoti A. Vis-à-vis: a focus on genetic features of cerebral cavernous malformations and brain arteriovenous malformations pathogenesis. Neurol Sci 2018; 40:243-251. [DOI: 10.1007/s10072-018-3674-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/01/2018] [Indexed: 01/07/2023]
|
8
|
Nickel AC, Wan XY, Saban DV, Weng YL, Zhang S, Keyvani K, Sure U, Zhu Y. Loss of programmed cell death 10 activates tumor cells and leads to temozolomide-resistance in glioblastoma. J Neurooncol 2018; 141:31-41. [PMID: 30392087 DOI: 10.1007/s11060-018-03017-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/25/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE Glioblastoma (GBM) is one of the most aggressive and incurable primary brain tumors. Identification of novel therapeutic targets is an urgent priority. Programmed cell death 10 (PDCD10), a ubiquitously expressed apoptotic protein, has shown a dual function in different types of cancers and in chemo-resistance. Recently, we reported that PDCD10 was downregulated in human GBM. The aim of this study was to explore the function of PDCD10 in GBM cells. METHODS PDCD10 was knocked down in three GBM cell lines (U87, T98g and LN229) by lentiviral-mediated shRNA transduction. U87 and T98g transduced cells were used for phenotype study and LN229 and T98g cells were used for apoptosis study. The role of PDCD10 in apoptosis and chemo-resistance was investigated after treatment with staurosporine and temozolomide. A GBM xenograft mouse model was used to confirm the function of PDCD10 in vivo. A protein array was performed in PDCD10-knockdown and control GBM cells. RESULTS Knockdown of PDCD10 in GBM cells promoted cell proliferation, adhesion, migration, invasion, and inhibited apoptosis and caspase-3 activation. PDCD10-knockdown accelerated tumor growth and increased tumor mass by 2.1-fold and led to a chemo-resistance of mice treated with temozolomide. Immunostaining revealed extensive Ki67-positive cells and less activation of caspase-3 in PDCD10-knockdown tumors. The protein array demonstrated an increased release of multiple growth factors from PDCD10-knockdown GBM cells. CONCLUSIONS Loss of programmed cell death 10 activates tumor cells and leads to temozolomide-resistance in GBM, suggesting PDCD10 as a potential target for GBM therapy.
Collapse
Affiliation(s)
- Ann-Christin Nickel
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Xue-Yan Wan
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dino-Vitali Saban
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yin-Lun Weng
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shu Zhang
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Ulrich Sure
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yuan Zhu
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
9
|
Nardella G, Visci G, Guarnieri V, Castellana S, Biagini T, Bisceglia L, Palumbo O, Trivisano M, Vaira C, Scerrati M, Debrasi D, D'Angelo V, Carella M, Merla G, Mazza T, Castori M, D'Agruma L, Fusco C. A single-center study on 140 patients with cerebral cavernous malformations: 28 new pathogenic variants and functional characterization of a PDCD10 large deletion. Hum Mutat 2018; 39:1885-1900. [PMID: 30161288 DOI: 10.1002/humu.23629] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/12/2018] [Accepted: 07/22/2018] [Indexed: 12/12/2022]
Abstract
Cerebral cavernous malformation (CCM) is a capillary malformation arising in the central nervous system. CCM may occur sporadically or cluster in families with autosomal dominant transmission, incomplete penetrance, and variable expressivity. Three genes are associated with CCM KRIT1, CCM2, and PDCD10. This work is a retrospective single-center molecular study on samples from multiple Italian clinical providers. From a pool of 317 CCM index patients, we found germline variants in either of the three genes in 80 (25.2%) probands, for a total of 55 different variants. In available families, extended molecular analysis found segregation in 60 additional subjects, for a total of 140 mutated individuals. From the 55 variants, 39 occurred in KRIT1 (20 novel), 8 in CCM2 (4 novel), and 8 in PDCD10 (4 novel). Effects of the three novel KRIT1 missense variants were characterized in silico. We also investigated a novel PDCD10 deletion spanning exon 4-10, on patient's fibroblasts, which showed significant reduction of interactions between KRIT1 and CCM2 encoded proteins and impaired autophagy process. This is the largest study in Italian CCM patients and expands the known mutational spectrum of KRIT1, CCM2, and PDCD10. Our approach highlights the relevance of seeking supporting information to pathogenicity of new variants for the improvement of management of CCM.
Collapse
Affiliation(s)
- Grazia Nardella
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Grazia Visci
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Vito Guarnieri
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Stefano Castellana
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Tommaso Biagini
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Luigi Bisceglia
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Orazio Palumbo
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Marina Trivisano
- Department of Neuroscience, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Carmela Vaira
- Department of Neurosurgery, Università Politecnica delle Marche, Ancona, Italy
| | - Massimo Scerrati
- Department of Neurosurgery, Università Politecnica delle Marche, Ancona, Italy
| | - Davide Debrasi
- Department of Pediatrics, Università Federico II, Naples, Italy
| | | | - Massimo Carella
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giuseppe Merla
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Marco Castori
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Leonardo D'Agruma
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carmela Fusco
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
10
|
Long SL, Li YK, Xie YJ, Long ZF, Shi JF, Mo ZC. Neurite Outgrowth Inhibitor B Receptor: A Versatile Receptor with Multiple Functions and Actions. DNA Cell Biol 2017; 36:1142-1150. [PMID: 29058484 DOI: 10.1089/dna.2017.3813] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Members of the reticulon protein family are predominantly distributed within the endoplasmic reticulum. The neurite outgrowth inhibitor (Nogo) has three subtypes, including Nogo-A (200 kDa), Nogo-B (55 kDa), and Nogo-C (25 kDa). Nogo-A and Nogo-C are potent Nogos that are predominantly expressed in the central nervous system. Nogo-B, the splice variant of reticulon-4, is expressed widely in multiple human organ systems, including the liver, lung, kidney, blood vessels, and inflammatory cells. Moreover, the Nogo-B receptor (NgBR) can interact with Nogo-B and can independently affect nervous system regeneration, the chemotaxis of endothelial cells, proliferation, and apoptosis. In recent years, it has been demonstrated that NgBR plays an important role in human pathophysiological processes, including lipid metabolism, angiogenesis, N-glycosylation, cell apoptosis, chemoresistance in human hepatocellular carcinoma, and epithelial-mesenchymal transition. The pathophysiologic effects of NgBR have garnered increased attention, and the detection and enhancement of NgBR expression may be a novel approach to monitor the development and to improve the prognosis of relevant human clinical diseases.
Collapse
Affiliation(s)
- Shuang-Lian Long
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Yu-Kun Li
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Yuan-Jie Xie
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Zhi-Feng Long
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Jin-Feng Shi
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Zhong-Cheng Mo
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| |
Collapse
|
11
|
Update on Novel CCM Gene Mutations in Patients with Cerebral Cavernous Malformations. J Mol Neurosci 2016; 61:189-198. [DOI: 10.1007/s12031-016-0863-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 11/11/2016] [Indexed: 10/20/2022]
|
12
|
Retta SF, Glading AJ. Oxidative stress and inflammation in cerebral cavernous malformation disease pathogenesis: Two sides of the same coin. Int J Biochem Cell Biol 2016; 81:254-270. [PMID: 27639680 PMCID: PMC5155701 DOI: 10.1016/j.biocel.2016.09.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
CCM proteins play pleiotropic roles in various redox-sensitive signaling pathways. CCM proteins modulate the crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses. Oxidative stress and inflammation are emerging as key focal determinants of CCM lesion formation, progression and severity. The pleiotropic functions of CCM proteins may prevent vascular dysfunctions triggered by local oxidative stress and inflammatory events. The distinct therapeutic compounds proposed so far for CCM disease share the ability to modulate redox signaling and autophagy.
Cerebral Cavernous Malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or is inherited as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM lesions exhibit a range of different phenotypes, including wide inter-individual differences in lesion number, size, and susceptibility to intracerebral hemorrhage (ICH). Lesions may remain asymptomatic or result in pathological conditions of various type and severity at any age, with symptoms ranging from recurrent headaches to severe neurological deficits, seizures, and stroke. To date there are no direct therapeutic approaches for CCM disease besides the surgical removal of accessible lesions. Novel pharmacological strategies are particularly needed to limit disease progression and severity and prevent de novo formation of CCM lesions in susceptible individuals. Useful insights into innovative approaches for CCM disease prevention and treatment are emerging from a growing understanding of the biological functions of the three known CCM proteins, CCM1/KRIT1, CCM2 and CCM3/PDCD10. In particular, accumulating evidence indicates that these proteins play major roles in distinct signaling pathways, including those involved in cellular responses to oxidative stress, inflammation and angiogenesis, pointing to pathophysiological mechanisms whereby the function of CCM proteins may be relevant in preventing vascular dysfunctions triggered by these events. Indeed, emerging findings demonstrate that the pleiotropic roles of CCM proteins reflect their critical capacity to modulate the fine-tuned crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses, providing a novel mechanistic scenario that reconciles both the multiple signaling pathways linked to CCM proteins and the distinct therapeutic approaches proposed so far. In addition, recent studies in CCM patient cohorts suggest that genetic susceptibility factors related to differences in vascular sensitivity to oxidative stress and inflammation contribute to inter-individual differences in CCM disease susceptibility and severity. This review discusses recent progress into the understanding of the molecular basis and mechanisms of CCM disease pathogenesis, with specific emphasis on the potential contribution of altered cell responses to oxidative stress and inflammatory events occurring locally in the microvascular environment, and consequent implications for the development of novel, safe, and effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Saverio Francesco Retta
- Department of Clinical and Biological Sciences, School of Medicine and Surgery, University of Torino, Regione Gonzole 10, 10043 Orbassano, Torino, Italy; CCM Italia Research Network(1).
| | - Angela J Glading
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, 14642 Rochester, NY, USA.
| |
Collapse
|
13
|
Gao Y, Yin Y, Xing X, Zhao Z, Lu Y, Sun Y, Zhuang Z, Wang M, Ji W, He Y. Arsenic-induced anti-angiogenesis via miR-425-5p-regulated CCM3. Toxicol Lett 2016; 254:22-31. [DOI: 10.1016/j.toxlet.2016.04.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/30/2016] [Accepted: 04/26/2016] [Indexed: 10/21/2022]
|
14
|
Spiegler S, Kirchmaier B, Rath M, Korenke GC, Tetzlaff F, van de Vorst M, Neveling K, Acker-Palmer A, Kuss AW, Gilissen C, Fischer A, Schulte-Merker S, Felbor U. FAM222B Is Not a Likely Novel Candidate Gene for Cerebral Cavernous Malformations. Mol Syndromol 2016; 7:144-52. [PMID: 27587990 DOI: 10.1159/000446884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2016] [Indexed: 12/11/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are prevalent slow-flow vascular lesions which harbour the risk to develop intracranial haemorrhages, focal neurological deficits, and epileptic seizures. Autosomal dominantly inherited CCMs were found to be associated with heterozygous inactivating mutations in 3 genes, CCM1 (KRIT1), CCM2 (MGC4607), and CCM3 (PDCD10) in 1999, 2003 and 2005, respectively. Despite the availability of high-throughput sequencing techniques, no further CCM gene has been published since. Here, we report on the identification of an autosomal dominantly inherited frameshift mutation in a gene of thus far unknown function, FAM222B (C17orf63), through exome sequencing of CCM patients mutation-negative for CCM1-3. A yeast 2-hybrid screen revealed interactions of FAM222B with the tubulin cytoskeleton and STAMBP which is known to be associated with microcephaly-capillary malformation syndrome. However, a phenotype similar to existing models was not found, neither in fam222bb/fam222ba double mutant zebrafish generated by transcription activator-like effector nucleases nor in an in vitro sprouting assay using human umbilical vein endothelial cells transfected with siRNA against FAM222B. These observations led to the assumption that aberrant FAM222B is not involved in the formation of CCMs.
Collapse
Affiliation(s)
- Stefanie Spiegler
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Bettina Kirchmaier
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt am Main, Germany; Hubrecht Institute - KNAW & UMC Utrecht, Utrecht, The Netherlands
| | - Matthias Rath
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | | | - Fabian Tetzlaff
- Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maartje van de Vorst
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Kornelia Neveling
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt am Main, Germany
| | - Andreas W Kuss
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Andreas Fischer
- Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Cells-in-Motion Cluster of Excellence, Faculty of Medicine, University of Münster, Münster, Germany; Hubrecht Institute - KNAW & UMC Utrecht, Utrecht, The Netherlands
| | - Ute Felbor
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
15
|
Kar S, Baisantry A, Nabavi A, Bertalanffy H. Role of Delta-Notch signaling in cerebral cavernous malformations. Neurosurg Rev 2016; 39:581-9. [DOI: 10.1007/s10143-015-0699-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/03/2015] [Accepted: 12/23/2015] [Indexed: 11/28/2022]
|
16
|
Wu Z, Qi Y, Guo Z, Li P, Zhou D. miR-613 suppresses ischemia-reperfusion-induced cardiomyocyte apoptosis by targeting the programmed cell death 10 gene. Biosci Trends 2016; 10:251-7. [DOI: 10.5582/bst.2016.01122] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Zhenhua Wu
- ICU, Departmentof Cardiac Surgery, Tianjin Chest Hospital
| | - Yujuan Qi
- ICU, Departmentof Cardiac Surgery, Tianjin Chest Hospital
| | - Zhigang Guo
- Department of Cardiac Surgery, Tianjin Chest Hospital
| | - Peijun Li
- ICU, Departmentof Cardiac Surgery, Tianjin Chest Hospital
| | - Ding Zhou
- TEDA International Cardiovascular Hospital
| |
Collapse
|
17
|
Rana U, Liu Z, Kumar SN, Zhao B, Hu W, Bordas M, Cossette S, Szabo S, Foeckler J, Weiler H, Chrzanowska-Wodnicka M, Holtz ML, Misra RP, Salato V, North PE, Ramchandran R, Miao QR. Nogo-B receptor deficiency causes cerebral vasculature defects during embryonic development in mice. Dev Biol 2015; 410:190-201. [PMID: 26746789 DOI: 10.1016/j.ydbio.2015.12.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/21/2015] [Accepted: 12/21/2015] [Indexed: 01/07/2023]
Abstract
Nogo-B receptor (NgBR) was identified as a receptor specific for Nogo-B. Our previous work has shown that Nogo-B and its receptor (NgBR) are essential for chemotaxis and morphogenesis of endothelial cells in vitro and intersomitic vessel formation via Akt pathway in zebrafish. Here, we further demonstrated the roles of NgBR in regulating vasculature development in mouse embryo and primitive blood vessel formation in embryoid body culture systems, respectively. Our results showed that NgBR homozygous knockout mice are embryonically lethal at E7.5 or earlier, and Tie2Cre-mediated endothelial cell-specific NgBR knockout (NgBR ecKO) mice die at E11.5 and have severe blood vessel assembly defects in embryo. In addition, mutant embryos exhibit dilation of cerebral blood vessel, resulting in thin-walled endothelial caverns. The similar vascular defects also were detected in Cdh5(PAC)-CreERT2 NgBR inducible ecKO mice. Murine NgBR gene-targeting embryonic stem cells (ESC) were generated by homologous recombination approaches. Homozygous knockout of NgBR in ESC results in cell apoptosis. Heterozygous knockout of NgBR does not affect ESC cell survival, but reduces the formation and branching of primitive blood vessels in embryoid body culture systems. Mechanistically, NgBR knockdown not only decreases both Nogo-B and VEGF-stimulated endothelial cell migration by abolishing Akt phosphorylation, but also decreases the expression of CCM1 and CCM2 proteins. Furthermore, we performed immunofluorescence (IF) staining of NgBR in human cerebral cavernous malformation patient tissue sections. The quantitative analysis results showed that NgBR expression levels in CD31 positive endothelial cells is significantly decreased in patient tissue sections. These results suggest that NgBR may be one of important genes coordinating the cerebral vasculature development.
Collapse
Affiliation(s)
- Ujala Rana
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Zhong Liu
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Suresh N Kumar
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Baofeng Zhao
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wenquan Hu
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michelle Bordas
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stephanie Cossette
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sara Szabo
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jamie Foeckler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; BloodCenter of Wisconsin, Milwaukee, WI 53226, USA
| | - Hartmut Weiler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; BloodCenter of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Mary L Holtz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ravindra P Misra
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Valerie Salato
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Paula E North
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ramani Ramchandran
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Qing Robert Miao
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
18
|
Lambertz N, El Hindy N, Kreitschmann-Andermahr I, Stein KP, Dammann P, Oezkan N, Mueller O, Sure U, Zhu Y. Downregulation of programmed cell death 10 is associated with tumor cell proliferation, hyperangiogenesis and peritumoral edema in human glioblastoma. BMC Cancer 2015; 15:759. [PMID: 26490252 PMCID: PMC4618952 DOI: 10.1186/s12885-015-1709-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022] Open
Abstract
Background Neovascularization and peritumoral edema are hallmarks of glioblastoma (GBM). Programmed cell death 10 (PDCD10) plays a pivotal role in regulating apoptosis, neoangiogenesis and vessel permeability and is implicated in certain tumor signaling pathways. However, little is known about PDCD10 in GBM. We aimed to investigate the expression pattern of PDCD10 and to identify the association of its expression with some molecular and clinical parameters in human GBM. Methods mRNA and protein expression of PDCD10 were examined respectively by real-time RT-PCR and Western blotting in GBM (n = 27), astrocytoma grade II (n = 13) and control (n = 11). The protein level of p-Akt and GFAP was detected by Western blot. Double-imunofluorecent staining was performed to reveal the cellular expression profile of PDCD10. Brain edema and microvascular density (MVD) were respectively analyzed based on pre-operative MRI and after laminin immnostaining. MGMT promoter methylation was detected by methylation specific PCR. Results mRNA and protein levels of PDCD10 were significantly downregulated in GBM, concomitantly accompanied by the activation of Akt. PDCD10 immunoreactivity was absent in proliferating tumor cells, endothelial cells and GFAP-positive cells, but exclusively present in the hypoxic pseudopalisading cells which underwent apoptosis. Moreover, loss of PDCD10 was associated with a higher MVD and a more severe peritumoral edema but not with MGMT promoter methylation in GBM. Conclusion We report for the first time that PDCD10 expression is downregulated in GBM, which is associated with the activation of Akt signaling protein. PDCD10 is potentially implicated in tumor proliferation and apoptosis, hyperangiogenesis and peritumoral edema in GBM.
Collapse
Affiliation(s)
- Nicole Lambertz
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Nicolai El Hindy
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | | | - Klaus Peter Stein
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany. .,Present Address: Department of Neurosurgery, KRH Klinikum Nordstadt, Haltenhoffstr. 41, 30167, Hannover, Germany.
| | - Philipp Dammann
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Neriman Oezkan
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Oliver Mueller
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Ulrich Sure
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Yuan Zhu
- Department of Neurosurgery, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
19
|
Zhu Y, Zhao K, Prinz A, Keyvani K, Lambertz N, Kreitschmann-Andermahr I, Lei T, Sure U. Loss of endothelial programmed cell death 10 activates glioblastoma cells and promotes tumor growth. Neuro Oncol 2015; 18:538-48. [PMID: 26254477 DOI: 10.1093/neuonc/nov155] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/15/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Neo-angiogenesis is a hallmark of glioblastoma (GBM) and is sustained by autocrine and paracrine interactions between neoplastic and nonneoplastic cells. Programmed cell death 10 (PDCD10) is ubiquitously expressed in nearly all tissues and plays crucial roles in regulating angiogenesis and apoptosis. We recently discovered the absence of PDCD10 expression in the tumor vessels of GBM patients. This raised the hypothesis that loss of endothelial PDCD10 affected GBM cell phenotyping and tumor progression. METHODS Endothelial PDCD10 was silenced by siRNA and lentiviral shRNA. The tumor cell phenotype was studied in direct and indirect co-culture of endothelial cells (ECs) with U87 or LN229. Angiogenic protein array was performed in the media of PDCD10-silenced ECs. Tumor angiogenesis and tumor growth were investigated in a human GBM xenograft mouse model. RESULTS Endothelial silence of PDCD10 significantly stimulated tumor cell proliferation, migration, adhesion, and invasion and inhibited apoptosis in co-cultures. Stable knockdown of endothelial PDCD10 increased microvessel density and the formation of a functional vascular network, leading to a 4-fold larger tumor mass in mice. Intriguingly, endothelial deletion of PDCD10 increased (≥2-fold) the release of 20 of 55 tested proangiogenic factors including VEGF, which in turn activated Erk1/2 and Akt in GBM cells. CONCLUSIONS For the first time, we provide evidence that loss of endothelial PDCD10 activates GBM cells and promotes tumor growth, most likely via a paracrine mechanism. PDCD10 shows a tumor-suppressor-like function in the cross talk between ECs and tumor cells and is potentially implicated in GBM progression.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Kai Zhao
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Anja Prinz
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Kathy Keyvani
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Nicole Lambertz
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Ilonka Kreitschmann-Andermahr
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Ting Lei
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| | - Ulrich Sure
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany (Y.Z., K.Z., A.P., N.L., I.K.-A., U.S.); Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany (K.K.); Department of Neurosurgery, Tongji Medical College, Wuhan, China (K.Z., T.L.)
| |
Collapse
|
20
|
Xu X, Ge S, Jia R, Zhou Y, Song X, Zhang H, Fan X. Hypoxia-induced miR-181b enhances angiogenesis of retinoblastoma cells by targeting PDCD10 and GATA6. Oncol Rep 2015; 33:2789-96. [PMID: 25872572 DOI: 10.3892/or.2015.3900] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/19/2015] [Indexed: 11/06/2022] Open
Abstract
Previous findings showed that miR-181b is upregulated under hypoxic conditions in retinoblastoma cells. Since hypoxia is a common feature of retinoblastoma that affects tumor progression as well as tumor therapy, in the present study, we investigated the regulatory mechanism of miR-181b under hypoxic conditions, and examined the role of miR-181b in retinoblastoma responses to hypoxia (chemoresistance and angiogenesis) and possible downstream genes. The level of hypoxia-inducible factor-1α (HIF-1α) and miR-181b was detected to examine the link between them. Tube formation and cell cytotoxicity assays were used to clarify the effects of miR-181b on hypoxic responses of retinoblastoma cells. Bioinformatics analysis was performed to predict potential targets of miR-181b and western blotting was used to verify these targets. The results showed a significantly increased expression of HIF-1α in hypoxia-treated retinoblastoma cells. Downregulation of HIF-1α using a small-interfering RNA (siRNA) knockdown technology did not decrease the expression of miR-181b. Through gain- and loss-of-function studies, miR-181b was demonstrated to significantly stimulate the ability of capillary tube formation of endothelial cells. Programmed cell death-10 (PDCD10) and GATA binding protein 6 (GATA6) were identified as the target genes of miR‑181b. To the best of our knowledge, results of the present study provide the first evidence that miR-181b was upregulated by hypoxia in retinoblastoma in an HIF-1α-independent manner. miR-181b increased tumor angiogenesis of retinoblastoma cells. Additionally, miR-181b exerts its angiogenic function, at least in part, by inhibiting PDCD10 and GATA6. Thus, it is a new potentially useful therapeutic target for retinoblastoma.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yixiong Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xin Song
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - He Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
21
|
Guerrero A, Iglesias C, Raguz S, Floridia E, Gil J, Pombo CM, Zalvide J. The cerebral cavernous malformation 3 gene is necessary for senescence induction. Aging Cell 2015; 14:274-83. [PMID: 25655101 PMCID: PMC4364839 DOI: 10.1111/acel.12316] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2014] [Indexed: 11/28/2022] Open
Abstract
Mutations in cerebral cavernous malformation 3 gene are known to result in development of vascular malformations and have recently been proposed to also give rise to meningiomas. We report in this study that lack of CCM3 unexpectedly impairs the senescence response of cells, and this is related to the inability of CCM3-deficient cells to induce the C/EBPβ transcription factor and implement the senescence-associated secretory phenotype. Induction of C/EBPβ and cytokines is also impaired in the absence of CCM3 in response to cytokines in nonsenescent cells, pointing to it being a primary defect and not secondary to impaired senescence. CCM3-deficient cells also have a defect in autophagy at late passages of culture, and this defect is also not dependent on impaired senescence, as it is evident in immortal cells after nutrient starvation. Further, these two defects may be related, as enforcing autophagy in CCM3-deficient late passage cells increases C/EBPβ cytokine expression. These results broaden our knowledge on the mechanisms by which CCM3 deficiency results in disease and open new avenues of research into both CCM3 and senescence biology.
Collapse
Affiliation(s)
- Ana Guerrero
- Department of Physiology; CIMUS; Instituto de Investigación Sanitaria IDIS; University of Santiago de Compostela; Santiago de Compostela A Coruña 15703 Spain
- Cell Proliferation Group; MRC Clinical Sciences Centre; Imperial College London; London W12 0NN UK
| | - Cristina Iglesias
- Department of Physiology; CIMUS; Instituto de Investigación Sanitaria IDIS; University of Santiago de Compostela; Santiago de Compostela A Coruña 15703 Spain
| | - Selina Raguz
- Cell Proliferation Group; MRC Clinical Sciences Centre; Imperial College London; London W12 0NN UK
- Quantitative Cell Biology; MRC Clinical Sciences Centre; Imperial College; London
| | - Ebel Floridia
- Department of Physiology; CIMUS; Instituto de Investigación Sanitaria IDIS; University of Santiago de Compostela; Santiago de Compostela A Coruña 15703 Spain
| | - Jesús Gil
- Cell Proliferation Group; MRC Clinical Sciences Centre; Imperial College London; London W12 0NN UK
| | - Celia M. Pombo
- Department of Physiology; CIMUS; Instituto de Investigación Sanitaria IDIS; University of Santiago de Compostela; Santiago de Compostela A Coruña 15703 Spain
| | - Juan Zalvide
- Department of Physiology; CIMUS; Instituto de Investigación Sanitaria IDIS; University of Santiago de Compostela; Santiago de Compostela A Coruña 15703 Spain
| |
Collapse
|
22
|
Yang YJ, Liu ZS, Lu SY, Li C, Hu P, Li YS, Liu NN, Tang F, Xu YM, Zhang JH, Li ZH, Feng XL, Zhou Y, Ren HL. Molecular cloning, expression and characterization of programmed cell death 10 from sheep (Ovis aries). Gene 2014; 558:65-74. [PMID: 25541025 DOI: 10.1016/j.gene.2014.12.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/27/2014] [Accepted: 12/19/2014] [Indexed: 11/30/2022]
Abstract
Programmed cell death 10 (PDCD10) is a highly conserved adaptor protein. Its mutations result in cerebral cavernous malformations (CCMs). In this study, PDCD10 cDNA from the buffy coat of Small Tail Han sheep (Ovis aries) was cloned from a suppression subtractive hybridization cDNA library, named OaPDCD10. The full-length cDNA of OaPDCD10 was 1343bp with a 639bp open reading frame (ORF) encoding 212 amino acid residues. Tissue distribution of OaPDCD10 mRNA determined that it was ubiquitously expressed in all tested tissue samples, and the highest expression was observed in the heart. The differential expression of OaPDCD10 between infected sheep (challenged with Brucella melitensis) and vaccinated sheep (vaccinated with Brucella suis S2) was also investigated. The results revealed that, compared to the control group, the expression of OaPDCD10 from infected and vaccinated sheep was both significantly up-regulated (p<0.05). Moreover, the expression levels of OaPDCD10 from the vaccinated sheep were significantly higher than the infected sheep (p<0.05) after 30days post-inoculation. The recombinant OaPDCD10 (rOaPDCD10) protein was expressed in Escherichia coli BL21 (DE3), and then purified by affinity chromatography. The rOaPDCD10 protein was demonstrated to induce apoptosis and promote cell proliferation. Our studies are intended to discover potential diagnostic biomarkers of brucellosis to discern infected sheep from vaccinated sheep, and OaPDCD10 could be considered as a potential diagnostic biomarker of brucellosis.
Collapse
Affiliation(s)
- Yong-Jie Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; Department of Food Science, College of Agriculture, Yanbian University, Yanji 133002, China
| | - Zeng-Shan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shi-Ying Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chuang Li
- Department of Food Science, College of Agriculture, Yanbian University, Yanji 133002, China
| | - Pan Hu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yan-Song Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Nan-Nan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Feng Tang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; College of Animal Husbandry and Veterinary, Liaoning Medical University, Jinzhou 121001, China
| | - Yun-Ming Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; Department of Husbandry and Veterinary Medicine, Jiangsu Polytechnic College of Agriculture and Forestry, Jurong 212400, China
| | - Jun-Hui Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhao-Hui Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiao-Li Feng
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hong-Lin Ren
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
23
|
Hwang J, Pallas DC. STRIPAK complexes: structure, biological function, and involvement in human diseases. Int J Biochem Cell Biol 2014; 47:118-48. [PMID: 24333164 PMCID: PMC3927685 DOI: 10.1016/j.biocel.2013.11.021] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/18/2013] [Accepted: 11/28/2013] [Indexed: 12/31/2022]
Abstract
The mammalian striatin family consists of three proteins, striatin, S/G2 nuclear autoantigen, and zinedin. Striatin family members have no intrinsic catalytic activity, but rather function as scaffolding proteins. Remarkably, they organize multiple diverse, large signaling complexes that participate in a variety of cellular processes. Moreover, they appear to be regulatory/targeting subunits for the major eukaryotic serine/threonine protein phosphatase 2A. In addition, striatin family members associate with germinal center kinase III kinases as well as other novel components, earning these assemblies the name striatin-interacting phosphatase and kinase (STRIPAK) complexes. Recently, there has been a great increase in functional and mechanistic studies aimed at identifying and understanding the roles of STRIPAK and STRIPAK-like complexes in cellular processes of multiple organisms. These studies have identified novel STRIPAK and STRIPAK-like complexes and have explored their roles in specific signaling pathways. Together, the results of these studies have sparked increased interest in striatin family complexes because they have revealed roles in signaling, cell cycle control, apoptosis, vesicular trafficking, Golgi assembly, cell polarity, cell migration, neural and vascular development, and cardiac function. Moreover, STRIPAK complexes have been connected to clinical conditions, including cardiac disease, diabetes, autism, and cerebral cavernous malformation. In this review, we discuss the expression, localization, and protein domain structure of striatin family members. Then we consider the diverse complexes these proteins and their homologs form in various organisms, emphasizing what is known regarding function and regulation. Finally, we explore possible roles of striatin family complexes in disease, especially cerebral cavernous malformation.
Collapse
Affiliation(s)
- Juyeon Hwang
- Department of Biochemistry and Winship Cancer Institute, and Biochemistry, Cell, Developmental Biology Graduate Program, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA.
| | - David C Pallas
- Department of Biochemistry and Winship Cancer Institute, and Biochemistry, Cell, Developmental Biology Graduate Program, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA.
| |
Collapse
|
24
|
Ou JM, Qui MK, Dai YX, Dong Q, Shen J, Dong P, Wang XF, Liu YB, Fei ZW. Combined blockade of AKT/mTOR pathway inhibits growth of human hemangioma via downregulation of proliferating cell nuclear antigen. Int J Immunopathol Pharmacol 2013; 25:945-53. [PMID: 23298485 DOI: 10.1177/039463201202500412] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in the tumorigenesis and progression of multiple tumors, and has been shown to be important therapeutic targets for cancer. The present study aimed to explore the role and molecular mechanisms of AKT/mTOR pathway in human hemangioma (HA). Twenty-five cases of human HA tissues were collected. The expression of AKT, mTOR and proliferating cell nuclear antigen (PCNA) proteins was evaluated using semi-quantitative immunohistochemistry in biopsy samples in different phases of HA. AKT/mTOR pathway was blocked by recombinant small hairpin RNA adenovirus vector rAd5-AKT+mTOR (rAd5-Am), used for infecting proliferating phase HA-derived endothelial cells (HDEC). The expression of AKT, mTOR and PCNA was detected by Real-time PCR and Western blot assays. Cell proliferative activities were determined by MTT assay, and cell cycle distribution and apoptosis were analyzed by flow cytometry. As a consequence, the expression of AKT, mTOR and PCNA was significantly increased in proliferative phase HA, while that was decreased in involutive phase. Combined blockade of AKT/mTOR pathway by rAd5-Am diminished cell proliferative activities, and induced cell apoptosis and cycle arrest with the decreased expression of AKT, mTOR and PCNA in proliferative phase HDEC. In conclusion, the activity of AKT/mTOR pathway was increased in proliferative phase HA, while it was decreased in involutive phase. Combined blockade of AKT/mTOR pathway might suppress cell proliferation via down-regulation of PCNA expression, and induce apoptosis and cycle arrest in proliferative phase HDEC, suggesting that AKT/mTOR pathway might represent the important therapeutic targets for human HA.
Collapse
Affiliation(s)
- J M Ou
- Department of General Surgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bacigaluppi S, Retta SF, Pileggi S, Fontanella M, Goitre L, Tassi L, La Camera A, Citterio A, Patrosso MC, Tredici G, Penco S. Genetic and cellular basis of cerebral cavernous malformations: implications for clinical management. Clin Genet 2013; 83:7-14. [DOI: 10.1111/j.1399-0004.2012.01892.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
26
|
Fischer A, Zalvide J, Faurobert E, Albiges-Rizo C, Tournier-Lasserve E. Cerebral cavernous malformations: from CCM genes to endothelial cell homeostasis. Trends Mol Med 2013; 19:302-8. [PMID: 23506982 DOI: 10.1016/j.molmed.2013.02.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/18/2013] [Accepted: 02/18/2013] [Indexed: 11/18/2022]
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions that can occur sporadically or as a consequence of inherited loss-of-function mutations, predominantly in the genes CCM1 (KRIT1), CCM2 (MGC4607, OSM, Malcavernin), or CCM3 (PDCD10, TFAR15). Inherited, familial CCM is characterized by the development of multiple lesions throughout a patient's life leading to recurrent cerebral hemorrhages. Recently, roles for the CCM proteins in maintaining vascular barrier functions and quiescence have been elucidated, and in this review we summarize the genetics and pathophysiology of this disease and discuss the molecular mechanisms through which CCM proteins may act within blood vessels.
Collapse
Affiliation(s)
- Andreas Fischer
- Vascular Signaling and Cancer (A270), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
27
|
You C, Sandalcioglu IE, Dammann P, Felbor U, Sure U, Zhu Y. Loss of CCM3 impairs DLL4-Notch signalling: implication in endothelial angiogenesis and in inherited cerebral cavernous malformations. J Cell Mol Med 2013; 17:407-18. [PMID: 23388056 PMCID: PMC3823022 DOI: 10.1111/jcmm.12022] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/28/2012] [Indexed: 12/17/2022] Open
Abstract
CCM3, a product of the cerebral cavernous malformation 3 or programmed cell death 10 gene (CCM3/PDCD10), is broadly expressed throughout development in both vertebrates and invertebrates. Increasing evidence indicates a crucial role of CCM3 in vascular development and in regulation of angiogenesis and apoptosis. Furthermore, loss of CCM3 causes inherited (familial) cerebral cavernous malformation (CCM), a common brain vascular anomaly involving aberrant angiogenesis. This study focused on signalling pathways underlying the angiogenic functions of CCM3. Silencing CCM3 by siRNA stimulated endothelial proliferation, migration and sprouting accompanied by significant downregulation of the core components of Notch signalling including DLL4, Notch4, HEY2 and HES1 and by activation of VEGF and Erk pathways. Treatment with recombinant DLL4 (rhDLL4) restored DLL4 expression and reversed CCM3-silence-mediated impairment of Notch signalling and reduced the ratio of VEGF-R2 to VEGF-R1 expression. Importantly, restoration of DLL4-Notch signalling entirely rescued the hyper-angiogenic phenotype induced by CCM3 silence. A concomitant loss of CCM3 and the core components of DLL4-Notch signalling were also demonstrated in CCM3-deficient endothelial cells derived from human CCM lesions (CCMEC) and in a CCM3 germline mutation carrier. This study defined DLL4 as a key downstream target of CCM3 in endothelial cells. CCM3/DLL4-Notch pathway serves as an important signalling for endothelial angiogenesis and is potentially implicated in the pathomechanism of human CCMs.
Collapse
Affiliation(s)
- Chao You
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Fidalgo M, Guerrero A, Fraile M, Iglesias C, Pombo CM, Zalvide J. Adaptor protein cerebral cavernous malformation 3 (CCM3) mediates phosphorylation of the cytoskeletal proteins ezrin/radixin/moesin by mammalian Ste20-4 to protect cells from oxidative stress. J Biol Chem 2012; 287:11556-65. [PMID: 22291017 DOI: 10.1074/jbc.m111.320259] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
While studying the functions of CCM3/PDCD10, a gene encoding an adaptor protein whose mutation results in vascular malformations, we have found that it is involved in a novel response to oxidative stress that results in phosphorylation and activation of the ezrin/radixin/moesin (ERM) family of proteins. This phosphorylation protects cells from accidental cell death induced by oxidative stress. We also present evidence that ERM phosphorylation is performed by the GCKIII kinase Mst4, which is activated and relocated to the cell periphery after oxidative stress. The cellular levels of Mst4 and its activation after oxidative stress depend on the presence of CCM3, as absence of the latter impairs the phosphorylation of ERM proteins and enhances death of cells exposed to reactive oxygen species. These findings shed new light on the response of cells to oxidative stress and identify an important pathophysiological situation in which ERM proteins and their phosphorylation play a significant role.
Collapse
Affiliation(s)
- Miguel Fidalgo
- Department of Physiology and Centro Singular de Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Spain
| | | | | | | | | | | |
Collapse
|
29
|
Cerebral cavernous malformations: from molecular pathogenesis to genetic counselling and clinical management. Eur J Hum Genet 2011; 20:134-40. [PMID: 21829231 DOI: 10.1038/ejhg.2011.155] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cerebral cavernous (or capillary-venous) malformations (CCM) have a prevalence of about 0.1-0.5% in the general population. Genes mutated in CCM encode proteins that modulate junction formation between vascular endothelial cells. Mutations lead to the development of abnormal vascular structures.In this article, we review the clinical features, molecular and genetic basis of the disease, and management.
Collapse
|
30
|
Cunningham K, Uchida Y, O'Donnell E, Claudio E, Li W, Soneji K, Wang H, Mukouyama YS, Siebenlist U. Conditional deletion of Ccm2 causes hemorrhage in the adult brain: a mouse model of human cerebral cavernous malformations. Hum Mol Genet 2011; 20:3198-206. [PMID: 21596842 DOI: 10.1093/hmg/ddr225] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cerebral cavernous malformations (CCM) are irregularly shaped and enlarged capillaries in the brain that are prone to hemorrhage, resulting in headaches, seizures, strokes and even death in patients. The disease affects up to 0.5% of the population and the inherited form has been linked to mutations in one of three genetic loci, CCM1, CCM2 and CCM3. To understand the pathophysiology underlying the vascular lesions in CCM, it is critical to develop a reproducible mouse genetic model of this disease. Here, we report that limited conditional ablation of Ccm2 in young adult mice induces observable neurological dysfunction and reproducibly results in brain hemorrhages whose appearance is highly reminiscent of the lesions observed in human CCM patients. We first demonstrate that conventional or endothelial-specific deletion of Ccm2 leads to fatal cardiovascular defects during embryogenesis, including insufficient vascular lumen formation as well as defective arteriogenesis and heart malformation. These findings confirm and extend prior studies. We then demonstrate that the inducible deletion of Ccm2 in adult mice recapitulates the CCM-like brain lesions in humans; the lesions display disrupted vascular lumens, enlarged capillary cavities, loss of proper neuro-vascular associations and an inflammatory reaction. The CCM lesions also exhibit damaged neuronal architecture, the likely cause of neurologic defects, such as ataxia and seizure. These mice represent the first CCM2 animal model for CCM and should provide the means to elucidate disease mechanisms and evaluate therapeutic strategies for human CCM.
Collapse
Affiliation(s)
- Kirk Cunningham
- Immune Activation Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|