1
|
Cao Y, Su H, Zeng J, Xie Y, Liu Z, Liu F, Qiu Y, Yi F, Lin J, Hammes HP, Zhang C. Integrin β8 prevents pericyte-myofibroblast transition and renal fibrosis through inhibiting the TGF-β1/TGFBR1/Smad3 pathway in diabetic kidney disease. Transl Res 2024; 265:36-50. [PMID: 37931653 DOI: 10.1016/j.trsl.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/08/2023]
Abstract
Diabetic kidney disease (DKD) is one of the leading causes to develop end-stage kidney disease worldwide. Pericytes are implicated in the development of tissue fibrosis. However, the underlying mechanisms of pericytes in DKD remain largely unknown. We isolated and cultured primary pericytes and rat mesangial cells (HBZY-1). Western blot and qRT-PCR analysis were used to explore the role and regulatory mechanism of Integrin β8/transforming growth factor beta 1 (TGF-β1) pathway. We also constructed pericyte-specific Integrin β8 knock-in mice as the research objects to determine the role of Integrin β8 in vivo. We discovered that reduced Integrin β8 expression was closely associated with pericyte transition in DKD. Overexpressed Integrin β8 in pericytes dramatically suppressed TGF-β1/TGF beta receptor 1 (TGFBR1)/Smad3 signaling pathway and protected glomerular endothelial cells (GECs) in vitro. In vivo, pericyte-specific Integrin β8 knock-in ameliorated pericyte transition, endothelium injury and renal fibrosis in STZ-induced diabetic mice. Mechanistically, Murine double minute 2 (MDM2) was found to increase the degradation of Integrin β8 and caused TGF-β1 release and activation. Knockdown MDM2 could partly reverse the decline of Integrin β8 and suppress pericytes transition. In conclusion, the present findings suggested that upregulated MDM2 expression contributes to the degradation of Integrin β8 and activation of TGF-β1/TGFBR1/Smad3 signaling pathway, which ultimately leads to pericyte transition during DKD progression. These results indicate MDM2/Integrin β8 might be considered as therapeutic targets for DKD.
Collapse
Affiliation(s)
- Yiling Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jieyu Zeng
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yaru Xie
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zezhou Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Qiu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jihong Lin
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
2
|
Wang YS, Wang SL, Liu XL, Kang ZC. Platelet-rich plasma promotes peripheral nerve regeneration after sciatic nerve injury. Neural Regen Res 2023; 18:375-381. [PMID: 35900433 PMCID: PMC9396478 DOI: 10.4103/1673-5374.346461] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The effect of platelet-rich plasma on nerve regeneration remains controversial. In this study, we established a rabbit model of sciatic nerve small-gap defects with preserved epineurium and then filled the gaps with platelet-rich plasma. Twenty-eight rabbits were divided into the following groups (7 rabbits/group): model, low-concentration PRP (2.5–3.5-fold concentration of whole blood platelets), medium-concentration PRP (4.5–6.5-fold concentration of whole blood platelets), and high-concentration PRP (7.5–8.5-fold concentration of whole blood platelets). Electrophysiological and histomorphometrical assessments and proteomics analysis were used to evaluate regeneration of the sciatic nerve. Our results showed that platelet-rich plasma containing 4.5–6.5- and 7.5–8.5-fold concentrations of whole blood platelets promoted repair of sciatic nerve injury. Proteomics analysis was performed to investigate the possible mechanism by which platelet-rich plasma promoted nerve regeneration. Proteomics analysis showed that after sciatic nerve injury, platelet-rich plasma increased the expression of integrin subunit β-8 (ITGB8), which participates in angiogenesis, and differentially expressed proteins were mainly enriched in focal adhesion pathways. Additionally, two key proteins, ribosomal protein S27a (RSP27a) and ubiquilin 1 (UBQLN1), which were selected after protein-protein interaction analysis, are involved in the regulation of ubiquitin levels in vivo. These data suggest that platelet-rich plasma promotes peripheral nerve regeneration after sciatic nerve injury by affecting angiogenesis and intracellular ubiquitin levels.
Collapse
|
3
|
Shabani Z, Schuerger J, Su H. Cellular loci involved in the development of brain arteriovenous malformations. Front Hum Neurosci 2022; 16:968369. [PMID: 36211120 PMCID: PMC9532630 DOI: 10.3389/fnhum.2022.968369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Brain arteriovenous malformations (bAVMs) are abnormal vessels that are prone to rupture, causing life-threatening intracranial bleeding. The mechanism of bAVM formation is poorly understood. Nevertheless, animal studies revealed that gene mutation in endothelial cells (ECs) and angiogenic stimulation are necessary for bAVM initiation. Evidence collected through analyzing bAVM specimens of human and mouse models indicate that cells other than ECs also are involved in bAVM pathogenesis. Both human and mouse bAVMs vessels showed lower mural cell-coverage, suggesting a role of pericytes and vascular smooth muscle cells (vSMCs) in bAVM pathogenesis. Perivascular astrocytes also are important in maintaining cerebral vascular function and take part in bAVM development. Furthermore, higher inflammatory cytokines in bAVM tissue and blood demonstrate the contribution of inflammatory cells in bAVM progression, and rupture. The goal of this paper is to provide our current understanding of the roles of different cellular loci in bAVM pathogenesis.
Collapse
Affiliation(s)
- Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Joana Schuerger
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Hua Su
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Hua Su, ; orcid.org/0000-0003-1566-9877
| |
Collapse
|
4
|
Genetics and Vascular Biology of Brain Vascular Malformations. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
5
|
Kashif H, Shah D, Sukumari-Ramesh S. Dysregulation of microRNA and Intracerebral Hemorrhage: Roles in Neuroinflammation. Int J Mol Sci 2021; 22:8115. [PMID: 34360881 PMCID: PMC8347974 DOI: 10.3390/ijms22158115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/23/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a major public health problem and devastating subtype of stroke with high morbidity and mortality. Notably, there is no effective treatment for ICH. Neuroinflammation, a pathological hallmark of ICH, contributes to both brain injury and repair and hence, it is regarded as a potential target for therapeutic intervention. Recent studies document that microRNAs, small non-coding RNA molecules, can regulate inflammatory brain response after ICH and are viable molecular targets to alter brain function. Therefore, there is an escalating interest in studying the role of microRNAs in the pathophysiology of ICH. Herein, we provide, for the first time, an overview of the microRNAs that play roles in ICH-induced neuroinflammation and identify the critical knowledge gap in the field, as it would help design future studies.
Collapse
Affiliation(s)
| | | | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.K.); (D.S.)
| |
Collapse
|
6
|
Potential Second-Hits in Hereditary Hemorrhagic Telangiectasia. J Clin Med 2020; 9:jcm9113571. [PMID: 33167572 PMCID: PMC7694477 DOI: 10.3390/jcm9113571] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant genetic disorder that presents with telangiectases in skin and mucosae, and arteriovenous malformations (AVMs) in internal organs such as lungs, liver, and brain. Mutations in ENG (endoglin), ACVRL1 (ALK1), and MADH4 (Smad4) genes account for over 95% of HHT. Localized telangiectases and AVMs are present in different organs, with frequencies which differ among affected individuals. By itself, HHT gene heterozygosity does not account for the focal nature and varying presentation of the vascular lesions leading to the hypothesis of a “second-hit” that triggers the lesions. Accumulating research has identified a variety of triggers that may synergize with HHT gene heterozygosity to generate the vascular lesions. Among the postulated second-hits are: mechanical trauma, light, inflammation, vascular injury, angiogenic stimuli, shear stress, modifier genes, and somatic mutations in the wildtype HHT gene allele. The aim of this review is to summarize these triggers, as well as the functional mechanisms involved.
Collapse
|
7
|
Cheng P, Ma L, Shaligram S, Walker EJ, Yang ST, Tang C, Zhu W, Zhan L, Li Q, Zhu X, Lawton MT, Su H. Effect of elevation of vascular endothelial growth factor level on exacerbation of hemorrhage in mouse brain arteriovenous malformation. J Neurosurg 2020; 132:1566-1573. [PMID: 31026826 PMCID: PMC6817409 DOI: 10.3171/2019.1.jns183112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/18/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE A high level of vascular endothelial growth factor (VEGF) has been implicated in brain arteriovenous malformation (bAVM) bleeding and rupture. However, direct evidence is missing. In this study the authors used a mouse bAVM model to test the hypothesis that elevation of focal VEGF levels in bAVMs exacerbates the severity of bAVM hemorrhage. METHODS Brain AVMs were induced in adult mice in which activin receptor-like kinase 1 (Alk1, a gene that causes AVM) gene exons 4-6 were floxed by intrabasal ganglia injection of an adenoviral vector expressing Cre recombinase to induce Alk1 mutation and an adeno-associated viral vector expressing human VEGF (AAV-VEGF) to induce angiogenesis. Two doses of AAV-VEGF (5 × 109 [high] or 2 × 109 [low]) viral genomes were used. In addition, the common carotid artery and external jugular vein were anastomosed in a group of mice treated with low-dose AAV-VEGF 6 weeks after the model induction to induce cerebral venous hypertension (VH), because VH increases the VEGF level in the brain. Brain samples were collected 8 weeks after the model induction. Hemorrhages in the bAVM lesions were quantified on brain sections stained with Prussian blue, which detects iron deposition. VEGF levels were quantified in bAVM tissue by enzyme-linked immunosorbent assay. RESULTS Compared to mice injected with a low dose of AAV-VEGF, the mice injected with a high dose had higher levels of VEGF (p = 0.003) and larger Prussian blue-positive areas in the bAVM lesion at 8 or 9 weeks after model induction (p = 0.002). VH increased bAVM hemorrhage in the low-dose AAV-VEGF group. The overall mortality in the high-dose AAV-VEGF group was 26.7%, whereas no mouse died in the low-dose AAV-VEGF group without VH. In contrast, VH caused a mortality of 50% in the low-dose AAV-VEGF group. CONCLUSIONS Using mouse bAVM models, the authors provided direct evidence that elevation of the VEGF level increases bAVM hemorrhage and mouse mortality.
Collapse
Affiliation(s)
- Philip Cheng
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Li Ma
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Sonali Shaligram
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Espen J. Walker
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Shun-Tai Yang
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Chaoliang Tang
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Wan Zhu
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Lei Zhan
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Qiang Li
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Xiaonan Zhu
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| | - Michael T. Lawton
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Hua Su
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, California
| |
Collapse
|
8
|
Bai YY, Niu JZ. miR‑222 regulates brain injury and inflammation following intracerebral hemorrhage by targeting ITGB8. Mol Med Rep 2019; 21:1145-1153. [PMID: 31894320 PMCID: PMC7003054 DOI: 10.3892/mmr.2019.10903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/30/2019] [Indexed: 12/11/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a disease associated with high mortality and morbidity. MicroRNAs (miRNAs) have been reported to be associated with the pathogenesis of numerous cerebrovascular diseases, including ICH. miR-222 has been revealed to play important roles in various physiological and pathological processes in cardiovascular diseases. However, its role in ICH remains largely unknown. The aim of the present study was to evaluate the potential effect of miR-222 on brain injury in ICH. The results revealed that the expression of miR-222 was significantly increased in ICH, and downregulation of miR-222 significantly reduced erythrocyte lysate-induced cell apoptosis by decreasing the levels of cleaved caspase-3, cleaved caspase-9 and Bax and increasing the level of Bcl-2. In addition, downregulation of miR-222 suppressed the inflammatory responses in erythrocyte lysate-induced microglia, and inhibited inflammation, brain water content and improved neurological functions in ICH mice. Mechanistically, integrin subunit β8 (ITGB8) was identified as a direct target of negative regulation by miR-222 in microglia cells, and up-regulation of ITGB8 led to the attenuation of inflammation and apoptosis. Collectively, the present findings indicated that miR-222 was a crucial regulator of inflammation via targeting of ITGB8, and represented a promising therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Yan-Yan Bai
- Department of Neurology, The First Hospital of Yulin, Yulin, Shaanxi 719000, P.R. China
| | - Jun-Zhi Niu
- Department of Information, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
9
|
Barbosa Do Prado L, Han C, Oh SP, Su H. Recent Advances in Basic Research for Brain Arteriovenous Malformation. Int J Mol Sci 2019; 20:ijms20215324. [PMID: 31731545 PMCID: PMC6862668 DOI: 10.3390/ijms20215324] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Arteriovenous malformations (AVMs) are abnormal connections of vessels that shunt blood directly from arteries into veins. Rupture of brain AVMs (bAVMs) can cause life-threatening intracranial bleeding. Even though the majority of bAVM cases are sporadic without a family history, some cases are familial. Most of the familial cases of bAVMs are associated with a genetic disorder called hereditary hemorrhagic telangiectasia (HHT). The mechanism of bAVM formation is not fully understood. The most important advances in bAVM basic science research is the identification of somatic mutations of genes in RAS-MAPK pathways. However, the mechanisms by which mutations of these genes lead to AVM formation are largely unknown. In this review, we summarized the latest advance in bAVM studies and discussed some pathways that play important roles in bAVM pathogenesis. We also discussed the therapeutic implications of these pathways.
Collapse
Affiliation(s)
- Leandro Barbosa Do Prado
- Center for Cerebrovascular Research, Department of Anesthesia, University of California, San Francisco, CA 94143, USA;
| | - Chul Han
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute/Dignity Health, Phoenix, AZ 85013, USA; (C.H.); (S.P.O.)
| | - S. Paul Oh
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute/Dignity Health, Phoenix, AZ 85013, USA; (C.H.); (S.P.O.)
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia, University of California, San Francisco, CA 94143, USA;
- Correspondence: ; Tel.: +01-415-206-3162
| |
Collapse
|
10
|
Feng Z, Ye L, Klebe D, Ding Y, Guo ZN, Flores JJ, Yin C, Tang J, Zhang JH. Anti-inflammation conferred by stimulation of CD200R1 via Dok1 pathway in rat microglia after germinal matrix hemorrhage. J Cereb Blood Flow Metab 2019; 39:97-107. [PMID: 28792282 PMCID: PMC6311673 DOI: 10.1177/0271678x17725211] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
CD200 has been reported to be neuroprotective in neurodegenerative diseases. However, the potential protective effects of CD200 in germinal matrix hemorrhage (GMH) have not been investigated. We examined the anti-inflammatory mechanisms of CD200 after GMH. A total of 167 seven-day-old rat pups were used. The time-dependent effect of GMH on the levels of CD200 and CD200 Receptor 1 (CD200R1) was evaluated by western blot. CD200R1 was localized by immunohistochemistry. The short-term (24 h) and long-term (28 days) outcomes were evaluated after CD200 fusion protein (CD200Fc) treatment by neurobehavioral assessment. CD200 small interfering RNA (siRNA) and downstream of tyrosine kinase 1 (Dok1) siRNA were injected intracerebroventricularly. Western blot was employed to study the mechanisms of CD200 and CD200R1. GMH induced significant developmental delay and caused impairment in both cognitive and motor functions in rat pups. CD200Fc ameliorated GMH-induced damage. CD200Fc increased expression of Dok1 and decreased IL-1beta and TNF-alpha levels. CD200R1 siRNA and Dok1 siRNA abolished the beneficial effects of CD200Fc, as demonstrated by enhanced expression levels of IL-1beta and TNF-alpha. CD200Fc inhibited GMH-induced inflammation and this effect may be mediated by CD200R1/Dok1 pathway. Thus, CD200Fc may serve as a potential treatment to ameliorate brain injury for GMH patients.
Collapse
Affiliation(s)
- Zhanhui Feng
- 1 Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Lan Ye
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,3 The Medical Function Laboratory of Experimental Teaching Center of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Damon Klebe
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Yan Ding
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Zhen-Ni Guo
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jerry J Flores
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Cheng Yin
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- 2 Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.,4 Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,5 Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
11
|
Zhu W, Saw D, Weiss M, Sun Z, Wei M, Shaligram S, Wang S, Su H. Induction of Brain Arteriovenous Malformation Through CRISPR/Cas9-Mediated Somatic Alk1 Gene Mutations in Adult Mice. Transl Stroke Res 2018; 10:557-565. [PMID: 30511203 DOI: 10.1007/s12975-018-0676-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/05/2018] [Accepted: 11/11/2018] [Indexed: 02/07/2023]
Abstract
Brain arteriovenous malformation (bAVM) is an important risk factor for intracranial hemorrhage. The pathogenesis of bAVM has not been fully understood. Animal models are important tools for dissecting bAVM pathogenesis and testing new therapies. We have developed several mouse bAVM models using genetically modified mice. However, due to the body size, mouse bAVM models have some limitations. Recent studies identified somatic mutations in sporadic human bAVM. To develop a feasible tool to create sporadic bAVM in rodent and animals larger than rodent, we made tests using the CRISPR/Cas9 technique to induce somatic gene mutations in mouse brain in situ. Two sequence-specific guide RNAs (sgRNAs) targeting mouse Alk1 exons 4 and 5 were cloned into pAd-Alk1e4sgRNA + e5sgRNA-Cas9 plasmid. These sgRNAs were capable to generate mutations in Alk1 gene in mouse cell lines. After packaged into adenovirus, Ad-Alk1e4sgRNA + e5sgRNA-Cas9 was co-injected with an adeno-associated viral vector expressing vascular endothelial growth factor (AAV-VEGF) into the brains of wild-type C57BL/6J mice. Eight weeks after viral injection, bAVMs were detected in 10 of 12 mice. Compared to the control (Ad-GFP/AAV-VEGF-injected) brain, 13% of Alk1 alleles were mutated and Alk1 expression was reduced by 26% in the Ad-Alk1e4sgRNA + e5sgRNA-Cas9/AAV-VEGF-injected brains. Around the Ad-Alk1e4sgRNA + e5sgRNA-Cas9/AAV-VEGF injected site, Alk1-null endothelial cells were detected. Our data demonstrated that CRISPR/Cas9 is a feasible tool for generating bAVM model in animals.
Collapse
Affiliation(s)
- Wan Zhu
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel Saw
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Miriam Weiss
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Zhengda Sun
- Department of Radiology, University of California, San Francisco, San Francisco, CA, USA
| | - Meng Wei
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Sonali Shaligram
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Sen Wang
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
12
|
Ye L, Feng Z, Doycheva D, Malaguit J, Dixon B, Xu N, Zhang JH, Tang J. CpG-ODN exerts a neuroprotective effect via the TLR9/pAMPK signaling pathway by activation of autophagy in a neonatal HIE rat model. Exp Neurol 2017; 301:70-80. [PMID: 29274721 DOI: 10.1016/j.expneurol.2017.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/28/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022]
Abstract
Hypoxic Ischemic Encephalopathy (HIE) is an injury caused to the brain due to prolonged lack of oxygen and blood supply which results in death or long-term disabilities. The main aim of this study was to investigate the role of Cytosine-phospho-guanine oligodeoxynucleotide (CpG-ODN) in autophagy after HIE. Ten-day old (P10) rat pups underwent right common carotid artery ligation followed by 2.5h of hypoxia as previously described by Rice-Vannucci. At 1h post HIE, rats were intranasally administered with recombinant CpG-ODN. Time-course expression levels of endogenous key proteins, TLR9, pAMPK/AMPK, LC3II/I, and LAMP1 involved in CpG-ODN's protective effects were measured using western blot. Short (48h) and long (4w) term neurobehavior studies were performed using righting reflex, negative geotaxis, water maze, foot fault and Rota rod tests. Brain samples were collected after long term for histological analysis. Furthermore, to elucidate the pathway via which CpG-ODN confers protection, TLR9 and AMPK inhibitors were used. Time course results showed that the expression of TLR9, pAMPK/AMPK, LC3II/I, LAMP1 increased after HIE. Neurobehavioral studies showed that HIE induced a significant delay in development and resulted in cognitive and motor function deficits. However, CpG-ODN ameliorated HIE-induced outcomes and improved long term neurological deficits. In addition, CpG-ODN increased expression of pAMPK/AMPK, p-ULK1/ULK1, P-AMBRA1/AMBRA1, LC3II/I and LAMP1 while inhibition of TLR9 and AMPK reversed those effects. In summary, CpG-ODN increased HIE-induced autophagy and improved short and long term neurobehavioral outcomes which may be mediated by the TLR9/pAMPK signaling pathway after HIE.
Collapse
Affiliation(s)
- Lan Ye
- The Medical Function Laboratory of Experimental Teaching Center of Basic Medicine, Guizhou Medical University, Guiyang 550004, China; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Zhanhui Feng
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Desislava Doycheva
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States.
| | - Jay Malaguit
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Brandon Dixon
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States.
| | - Ningbo Xu
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - John H Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Jiping Tang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States.
| |
Collapse
|
13
|
Abstract
During vascular development, endothelial cells (ECs) and neighboring stromal cells interact and communicate through autocrine and paracrine signaling mechanisms involving extracellular matrix (ECM) proteins and their cell surface integrin adhesion receptors. Integrin-mediated adhesion and signaling pathways are crucial for normal vascular development and physiology, and alterations in integrin expression and/or function drive several vascular-related pathologies including thrombosis, autoimmune disorders, and cancer. The purpose of this chapter is to discuss integrin adhesion and signaling pathways important for EC growth, survival, and migration. Integrin-mediated paracrine links between ECs and surrounding stromal cells in the organ microenvironment will also be discussed. Lastly, we will review roles for integrins in vascular pathologies and discuss possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Paola A Guerrero
- University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joseph H McCarty
- University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
14
|
Guerrero PA, Tchaicha JH, Chen Z, Morales JE, McCarty N, Wang Q, Sulman EP, Fuller G, Lang FF, Rao G, McCarty JH. Glioblastoma stem cells exploit the αvβ8 integrin-TGFβ1 signaling axis to drive tumor initiation and progression. Oncogene 2017; 36:6568-6580. [PMID: 28783169 DOI: 10.1038/onc.2017.248] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is a primary brain cancer that contains populations of stem-like cancer cells (GSCs) that home to specialized perivascular niches. GSC interactions with their niche influence self-renewal, differentiation and drug resistance, although the pathways underlying these events remain largely unknown. Here, we report that the integrin αvβ8 and its latent transforming growth factor β1 (TGFβ1) protein ligand have central roles in promoting niche co-option and GBM initiation. αvβ8 integrin is highly expressed in GSCs and is essential for self-renewal and lineage commitment in vitro. Fractionation of β8high cells from freshly resected human GBM samples also reveals a requirement for this integrin in tumorigenesis in vivo. Whole-transcriptome sequencing reveals that αvβ8 integrin regulates tumor development, in part, by driving TGFβ1-induced DNA replication and mitotic checkpoint progression. Collectively, these data identify the αvβ8 integrin-TGFβ1 signaling axis as crucial for exploitation of the perivascular niche and identify potential therapeutic targets for inhibiting tumor growth and progression in patients with GBM.
Collapse
Affiliation(s)
- P A Guerrero
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| | - J H Tchaicha
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| | - Z Chen
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| | - J E Morales
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| | - N McCarty
- The Brown Institute for Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Q Wang
- Department of Radiation Oncology, M. D. Anderson Cancer Center, Houston, TX, USA.,Department of Genomic Medicine, M. D. Anderson Cancer Center, Houston, TX, USA
| | - E P Sulman
- Department of Radiation Oncology, M. D. Anderson Cancer Center, Houston, TX, USA.,Department of Genomic Medicine, M. D. Anderson Cancer Center, Houston, TX, USA.,Department of Translational Molecular Pathology, M. D. Anderson Cancer Center, Houston, TX, USA
| | - G Fuller
- Departments of Pathology, M. D. Anderson Cancer Center, Houston, TX, USA
| | - F F Lang
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| | - G Rao
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| | - J H McCarty
- Department of Neurosurgery, M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
15
|
Wang X, Hao Q, Zhao Y, Guo Y, Ge W. Dysregulation of cell-cell interactions in brain arteriovenous malformations: A quantitative proteomic study. Proteomics Clin Appl 2017; 11. [PMID: 28083997 DOI: 10.1002/prca.201600093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/08/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Xia Wang
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; Beijing 100005 China
| | - Qiang Hao
- Department of Neurosurgery, Beijing Tiantan Hospital; Capital Medical University; Beijing 100050 China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital; Capital Medical University; Beijing 100050 China
| | - Yi Guo
- Department of Neurosurgery; Tsinghua Changgung Hospital; Beijing 102218 China
- Department of Neurosurgery; Affiliated Hospital of Hebei University; Baoding 071000 China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; Beijing 100005 China
| |
Collapse
|