1
|
Zhang X, Zheng J, Zhang L, Zhang J, Feng L, Zhang L, Huang X. Transcriptomic and proteomic integrated analysis reveals molecular mechanisms of 3D bioprinted vaginal scaffolds in vaginal regeneration. Sci Rep 2025; 15:18601. [PMID: 40436951 PMCID: PMC12119898 DOI: 10.1038/s41598-025-00507-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
3D Bioprinting technology has been applied to vaginal reconstruction with satisfactory results. Understanding the transcriptome and proteome of regenerated vaginas is essential for knowing how biomaterials and seed cells contribute to vaginal regeneration. There are no reports on the systemic analysis of vaginal regeneration transcriptomes or proteomes. This study aims to explore the transcriptomic and proteomic features of vaginal tissue reconstructed with 3D bioprinted scaffolds. The scaffolds were made with biomaterials and bone marrow-derived mesenchymal stem cells (BMSCs) and then transplanted into a rabbit model.rna sequencing was used to analyze the transcriptomes of reconstructed and normal vaginal tissues, identifying 11,956 differentially expressed genes (DEGs). Proteomic analysis using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and data-independent acquisition (DIA) identified 7,363 differentially expressed proteins (DEPs). Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses were performed on DEGs and deps. Results showed that DEGs and deps.were involved in extracellular matrix remodeling, angiogenesis, inflammatory response, epithelialization, and muscle formation. This study shows that 3D bioprinted scaffolds are feasible for vaginal reconstruction and offers new insights into the molecular mechanisms involved.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
- Department of Pelvic floor clinic, Cangzhou Central Hospital, Cangzhou, 061600, Hebei Province, China
| | - Jiahua Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Liye Zhang
- Chengde Medical University, Chengde, 067000, Hebei Province, China
| | - Jingkun Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Li Feng
- Department of Gynecology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, 050000, Hebei Province, China
| | - Lin Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Xianghua Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
2
|
Utami AM, Halfwerk JBG, de Boer OJ, Mackaaij C, Pabittei DR, van der Horst CMAM, Meijer-Jorna LB, van der Wal AC. Relative expression of hormone receptors by endothelial and smooth muscle cells in proliferative and non-proliferative areas of congenital arteriovenous malformations. Eur J Med Res 2023; 28:449. [PMID: 37864259 PMCID: PMC10588228 DOI: 10.1186/s40001-023-01436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 10/07/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Episodic growth due to microvascular proliferations (MVP) has been reported in congenital arteriovenous malformations (AVM), which are normally quiescent lesions composed of mature malformed vessels. Since AVM also may worsen under conditions of hormonal dysregulation, we hypothesized that hormonal influences may stimulate this process of vasoproliferative growth through potential interactions with hormone receptors (HR). METHODS 13 Cases of AVM tissue with histologically documented vasoproliferative growth were analyzed quantitatively for the presence and tissue localization of estrogen receptor (ER), progesterone receptor (PGR), growth hormone receptor (GHR) and follicle-stimulating hormone receptor (FSHR) in relation to resident cells of interest (endothelial cells (EC), smooth muscle cells (SMC) and mast cells (MC)) by applying multiplex immunohistochemistry (IHC) staining. Expression patterns in lesions with MVP and mature vessels were quantified and compared. Available fresh frozen tissues of 3 AVM samples were used to confirm the presence of HR using Reverse-Transcriptase quantitative Polymerase Chain Reaction (RT-qPCR). RESULTS All four HR studied were expressed in all cases within EC and SMC in areas of MVP and mature vessels, but not in normal skin tissue. ER, GHR, and FSHR showed more expression in EC of MVP and in SMC of mature vessels. RT-qPCR confirmed presence of all 4 HR in both areas. CONCLUSION Expression of ER, PGR, GHR, and FSHR in vasoproliferative areas of congenital AVM could explain onset of sudden symptomatic growth, as has observed in a subpopulation of patients. These findings may have implications for eventual anti-hormonal targeted therapy in the lesions involved.
Collapse
Affiliation(s)
- A M Utami
- Department of Pathology, Amsterdam University Medical Center-Location AMC, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Anatomical Pathology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia.
| | - J B G Halfwerk
- Department of Pathology, Amsterdam University Medical Center-Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - O J de Boer
- Department of Pathology, Amsterdam University Medical Center-Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - C Mackaaij
- Department of Anatomy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - D R Pabittei
- Department of Physiology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - C M A M van der Horst
- Department of Plastic Surgery, Amsterdam University Medical Center-Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - L B Meijer-Jorna
- Symbiant Pathology Expert Center, NWZ- Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - A C van der Wal
- Department of Pathology, Amsterdam University Medical Center-Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Alkhaibary A, Alnefaie N, Alharbi A, Alammar H, Arishy AM, Alshaya W, Khairy S. Intracranial dural arteriovenous fistula: a comprehensive review of the history, management, and future prospective. Acta Neurol Belg 2022; 123:359-366. [PMID: 36374476 DOI: 10.1007/s13760-022-02133-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022]
Abstract
Dural arteriovenous fistulas (DAVF) are abnormal acquired intracranial vascular malformations consisting of pathological connections located within the dura between the pial arteries and the veno vasora, comprising the walls of the dural sinuses, bridging veins, or transosseous emissary veins. Dural arteriovenous fistulas are distinguished from arteriovenous malformations by their arterial supply from the vessels that perfuse the dura mater and lack of a parenchymal nidus. They are most commonly situated at the transverse and cavernous sinuses. The mechanism of development behind dural arteriovenous fistula can be explained by the molecular and anatomical factors. Multiple classification systems have been proposed throughout history including; Djindjian and Merland, Cognard, and Borden classification systems. The aggressiveness of the clinical course in intracranial dural arteriovenous fistula can be predicted through the angiographic patterns of venous drainage, more specifically, the presence of cortical venous drainage, the presence of venous ectasia, and the aggressiveness of clinical presentation. Intracranial dural arteriovenous fistulas might be discovered incidentally. However, if symptomatic, the clinical presentation ranges from mild neurological deficits to severe, lethal intracranial hemorrhage. Angiography is the imaging of choice to investigate, diagnose, and plan treatment for intracranial dural arteriovenous fistula. The management algorithm of intracranial dural arteriovenous fistula can be broadly divided into conservative, surgical, endovascular, and/or radiosurgical options. With the advent of endovascular therapies, surgery has fallen out of favor for managing intracranial dural arteriovenous fistulas. In the present article, the pathophysiology, classifications, natural history, clinical manifestations, radiological features, management, and complications are comprehensively reviewed.
Collapse
|
4
|
Sato Y, Falcone-Juengert J, Tominaga T, Su H, Liu J. Remodeling of the Neurovascular Unit Following Cerebral Ischemia and Hemorrhage. Cells 2022; 11:2823. [PMID: 36139398 PMCID: PMC9496956 DOI: 10.3390/cells11182823] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Formulated as a group effort of the stroke community, the transforming concept of the neurovascular unit (NVU) depicts the structural and functional relationship between brain cells and the vascular structure. Composed of both neural and vascular elements, the NVU forms the blood-brain barrier that regulates cerebral blood flow to meet the oxygen demand of the brain in normal physiology and maintain brain homeostasis. Conversely, the dysregulation and dysfunction of the NVU is an essential pathological feature that underlies neurological disorders spanning from chronic neurodegeneration to acute cerebrovascular events such as ischemic stroke and cerebral hemorrhage, which were the focus of this review. We also discussed how common vascular risk factors of stroke predispose the NVU to pathological changes. We synthesized existing literature and first provided an overview of the basic structure and function of NVU, followed by knowledge of how these components remodel in response to ischemic stroke and brain hemorrhage. A greater understanding of the NVU dysfunction and remodeling will enable the design of targeted therapies and provide a valuable foundation for relevant research in this area.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Jaime Falcone-Juengert
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hua Su
- Department of Anesthesia, UCSF, San Francisco, CA 94143, USA
- Center for Cerebrovascular Research, UCSF, San Francisco, CA 94143, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| |
Collapse
|
5
|
Sim SY. Pathophysiology and classification of intracranial and spinal dural AVF. J Cerebrovasc Endovasc Neurosurg 2022; 24:203-209. [PMID: 35443276 PMCID: PMC9537653 DOI: 10.7461/jcen.2022.e2021.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Dural arteriovenous fistulas (DAVFs) are pathologic shunts between pachymeningeal arteries and dural venous channel. DAVFs are relatively rare, however, DAVFs can lead to significant morbidity and mortality due to intracranial hemorrhage and non-hemorrhagic neurologic deterioration related to leptomeningeal venous drainage. The etiology and pathophysiology of DAVFs is not fully understood. Several hypotheses for development of DAVF and classifications for predicting risk of hemorrhage and neurological deficit have been proposed to help clinical decision making according to its natural history. Herein, incidence, etiology, pathophysiology of development of intracranial and spinal DAVF including their classifications are briefly described with short historical review.
Collapse
Affiliation(s)
- Sook Young Sim
- Department of Neurosurgery, Inje University Ilsan Paik Hospital, Goyang, Korea
| |
Collapse
|
6
|
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]
|
7
|
Abstract
Brain arteriovenous malformation (bAVM) is the most common cause of intracranial hemorrhage (ICH), particularly in young patients. However, the exact cause of bAVM bleeding and rupture is not yet fully understood. In bAVMs, blood bypasses the entire capillary bed and directly flows from arteries to veins. The vessel walls in bAVMs have structural defects, which impair vascular integrity. Mural cells are essential structural and functional components of blood vessels and play a critical role in maintaining vascular integrity. Changes in mural cell number and coverage have been implicated in bAVMs. In this review, we discussed the roles of mural cells in bAVM pathogenesis. We focused on 1) the recent advances in human and animal studies of bAVMs; 2) the importance of mural cells in vascular integrity; 3) the regulatory signaling pathways that regulate mural cell function. More specifically, the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-β (PDGFR-β), EphrinB2/EphB4, and angiopoietins/tie2 signaling pathways that regulate mural cell-recruitment during vascular remodeling were discussed in detail.
Collapse
|
8
|
Utami AM, Azahaf S, de Boer OJ, van der Horst CMAM, Meijer-Jorna LB, van der Wal AC. A literature review of microvascular proliferation in arteriovenous malformations of skin and soft tissue. J Clin Transl Res 2021; 7:540-557. [PMID: 34541367 PMCID: PMC8445624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/07/2020] [Accepted: 06/16/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND AIM Arteriovenous malformations (AVM) are defined as being quiescent vascular masses composed of mature vessels. However, recent studies reported areas of microvascular proliferation (MVP) in AVM, indicating a process of angiogenesis. As this finding questions the previous definition, the primary objective of this review was to evaluate whether angiogenesis occurs in vascular malformations of skin and soft tissue, and second, to identify potential factors involved in MVP. METHOD Due to the multifaceted nature of this subject, a hermeneutic methodology was used to select articles that were likely to provide a deeper understanding of MVP in vascular malformations. Through citation tracking and database searching in PubMed and Web of Science, relevant articles were identified. All study designs concerning occurrence of MVP in AVM of skin and soft tissue in all age groups were included in the study. The Newcastle-Ottawa scale was used for quality assessment. RESULTS 16 studies were included in this review which reported occurrence of MVP areas in between the otherwise mature vessels of vascular malformations. In these studies, angiogenesis was reported only in AVM-type of vascular malformations. Increased levels of pro-angiogenic factors were also reported and proliferation was found most prominently during adolescence. Finally, several types of hormone receptors also have been described in tissues of AVM. CONCLUSION Overall, the reviewed data support occurrence of active angiogenesis, highlighted by the presence of MVP in the arteriovenous type of vascular malformations, and a possible concurrent lesion progression towards a higher Schobinger stage of clinical severity. The relative scarcity of data at present implies that further research is required to elucidate the nature of MVP in AVM, which could have implications for developing targeted pharmacotherapy. RELEVANCE FOR PATIENTS Active angiogenesis caused by MVP in AVM patients is known to be correlating to clinical symptoms and contributing to the progression of the disease, recurrence rate, and patient's quality of life.
Collapse
Affiliation(s)
- Amalia Mulia Utami
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pathology Anatomy, Faculty of Medicine, Universitas Hasanuddin, Makassar, Indonesia
| | - Siham Azahaf
- Amsterdam University Medical Center, Vrije University, Amsterdam, The Netherlands
| | - Onno J. de Boer
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Chantal M. A. M. van der Horst
- Department of Plastic Surgery, Amsterdam University Medical Center-location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lorine B. Meijer-Jorna
- Symbiant Pathology Expert Center, NWZ- Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands
| | - Allard C. van der Wal
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Abstract
The complex development of the brain vascular system can be broken down by embryonic stages and anatomic locations, which are tightly regulated by different factors and pathways in time and spatially. The adult brain is relatively quiescent in angiogenesis. However, under disease conditions, such as trauma, stroke, or tumor, angiogenesis can be activated in the adult brain. Disruption of any of the factors or pathways may lead to malformed vessel development. In this chapter, we will discuss factors and pathways involved in normal brain vasculogenesis and vascular maturation, and the pathogenesis of several brain vascular malformations.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - Sonali S Shaligram
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California San Francisco, San Francisco, CA, United States
| | - Hua Su
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
Sadaghianloo N, Contenti J, Dufies M, Parola J, Rouleau M, Lee S, Peyron J, Fabbri L, Hassen‐Khodja R, Pouysségur J, Bost F, Jean‐Baptiste E, Dardik A, Mazure NM. Co-culture of human fibroblasts, smooth muscle and endothelial cells promotes osteopontin induction in hypoxia. J Cell Mol Med 2020; 24:2931-2941. [PMID: 32032472 PMCID: PMC7077551 DOI: 10.1111/jcmm.14905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/20/2019] [Accepted: 11/23/2019] [Indexed: 12/17/2022] Open
Abstract
Arteriovenous fistulas (AVFs) are the preferred vascular access for haemodialysis of patients suffering from end-stage renal disease, a worldwide public health problem. However, they are prone to a high rate of failure due to neointimal hyperplasia and stenosis. This study aimed to determine if osteopontin (OPN) was induced in hypoxia and if OPN could be responsible for driving AVF failure. Identification of new factors that participate in remodelling of AVFs is a challenge. Three cell lines representing the cells of the three layers of the walls of arteries and veins, fibroblasts, smooth muscle cells and endothelial cells, were tested in mono- and co-culture in vitro for OPN expression and secretion in normoxia compared to hypoxia after silencing the hypoxia-inducible factors (HIF-1α, HIF-2α and HIF-1/2α) with siRNA or after treatment with an inhibitor of NF-kB. None of the cells in mono-culture showed OPN induction in hypoxia, whereas cells in co-culture secreted OPN in hypoxia. The changes in oxygenation that occur during AVF maturation up-regulate secretion of OPN through cell-cell interactions between the different cell layers that form AVF, and in turn, these promote endothelial cell proliferation and could participate in neointimal hyperplasia.
Collapse
Affiliation(s)
- Nirvana Sadaghianloo
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Department of Vascular SurgeryCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Julie Contenti
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Department of Emergency MedicineCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | | | - Julien Parola
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
| | | | - Shinrong Lee
- Department of Surgery and the Vascular Biology and Therapeutics ProgramYale UniversityNew HavenCTUSA
- Department of Vascular SurgeryVA Connecticut Healthcare SystemsWest HavenCTUSA
| | - Jean‐François Peyron
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
| | - Lucilla Fabbri
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Réda Hassen‐Khodja
- Department of Vascular SurgeryCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Jacques Pouysségur
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Centre Scientifique de Monaco (CSM)MonacoMonaco
| | - Frédéric Bost
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
| | - Elixène Jean‐Baptiste
- Department of Vascular SurgeryCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Alan Dardik
- Department of Surgery and the Vascular Biology and Therapeutics ProgramYale UniversityNew HavenCTUSA
- Department of Vascular SurgeryVA Connecticut Healthcare SystemsWest HavenCTUSA
| | - Nathalie M. Mazure
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| |
Collapse
|
12
|
Jia YC, Fu JY, Huang P, Zhang ZP, Chao B, Bai J. Characterization of Endothelial Cells Associated with Cerebral Arteriovenous Malformation. Neuropsychiatr Dis Treat 2020; 16:1015-1022. [PMID: 32368063 PMCID: PMC7182449 DOI: 10.2147/ndt.s248356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/31/2020] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Cerebral arteriovenous malformation (cAVM) is a disease characterized by the angiogenesis and remodeling of veins. However, whether vascular endothelial cells (ECs) exhibit morphological and functional changes during cAVM remains unclear. This study aimed to investigate the role of ECs in the pathogenesis of cAVM. METHODS Rat model of cAVM was established by anastomosing the common carotid artery with the external jugular vein. The digital subtraction angiography (DSA), HE, Masson and immunohistochemical staining were performed to evaluate the model. ECs were isolated from AVM rat model or control rats, and characterized by MTT, cell scratch, and tube formation assays. The secretion of vascular endothelial growth factor (VEGF) was detected by ELISA. RESULTS AVM rat model showed typical pathological characteristics of cAVM. In addition, the proliferation, migration and tube formation abilities of ECs of arterialized vein (AV-ECs) were significantly better than those of ECs of normal vein (NV-ECs). Moreover, the levels of secreted VEGF were significantly higher in AV-ECs than in NV-ECs. CONCLUSION AV-ECs isolated from AVM rat model showed increased proliferation, migration and angiogenesis and may be potential target for the treatment of cAVM.
Collapse
Affiliation(s)
- Yu-Chen Jia
- Inner Mongolia Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Jia-Yue Fu
- Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Ping Huang
- Department of Neurosurgery, Affiliated Hospital, Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Zhan-Pu Zhang
- Department of Neurosurgery, Affiliated Hospital, Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Bo Chao
- Department of Neurosurgery, Affiliated Hospital, Inner Mongolia Medical University, Hohhot, People's Republic of China
| | - Jie Bai
- Department of Neurosurgery, Affiliated Hospital, Inner Mongolia Medical University, Hohhot, People's Republic of China
| |
Collapse
|
13
|
Shaligram SS, Winkler E, Cooke D, Su H. Risk factors for hemorrhage of brain arteriovenous malformation. CNS Neurosci Ther 2019; 25:1085-1095. [PMID: 31359618 PMCID: PMC6776739 DOI: 10.1111/cns.13200] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/16/2022] Open
Abstract
Patients with brain arteriovenous malformation (bAVM) are at risk of intracranial hemorrhage (ICH). Overall, bAVM accounts for 25% of hemorrhagic strokes in adults <50 years of age. The treatment of unruptured bAVMs has become controversial, because the natural history of these patients may be less morbid than invasive therapies. Available treatments include observation, surgical resection, endovascular embolization, stereotactic radiosurgery, or combination thereof. Knowing the risk factors for bAVM hemorrhage is crucial for selecting appropriate therapeutic strategies. In this review, we discussed several biological risk factors, which may contribute to bAVM hemorrhage.
Collapse
Affiliation(s)
- Sonali S Shaligram
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative care, University of California, San Francisco, California
| | - Ethan Winkler
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Daniel Cooke
- Department of Radiology, University of California, San Francisco, California
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative care, University of California, San Francisco, California
| |
Collapse
|
14
|
Shen F, Jiang L, Han F, Degos V, Chen S, Su H. Increased Inflammatory Response in Old Mice is Associated with More Severe Neuronal Injury at the Acute Stage of Ischemic Stroke. Aging Dis 2019; 10:12-22. [PMID: 30705764 PMCID: PMC6345332 DOI: 10.14336/ad.2018.0205] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
Stroke occurs mostly in patients with advanced age. Elderly patients have a less favorable prognosis compared with young adult patients. To understand the underlying mechanisms, we tested our hypothesis that an increased inflammatory response to acute ischemic injury in old stroke mice leads to more severe brain damage and behavioral dysfunction. An ischemic stroke model was created in 2- and 12-month-old C57BL/6 mice through permanent occlusion of the left distal middle cerebral artery (dMCAO). Infarct/atrophy volumes were quantified by staining the brain sections with Cresyl Violet. Sensorimotor function was assessed using the corner test and adhesive removal test. Quantification of CD68+ cells in the peri-infarct region was performed at 1, 3 and 14 days after dMCAO. Interleukin-6 (IL-6), interleukin-1 β (IL-1β) and vascular endothelial growth factor (VEGF) levels in the ischemic brain tissue were measured using ELISA. Western blot was used to determine the expression levels of tight junction proteins, claudin-5 and zonula occludens (ZO)-1. Blood-brain barrier permeability was measured by Evans blue (EB) extravasation. Gelatinase B (MMP-9, type IV collagenase) was measured by gel zymography. Compared to 2-month-old mice, 12-month-old mice had more severe behavioral deficits at both the acute and chronic stages of stroke. Compared with the 2-month-old mice, 12-month-old mice had larger infarct/atrophy volumes at 1 and 14 days after dMCAO, higher levels of IL-6 and IL-1β, higher MMP9 activity, and lower levels of claudin-5 and ZO-1 at 1 and 3 days after dMCAO. 12-month-old mice also had more CD68+ cells in the peri-infarct region at 1, 3 and 14 days after dMCAO and more EB leakage at 3 days after dMCAO. A higher inflammatory response at the acute stage of ischemic stroke in old mice is associated with more severe neuronal injury and long-term behavioral dysfunction.
Collapse
Affiliation(s)
- Fanxia Shen
- 1Department of Neurology & Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Lidan Jiang
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Frank Han
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Vincent Degos
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Shengdi Chen
- 1Department of Neurology & Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Su
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
15
|
Growth arrest-specific gene 6 transfer promotes mesenchymal stem cell survival and cardiac repair under hypoxia and ischemia via enhanced autocrine signaling and paracrine action. Arch Biochem Biophys 2018; 660:108-120. [PMID: 30365934 DOI: 10.1016/j.abb.2018.10.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/08/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023]
Abstract
Poor cell viability after transplantation has restricted the therapeutic capacity of mesenchymal stem cells (MSCs) for cardiac dysfunction after myocardial infarction (MI). Growth arrest-specific gene 6 (Gas6) encodes a secreted γ-carboxyglutamic acid (Gla)-containing protein that functions in cell growth, adhesion, chemotaxis, mitogenesis and cell survival. In this study, we genetically modified MSCs with Gas6 and evaluated cell survival, cardiac function, and infarct size in a rat model of MI via intramyocardial delivery. Functional studies demonstrated that Gas6 transfer significantly reduced MSC apoptosis, increased survival of MSCs in vitro and in vivo, and that Gas6-engineered MSCs (MSCGas6)-treated animals had smaller infarct size and showed remarkably functional recovery as compared with control MSCs (MSCNull)-treated animals. Mechanistically, Gas6 could enhance phosphatidylinositol 3-kinase (PI3K)/Akt signaling and improve hypoxia-inducible factor-1 alpha (HIF-1α)-driven secretion of four major growth factors (VEGF, bFGF, SDF and IGF-1) in MSCs under hypoxia in an Axl-dependent autocrine manner. The paracrine action of MSCGas6 was further validated by coculture neonatal rat cardiomyocytes with conditioned medium from hypoxia-treated MSCGas6, as well as by pretreatment cardiomyocytes with the specific receptor inhibitors of VEGF, bFGF, SDF and IGF-1. Collectively, our data suggest that Gas6 may advance the efficacy of MSC therapy for post-infarcted heart failure via enhanced Gas6/Axl autocrine prosurvival signaling and paracrine cytoprotective action.
Collapse
|
16
|
Interplay between VEGF and Nrf2 regulates angiogenesis due to intracranial venous hypertension. Sci Rep 2016; 6:37338. [PMID: 27869147 PMCID: PMC5116754 DOI: 10.1038/srep37338] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/21/2016] [Indexed: 12/18/2022] Open
Abstract
Venous hypertension(VH) plays an important role in the pathogenesis of cerebral arteriovenous malformations (AVMs) and is closely associated with the HIF-1α/VEGF signaling pathway. Nuclear factor erythroid 2-related factor 2(Nrf2) significantly influences angiogenesis; however, the interplay between Nrf2 and VEGF under VH in brain AVMs remains unclear. Therefore, our study aimed to investigate the interplay between Nrf2 and VEGF due to VH in brain AVMs. Immunohistochemistry indicated that Nrf2 and VEGF were highly expressed in human brain AVM tissues. In vivo, we established a VH model in both wild-type (WT) and siRNA-mediated Nrf2 knockdown rats. VH significantly increased the expression of Nrf2 and VEGF. Loss of Nrf2 markedly inhibited the upregulation of VEGF, as determined by Western blot analysis and qRT-PCR. In vitro, primary brain microvascular endothelial cells (BMECs) were isolated from WT and Nrf2−/− mice, and a VEGF-Nrf2 positive feed-back loop was observed in BMECs. By trans well assay and angiogenesis assay, Nrf2 knockout significantly inhibited the migration and vascular tube formation of BMECs. These findings suggest that the interplay between Nrf2 and VEGF can contribute to VH-induced angiogenesis in brain AVMs pathogenesis.
Collapse
|
17
|
Kim H, Pawlikowska L, Su H, Young WL. Genetics and Vascular Biology of Angiogenesis and Vascular Malformations. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00012-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Yang ST, Rodriguez-Hernandez A, Walker EJ, Young WL, Su H, Lawton MT. Adult mouse venous hypertension model: common carotid artery to external jugular vein anastomosis. J Vis Exp 2015:50472. [PMID: 25650793 DOI: 10.3791/50472] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The understanding of the pathophysiology of brain arteriovenous malformations and arteriovenous fistulas has improved thanks to animal models. A rat model creating an artificial fistula between the common carotid artery (CCA) and the external jugular vein (EJV) has been widely described and proved technically feasible. This construct provokes a consistent cerebral venous hypertension (CVH), and therefore has helped studying the contribution of venous hypertension to formation, clinical symptoms, and prognosis of brain AVMs and dural AVFs. Equivalent mice models have been only scarcely described and have shown trouble with stenosis of the fistula. An established murine model would allow the study of not only pathophysiology but also potential genetic therapies for these cerebrovascular diseases. We present a model of arteriovenous fistula that produces a durable intracranial venous hypertension in the mouse. Microsurgical anastomosis of the murine CCA and EJV can be difficult due to diminutive anatomy and frequently result in a non-patent fistula. In this step-by-step protocol we address all the important challenges encountered during this procedure. Avoiding excessive retraction of the vein during the exposure, using 11-0 sutures instead of 10-0, and making a carefully planned end-to-side anastomosis are some of the critical steps. Although this method requires advanced microsurgical skills and a longer learning curve that the equivalent in the rat, it can be consistently developed. This novel model has been designed to integrate transgenic mouse techniques with a previously well-established experimental system that has proved useful to study brain AVMs and dural AVFs. By opening the possibility of using transgenic mice, a broader spectrum of valid models can be achieved and genetic treatments can also be tested. The experimental construct could also be further adapted to the study of other cerebrovascular diseases related with venous hypertension such as migraine, transient global amnesia, transient monocular blindness, etc.
Collapse
Affiliation(s)
- Shun-Tai Yang
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco
| | | | - Espen J Walker
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco
| | - William L Young
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco; Department of Neurological Surgery, University of California, San Francisco; Department of Neurology, University of California, San Francisco
| | - Hua Su
- Department of Anesthesia and Perioperative Care and Center for Cerebrovascular Research, University of California, San Francisco
| | - Michael T Lawton
- Department of Neurological Surgery, University of California, San Francisco;
| |
Collapse
|
19
|
Koyanagi M, Sakai N, Adachi H, Ueno Y, Kunieda T, Imamura H, Kikuchi H. “Loop-like formation” in the cortical venous reflux of dural arteriovenous fistula with intracranial hemorrhage. J Neuroradiol 2014; 41:316-21. [DOI: 10.1016/j.neurad.2013.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 11/07/2013] [Accepted: 11/13/2013] [Indexed: 10/25/2022]
Affiliation(s)
- Masaomi Koyanagi
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan.
| | - Nobuyuki Sakai
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan
| | - Hidemitsu Adachi
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan
| | - Yasushi Ueno
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan
| | - Takeharu Kunieda
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan
| | - Hirotoshi Imamura
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan
| | - Haruhiko Kikuchi
- Department of Neurosurgery, Kobe City Medical Center General Hospital; Kobe, Hyogo, Japan
| |
Collapse
|
20
|
Natural history, current concepts, classification, factors impacting endovascular therapy, and pathophysiology of cerebral and spinal dural arteriovenous fistulas. Clin Neurol Neurosurg 2014; 121:64-75. [DOI: 10.1016/j.clineuro.2014.01.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/12/2014] [Accepted: 01/19/2014] [Indexed: 11/24/2022]
|
21
|
Gutiérrez-González R, Pérez-Zamarron A, Rodríguez-Boto G. Normal perfusion pressure breakthrough phenomenon: experimental models. Neurosurg Rev 2014; 37:559-67. [PMID: 24777643 DOI: 10.1007/s10143-014-0549-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 08/25/2013] [Accepted: 02/08/2014] [Indexed: 11/28/2022]
Abstract
One of the most life-threatening complications after the obliteration of intracranial arteriovenous malformations is the development of oedema and/or multifocal haemorrhage. Two main theories have been postulated so far in order to explain this situation. On one hand, "normal perfusion pressure breakthrough phenomenon" is based on the loss of cerebral vessel autoregulation due to the chronic vasodilation of perinidal microcirculation. On the other hand, the "occlusive hyperaemia" deals with thrombotic and venous obstruction phenomena that may also generate such manifestations. The aim of this study is to resume the main concepts of the "normal perfusion pressure breakthrough phenomenon" theory as well as the related animal models described up to date, their advantages and disadvantages, and the main conclusions obtained as a result of the experimental research.
Collapse
Affiliation(s)
- Raquel Gutiérrez-González
- Department of Neurosurgery, Fundación Jiménez Díaz (IIS-FJD), Avda Reyes Católicos 2, 28040, Madrid, Spain,
| | | | | |
Collapse
|
22
|
Brain arteriovenous malformation modeling, pathogenesis, and novel therapeutic targets. Transl Stroke Res 2014; 5:316-29. [PMID: 24723256 DOI: 10.1007/s12975-014-0343-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 02/07/2023]
Abstract
Patients harboring brain arteriovenous malformation (bAVM) are at life-threatening risk of rupture and intracranial hemorrhage (ICH). The pathogenesis of bAVM has not been completely understood. Current treatment options are invasive, and ≈ 20 % of patients are not offered interventional therapy because of excessive treatment risk. There are no specific medical therapies to treat bAVMs. The lack of validated animal models has been an obstacle for testing hypotheses of bAVM pathogenesis and testing new therapies. In this review, we summarize bAVM model development and bAVM pathogenesis and potential therapeutic targets that have been identified during model development.
Collapse
|
23
|
Li Q, Zhang Q, Huang QH, Fang YB, Zhang ZL, Xu Y, Liu JM. A pivotal role of the vascular endothelial growth factor signaling pathway in the formation of venous hypertension-induced dural arteriovenous fistulas. Mol Med Rep 2014; 9:1551-8. [PMID: 24626343 PMCID: PMC4020488 DOI: 10.3892/mmr.2014.2037] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 01/24/2014] [Indexed: 11/17/2022] Open
Abstract
Dural arteriovenous fistulas (DAVFs) are associated with venous hypertension. Numerous studies have revealed high expression levels of vascular endothelial growth factor (VEGF) in human DAVF specimens, as well as in animal models of experimental venous hypertension. The objective of the present study was to clarify whether the VEGF signaling pathway is important in the development of DAVFs. Rats (n=216) were randomly divided into six groups. In the rats from five groups (groups A and C-E, n=45 in each group; group B, n=12), experimental venous hypertension was induced by right common carotid artery (CCA)-external jugular vein (EJV) anastomosis, superior sinus occlusion and left transver sinus occlusion, while the remaining group (group F, n=24) underwent sham surgery. The rats in group A received a VEGF recombinant adenovirus injection into the distal section of the right EJV 30 min prior to anastomosis of the CCA and EJV. An equivalent control adenovirus was injected into the right EJV of group B rats prior to anastomosis. The rats in group C received no virus prior to anastomosis and no medicine subsequent to surgery. The group D rats were lavaged with Vatalanib, a VEGF receptor (VEGFR) inhibitor, and the group E rats were lavaged with an equal quantity of saline weekly following surgery. Six rats from groups A-E and one rat from group F were sacrificed in the first, second, fourth and twelfth weeks after surgery for immunohistochemical analysis of VEGF expression and analysis of microvessel density. Cerebral angiography was performed on the remaining rats in each group on the twelfth week after surgery. The results revealed that following transfection with VEGF recombinant adenovirus, angiogenesis in the dura mater of venous hypertensive rats was increased subsequent to the increase in the VEGF expression levels of the brain and dura mater. The rate of DAVF induction by venous hypertension was significantly reduced by the VEGFR antagonist due to reduced angiogenesis in the dura mater. In conclusion, VEGF and its receptor may be important in the formation of venous hypertension-induced DAVFs.
Collapse
Affiliation(s)
- Qiang Li
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Qi Zhang
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Qing-Hai Huang
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Yi-Bin Fang
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Zhao-Long Zhang
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Yi Xu
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Jian-Min Liu
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
24
|
Lim CS, Kiriakidis S, Sandison A, Paleolog EM, Davies AH. Hypoxia-inducible factor pathway and diseases of the vascular wall. J Vasc Surg 2013; 58:219-30. [PMID: 23643279 DOI: 10.1016/j.jvs.2013.02.240] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 02/11/2013] [Accepted: 02/16/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Hypoxia may contribute to the pathogenesis of various diseases of the vascular wall. Hypoxia-inducible factors (HIFs) are nuclear transcriptional factors that regulate the transcription of genes that mediate cellular and tissue homeostatic responses to altered oxygenation. This article reviews the published literature on and discusses the role of the HIF pathway in diseases involving the vascular wall, including atherosclerosis, arterial aneurysms, pulmonary hypertension, vascular graft failure, chronic venous diseases, and vascular malformation. METHODS PubMed was searched with the terms "hypoxia-inducible factor" or "HIF" and "atherosclerosis," "carotid stenosis," "aneurysm," "pulmonary artery hypertension," "varicose veins," "venous thrombosis," "graft thrombosis," and "vascular malformation." RESULTS In atherosclerotic plaque, HIF-1α was localized in macrophages and smooth muscle cells bordering the necrotic core. Increased HIF-1α may contribute to atherosclerosis through alteration of smooth muscle cell proliferation and migration, angiogenesis, and lipid metabolism. The expression of HIF-1α is significantly elevated in aortic aneurysms compared with nonaneurysmal arteries. In pulmonary hypertension, HIF-1α contributes to the increase of intracellular K(+) and Ca(2+) leading to vasoconstriction of pulmonary smooth muscle cells. Alteration of the HIF pathway may contribute to vascular graft failure through the formation of intimal hyperplasia. In chronic venous disease, HIF pathway dysregulation contributes to formation of varicose veins and venous thromboembolism. However, whether the activation of the HIF pathway is protective or destructive to the venous wall is unclear. Increased activation of the HIF pathway causes aberrant expression of angiogenic factors contributing to the formation and maintenance of vascular malformations. CONCLUSIONS Pathologic vascular wall remodelling of many common diseases of the blood vessels has been found to be associated with altered activity of the HIF pathway. Therefore, understanding the role of the HIF pathway in diseases of the vascular wall is important to identify novel therapeutic strategies in the management of these pathologies.
Collapse
Affiliation(s)
- Chung S Lim
- Academic Section of Vascular Surgery, Department of Surgery and Cancer, Faculty of Medicine, Charing Cross Hospital Campus, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
25
|
Hetts SW, Keenan K, Fullerton HJ, Young WL, English JD, Gupta N, Dowd CF, Higashida RT, Lawton MT, Halbach VV. Pediatric intracranial nongalenic pial arteriovenous fistulas: clinical features, angioarchitecture, and outcomes. AJNR Am J Neuroradiol 2012; 33:1710-9. [PMID: 22766672 DOI: 10.3174/ajnr.a3194] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE NGAVFs are rare vascular malformations usually presenting in infancy or childhood. We sought to identify clinical and angiographic predictors of clinical outcome for these lesions. MATERIALS AND METHODS Retrospective review of a neurointerventional data base identified 386 pediatric patients with intracranial AVFs and AVMs, from which a cohort of 25 patients with NGAVF were selected for medical record and imaging analysis. RESULTS NGAVFs constituted 7.3% of pediatric intracranial vascular lesions with a nondural arteriovenous shunt. Seven of 8 patients who presented in the first month of life had CHF and harbored large, complex fistulas with multiple sites of arteriovenous shunting. Single-hole fistulas predominated later in childhood and more frequently presented with seizures, hemorrhage, or focal neurologic deficits. More treatment procedures were performed in subjects presenting at ≤ 2 years of age compared with older children (median = 3 versus 2, P = .041), and in those harboring a multi-hole fistula versus those with a single-hole fistula (median = 3 versus 2, P = .003). Eighteen patients (72%) had complete posttreatment elimination of NGAVF shunting. Compared with patients presenting at >2 years of age, patients presenting in the first 2 years of life were more likely to have a multi-hole fistula (100% versus 25%, P = .0001) and to have a poor clinical outcome (54% versus 0%, P = .0052), defined as a pediatric mRS of ≥ 3. CONCLUSIONS The morbidity of NGAVF appears higher than previously reported despite a somewhat higher rate of angiographic cure. Poor clinical outcome occurred primarily in patients with multi-hole NGAVFs presenting at ≤ 2 years of age.
Collapse
Affiliation(s)
- S W Hetts
- Department of Radiology, University of California-San Francisco, San Francisco, California, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Potential contribution of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 to blood-brain barrier disruption and brain edema after experimental subarachnoid hemorrhage. J Mol Neurosci 2012; 48:273-80. [PMID: 22528459 DOI: 10.1007/s12031-012-9769-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 04/01/2012] [Indexed: 01/27/2023]
Abstract
The current research aimed to investigate the role of hypoxia-inducible factor-1α (HIF-1α), aquaporin-4 (AQP-4), and matrix metalloproteinase-9 (MMP-9) in blood-brain barrier (BBB) dysfunction and cerebral edema formation in a rat subarachnoid hemorrhage (SAH) model. The SAH model was induced by injection of 0.3 ml fresh arterial, non-heparinized blood into the prechiasmatic cistern in 20 s. Anti-AQP-4 antibody, minocycline (an inhibitor of MMP-9), or 2-methoxyestradiol (an inhibitor of HIF-1α), was administered intravenously at 2 and 24 h after SAH. Brain samples were extracted at 48 h after SAH and examined for protein expressions, BBB impairment, and brain edema. Following SAH, remarkable edema and BBB extravasations were observed. Compared with the control group, the SAH animals have significantly upregulated expressions of HIF-1α, AQP-4, and MMP-9, in addition to decreased amounts of laminin and tight junction proteins. Brain edema was repressed after inhibition of AQP-4, MMP-9, or HIF-1α. Although BBB permeability was also ameliorated after inhibition of either HIF-1α or MMP-9, it was not modulated after inhibition of AQP-4. Inhibition of MMP-9 reversed the loss of laminin. Finally, inhibition of HIF-1α significantly suppressed the level of AQP-4 and MMP-9, which could induce the expression of laminin and tight junction proteins. Our results suggest that HIF-1α plays a role in brain edema formation and BBB disruption via a molecular signaling pathway involving AQP-4 and MMP-9. Pharmacological intervention of this pathway in patients with SAH may provide a novel therapeutic strategy for early brain injury.
Collapse
|
27
|
Cerebrovascular disorders: molecular insights and therapeutic opportunities. Nat Neurosci 2011; 14:1390-7. [DOI: 10.1038/nn.2947] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, Ding JY, Dornbos D, Li X, Guthikonda M, Rossi NF, Ding Y. The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg 2010; 114:92-101. [PMID: 20617879 DOI: 10.3171/2010.6.jns10207] [Citation(s) in RCA: 229] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECT The present study investigated the role of hypoxia-inducible factor-1α (HIF-1α), aquaporin-4 (AQP-4), and matrix metalloproteinase-9 (MMP-9) in blood-brain barrier (BBB) permeability alterations and brain edema formation in a rodent traumatic brain injury (TBI) model. METHODS The brains of adult male Sprague-Dawley rats (400-425 g) were injured using the Marmarou closed-head force impact model. Anti-AQP-4 antibody, minocycline (an inhibitor of MMP-9), or 2-methoxyestradiol (2ME2, an inhibitor of HIF-1α), was administered intravenously 30 minutes after injury. The rats were killed 24 hours after injury and their brains were examined for protein expression, BBB permeability, and brain edema. Expression of HIF-1α, AQP-4, and MMP-9 as well as expression of the vascular basal lamina protein (laminin) and tight junction proteins (zona occludens-1 and occludin) was determined by Western blotting. Blood-brain barrier disruption was assessed by FITC-dextran extravasation, and brain edema was measured by the brain water content. RESULTS Significant (p < 0.05) edema and BBB extravasations were observed following TBI induction. Compared with sham-operated controls, the injured animals were found to have significantly (p < 0.05) enhanced expression of HIF-1α, AQP-4, and MMP-9, in addition to reduced amounts (p < 0.05) of laminin and tight junction proteins. Edema was significantly (p < 0.01) decreased after inhibition of AQP-4, MMP-9, or HIF-1α. While BBB permeability was significantly (p < 0.01) ameliorated after inhibition of either HIF-1α or MMP-9, it was not affected following inhibition of AQP-4. Inhibition of MMP reversed the loss of laminin (p < 0.01). Finally, while inhibition of HIF-1α significantly (p < 0.05) suppressed the expression of AQP-4 and MMP-9, such inhibition significantly (p < 0.05) increased the expression of laminin and tight junction proteins. CONCLUSIONS The data support the notion that HIF-1α plays a role in brain edema formation and BBB disruption via a molecular pathway cascade involving AQP-4 and MMP-9. Pharmacological blockade of this pathway in patients with TBI may provide a novel therapeutic strategy.
Collapse
Affiliation(s)
- Tetsuhiro Higashida
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|