1
|
Garcia-Martínez T, Gornatti DG, Ortiz M, Cañellas G, Heine-Suñer D, Vives-Bauzà C. The Triad of Blood-Brain Barrier Integrity: Endothelial Cells, Astrocytes, and Pericytes in Perinatal Stroke Pathophysiology. Int J Mol Sci 2025; 26:1886. [PMID: 40076511 PMCID: PMC11900453 DOI: 10.3390/ijms26051886] [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: 01/29/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pediatric stroke, a significant cause of long-term neurological deficits in children, often arises from disruptions within neurovascular unit (NVU) components. The NVU, a dynamic ensemble of astrocytes, endothelial cells, pericytes, and microglia, is vital for maintaining cerebral homeostasis and regulating vascular brain development. Its structural integrity, particularly at the blood-brain barrier (BBB), depends on intercellular junctions and the basement membrane, which together restrict paracellular transport and shield the brain from systemic insults. Dysfunction in this intricate system is increasingly linked to pediatric stroke and related cerebrovascular conditions. Mutations disrupting endothelial cell adhesion or pericyte-endothelial interactions can compromise BBB stability, leading to pathological outcomes such as intraventricular hemorrhage in the germinal matrix, a hallmark of vascular brain immaturity. Additionally, inflammation, ferroptosis, necroptosis, and autophagy are key cellular processes influencing brain damage and repair. Excessive activation of these mechanisms can exacerbate NVU injury, whereas targeted therapeutic modulation offers potential pathways to mitigate damage and support recovery. This review explores the cellular and molecular mechanisms underlying NVU dysfunction, BBB disruption, and subsequent brain injury in pediatric stroke. Understanding the interplay between genetic mutations, environmental stressors, and NVU dynamics provides new insights into stroke pathogenesis. The susceptibility of the germinal matrix to vascular rupture further emphasizes the critical role of NVU integrity in early brain development. Targeting inflammatory pathways and cell death mechanisms presents promising strategies to preserve NVU function and improve outcomes for affected neonates.
Collapse
Affiliation(s)
- Tania Garcia-Martínez
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Denise G. Gornatti
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Marina Ortiz
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
| | - Guillem Cañellas
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), 07122 Palma, Spain
| | - Damià Heine-Suñer
- Genomics of Health Research, Unit of Molecular Diagnostics and Clinical Genetics, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07013 Palma, Spain;
| | - Cristòfol Vives-Bauzà
- Neurobiology, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain; (T.G.-M.); (D.G.G.); (M.O.); (G.C.)
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), 07122 Palma, Spain
| |
Collapse
|
2
|
Al-Eitan LN, Alahmad SZ, Ajeen SA, Altawil AY, Khair IY, Kharmah HSA, Alghamdi MA. Evaluation of the metabolic activity, angiogenic impacts, and GSK-3β signaling of the synthetic cannabinoid MMB-2201 on human cerebral microvascular endothelial cells. J Cannabis Res 2024; 6:43. [PMID: 39707578 DOI: 10.1186/s42238-024-00255-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
Angiogenesis is an intrinsic physiological process involving the formation of new capillaries from existing ones. Synthetic cannabinoids refer to a class of human-made chemicals that are primarily designed to mimic the effects of delta-9-tetrahydrocannabinol, the primary psychoactive compound in cannabis. Studies investigating the association between synthetic cannabinoids and cellular reactions are limited, and the available scientific evidence is insufficient. Consequently, the primary goal was to examine the effects of the synthetic cannabinoid MDMB-2201 on brain angiogenesis in vitro to provide a comprehensive analysis of MMB-2201's potential therapeutic or adverse effects on vascular development and related health conditions. Human Cerebral Microvascular Endothelial Cells (HBEC-5i) were incubated with MMB-2201, and their metabolic activity, migration rate, and tubular structure formation were examined. Expression levels of several angiogenesis-related proteins such as vascular endothelial growth factor (VEGF), Angiopoietin-1 (ANG-1), and Angiopoietin-2 (ANG-2) were assessed using western blot, ELISA, and real-time PCR. Furthermore, the phosphorylation of glycogen synthase kinase 3 beta (GSK-3β) at Ser9 induced by MMB-2201 was evaluated. HBEC-5i cells showed a significant increase in metabolic rate, enhanced migration, and sprouting of brain endothelial cells. Moreover, there was a noticeable increase in the mRNA and protein levels of VEGF, ANG-1, and ANG-2, as well as in the phosphorylation rate of GSK-3β at Ser9. This study paves the way for a novel pharmacological approach to addressing various angiogenesis-related diseases by targeting cannabinoid receptor type-1. Further exploration using different antagonists or agonists of cannabinoid receptors, depending on the specific characteristics of the disorders, may be necessary.
Collapse
Affiliation(s)
- Laith Naser Al-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Saif Zuhair Alahmad
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Sufyan Ali Ajeen
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Ahmad Younis Altawil
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Iliya Yacoub Khair
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Hana Salah Abu Kharmah
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Mansour Abdullah Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 62529, Saudi Arabia
- Genomics and Personalized Medicine Unit, The Centre for Medical and Health Research, King Khalid University, Abha, 62529, Saudi Arabia
| |
Collapse
|
3
|
Khodaverdi K, Bakhshi A, Mozafari MR, Naghib SM. A review of chitosan-based nanocarriers as drug delivery systems for brain diseases: Critical challenges, outlooks and promises. Int J Biol Macromol 2024; 278:134962. [PMID: 39179064 DOI: 10.1016/j.ijbiomac.2024.134962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
The administration of medicinal drugs orally or systemically limits the treatment of specific central nervous system (CNS) illnesses, such as certain types of brain cancers. These methods can lead to severe adverse reactions and inadequate transport of drugs to the brain, resulting in limited effectiveness. The CNS homeostasis is maintained by various barriers within the brain, such as the endothelial, epithelial, mesothelial, and glial barriers, which strictly control the movement of chemicals, solutes, and immune cells. Brain capillaries consist of endothelial cells (ECs) and perivascular pericytes, with pericytes playing a crucial role in maintaining the blood-brain barrier (BBB), influencing new blood vessel formation, and exhibiting secretory capabilities. This article summarizes the structural components and anatomical characteristics of the BBB. Intranasal administration, a non-invasive method, allows drugs to reach the brain by bypassing the BBB, while direct cerebral administration targets specific brain regions with high concentrations of therapeutic drugs. Technical and mechanical tools now exist to bypass the BBB, enabling the development of more potent and safer medications for neurological disorders. This review also covers clinical trials, formulations, challenges, and patents for a comprehensive perspective.
Collapse
Affiliation(s)
- Khashayar Khodaverdi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Ali Bakhshi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran; Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia; Biomaterials and Tissue Engineering Research Group, Department of Interdisciplinary Technologies, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| |
Collapse
|
4
|
Grinchevskaya LR, Salikhova DI, Silachev DN, Goldshtein DV. Neural and Glial Regulation of Angiogenesis in CNS in Ischemic Stroke. Bull Exp Biol Med 2024:10.1007/s10517-024-06219-4. [PMID: 39266920 DOI: 10.1007/s10517-024-06219-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Indexed: 09/14/2024]
Abstract
CNS diseases associated with compromised blood supply and/or vascular integrity are one of the leading causes of mortality and disability in adults worldwide and are also among 10 most common causes of death in children. Angiogenesis is an essential element of regeneration processes upon nervous tissue damage and can play a crucial role in neuroprotection. Here we review the features of cerebral vascular regeneration after ischemic stroke, including the complex interactions between endothelial cells and other brain cell types (neural stem cells, astrocytes, microglia, and oligodendrocytes). The mechanisms of reciprocal influence of angiogenesis and neurogenesis, the role of astrocytes in the formation of the blood-brain barrier, and roles of microglia and oligodendrocytes in vascular regeneration are discussed. Understanding the mechanisms of angiogenesis regulation in CNS is of critical importance for the development of new treatments of neurovascular pathologies.
Collapse
Affiliation(s)
- L R Grinchevskaya
- Research Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University, Moscow, Russia
| | - D I Salikhova
- Research Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University, Moscow, Russia.
- Research Centre for Medical Genetics, Moscow, Russia.
| | - D N Silachev
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - D V Goldshtein
- Research Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University, Moscow, Russia
- Research Centre for Medical Genetics, Moscow, Russia
| |
Collapse
|
5
|
Rizzuti M, Melzi V, Brambilla L, Quetti L, Sali L, Ottoboni L, Meneri M, Ratti A, Verde F, Ticozzi N, Comi GP, Corti S, Abati E. Shaping the Neurovascular Unit Exploiting Human Brain Organoids. Mol Neurobiol 2024; 61:6642-6657. [PMID: 38334812 PMCID: PMC11338975 DOI: 10.1007/s12035-024-03998-9] [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: 04/14/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Brain organoids, three-dimensional cell structures derived from pluripotent stem cells, closely mimic key aspects of the human brain in vitro, providing a powerful tool for studying neurodevelopment and disease. The neuroectodermal induction protocol employed for brain organoid generation primarily gives rise to the neural cellular component but lacks the vital vascular system, which is crucial for the brain functions by regulating differentiation, migration, and circuit formation, as well as delivering oxygen and nutrients. Many neurological diseases are caused by dysfunctions of cerebral microcirculation, making vascularization of human brain organoids an important tool for pathogenetic and translational research. Experimentally, the creation of vascularized brain organoids has primarily focused on the fusion of vascular and brain organoids, on organoid transplantation in vivo, and on the use of microfluidic devices to replicate the intricate microenvironment of the human brain in vitro. This review summarizes these efforts and highlights the importance of studying the neurovascular unit in a forward-looking perspective of leveraging their use for understanding and treating neurological disorders.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linda Ottoboni
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Nicola Ticozzi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
6
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
7
|
Liu D, Liao P, Li H, Tong S, Wang B, Lu Y, Gao Y, Huang Y, Zhou H, Shi L, Papadimitriou J, Zong Y, Yuan J, Chen P, Chen Z, Ding P, Zheng Y, Zhang C, Zheng M, Gao J. Regulation of blood-brain barrier integrity by Dmp1-expressing astrocytes through mitochondrial transfer. SCIENCE ADVANCES 2024; 10:eadk2913. [PMID: 38941455 PMCID: PMC11212732 DOI: 10.1126/sciadv.adk2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/24/2024] [Indexed: 06/30/2024]
Abstract
The blood-brain barrier (BBB) acts as the crucial physical filtration structure in the central nervous system. Here, we investigate the role of a specific subset of astrocytes in the regulation of BBB integrity. We showed that Dmp1-expressing astrocytes transfer mitochondria to endothelial cells via their endfeet for maintaining BBB integrity. Deletion of the Mitofusin 2 (Mfn2) gene in Dmp1-expressing astrocytes inhibited the mitochondrial transfer and caused BBB leakage. In addition, the decrease of MFN2 in astrocytes contributes to the age-associated reduction of mitochondrial transfer efficiency and thus compromises the integrity of BBB. Together, we describe a mechanism in which astrocytes regulate BBB integrity through mitochondrial transfer. Our findings provide innnovative insights into the cellular framework that underpins the progressive breakdown of BBB associated with aging and disease.
Collapse
Affiliation(s)
- Delin Liu
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Sihan Tong
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Bingqi Wang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yafei Lu
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yigang Huang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Linjing Shi
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - John Papadimitriou
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Department of Pathology, Pathwest, Nedlands, Western Australia 6009, Australia
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Jun Yuan
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Peilin Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Ziming Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yongqiang Zheng
- Department of Orthopaedics, Jinjiang Municipal Hospital, Jinjiang, Fujian Province, 362200, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Orthopaedics, Jinjiang Municipal Hospital, Jinjiang, Fujian Province, 362200, China
| |
Collapse
|
8
|
Biouss G, Antounians L, Aguet J, Kopcalic K, Fakhari N, Baranger J, Mertens L, Villemain O, Zani A. The brain of fetuses with congenital diaphragmatic hernia shows signs of hypoxic injury with loss of progenitor cells, neurons, and oligodendrocytes. Sci Rep 2024; 14:13680. [PMID: 38871804 DOI: 10.1038/s41598-024-64412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a birth defect characterized by incomplete closure of the diaphragm, herniation of abdominal organs into the chest, and compression of the lungs and the heart. Besides complications related to pulmonary hypoplasia, 1 in 4 survivors develop neurodevelopmental impairment, whose etiology remains unclear. Using a fetal rat model of CDH, we demonstrated that the compression exerted by herniated organs on the mediastinal structures results in decreased brain perfusion on ultrafast ultrasound, cerebral hypoxia with compensatory angiogenesis, mature neuron and oligodendrocyte loss, and activated microglia. In CDH fetuses, apoptosis was prominent in the subventricular and subgranular zones, areas that are key for neurogenesis. We validated these findings in the autopsy samples of four human fetuses with CDH compared to age- and sex-matched controls. This study reveals the molecular mechanisms and cellular changes that occur in the brain of fetuses with CDH and creates opportunities for therapeutic targets.
Collapse
Affiliation(s)
- George Biouss
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Julien Aguet
- Department of Diagnostic Imaging, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Medical Imaging, University of Toronto, 263 McCaul Street, Toronto, ON, M5T 1W7, Canada
| | - Katarina Kopcalic
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Nikan Fakhari
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Jerome Baranger
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Luc Mertens
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Olivier Villemain
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Surgery, University of Toronto, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
9
|
Zhang S, Cheng Y, Guan Y, Wen J, Chen Z. Hydrogen Sulfide Exerted a Pro-Angiogenic Role by Promoting the Phosphorylation of VEGFR2 at Tyr797 and Ser799 Sites in Hypoxia-Reoxygenation Injury. Int J Mol Sci 2024; 25:4340. [PMID: 38673925 PMCID: PMC11050214 DOI: 10.3390/ijms25084340] [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: 03/05/2024] [Revised: 03/25/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The protective effects of hydrogen sulfide (H2S) against ischemic brain injury and its role in promoting angiogenesis have been established. However, the specific mechanism underlying these effects remains unclear. This study is designed to investigate the regulatory impact and mechanism of H2S on VEGFR2 phosphorylation. Following expression and purification, the recombinant His-VEGFR2 protein was subjected to LC-PRM/MS analysis to identify the phosphorylation sites of VEGFR2 upon NaHS treatment. Adenovirus infection was used to transfect primary rat brain artery endothelial cells (BAECs) with the Ad-VEGFR2WT, Ad-VEGFR2Y797F, and Ad-VEGFR2S799A plasmids. The expression of VEGFR2 and recombinant Flag-VEGFR2, along with Akt phosphorylation, cell proliferation, and LDH levels, was assessed. The migratory capacity and tube-forming potential of BAECs were assessed using wound healing, transwell, and tube formation assays. NaHS notably enhanced the phosphorylation of VEGFR2 at Tyr797 and Ser799 sites. These phosphorylation sites were identified as crucial for mediating the protective effects of NaHS against hypoxia-reoxygenation (H/R) injury. NaHS significantly enhanced the Akt phosphorylation, migratory capacity, and tube formation of BAECs and upregulated the expression of VEGFR2 and recombinant proteins. These findings suggest that Tyr797 and Ser799 sites of VEGFR2 serve as crucial mediators of H2S-induced pro-angiogenic effects and protection against H/R injury.
Collapse
Affiliation(s)
- Sen Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (S.Z.); (Y.G.)
| | - Yongfeng Cheng
- Clinical Medical College, Anhui Medical University, Hefei 230012, China;
| | - Yining Guan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (S.Z.); (Y.G.)
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (S.Z.); (Y.G.)
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (S.Z.); (Y.G.)
| |
Collapse
|
10
|
Margetts TJ, Wang HS, Karnik SJ, Plotkin LI, Movila A, Oblak AL, Fehrenbacher JC, Kacena MA. From the Mind to the Spine: The Intersecting World of Alzheimer's and Osteoporosis. Curr Osteoporos Rep 2024; 22:152-164. [PMID: 38334917 PMCID: PMC10912148 DOI: 10.1007/s11914-023-00848-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 02/10/2024]
Abstract
PURPOSE OF REVIEW This comprehensive review delves into the intricate interplay between Alzheimer's disease (AD) and osteoporosis, two prevalent conditions with significant implications for individuals' quality of life. The purpose is to explore their bidirectional association, underpinned by common pathological processes such as aging, genetic factors, inflammation, and estrogen deficiency. RECENT FINDINGS Recent advances have shown promise in treating both Alzheimer's disease (AD) and osteoporosis by targeting disease-specific proteins and bone metabolism regulators. Monoclonal antibodies against beta-amyloid and tau for AD, as well as RANKL and sclerostin for osteoporosis, have displayed therapeutic potential. Additionally, ongoing research has identified neuroinflammatory genes shared between AD and osteoporosis, offering insight into the interconnected inflammatory mechanisms. This knowledge opens avenues for innovative dual-purpose therapies that could address both conditions, potentially revolutionizing treatment approaches for AD and osteoporosis simultaneously. This review underscores the potential for groundbreaking advancements in early diagnosis and treatment by unraveling the intricate connection between AD and bone health. It advocates for a holistic, patient-centered approach to medical care that considers both cognitive and bone health, ultimately aiming to enhance the overall well-being of individuals affected by these conditions. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Tyler J Margetts
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah S Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lilian I Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
| | - Alexandru Movila
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, 46202, USA
| | - Adrian L Oblak
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| |
Collapse
|
11
|
Stankovic I, Notaras M, Wolujewicz P, Lu T, Lis R, Ross ME, Colak D. Schizophrenia endothelial cells exhibit higher permeability and altered angiogenesis patterns in patient-derived organoids. Transl Psychiatry 2024; 14:53. [PMID: 38263175 PMCID: PMC10806043 DOI: 10.1038/s41398-024-02740-2] [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: 07/10/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024] Open
Abstract
Schizophrenia (SCZ) is a complex neurodevelopmental disorder characterized by the manifestation of psychiatric symptoms in early adulthood. While many research avenues into the origins of SCZ during brain development have been explored, the contribution of endothelial/vascular dysfunction to the disease remains largely elusive. To model the neuropathology of SCZ during early critical periods of brain development, we utilized patient-derived induced pluripotent stem cells (iPSCs) to generate 3D cerebral organoids and define cell-specific signatures of disease. Single-cell RNA sequencing revealed that while SCZ organoids were similar in their macromolecular diversity to organoids generated from healthy controls (CTRL), SCZ organoids exhibited a higher percentage of endothelial cells when normalized to total cell numbers. Additionally, when compared to CTRL, differential gene expression analysis revealed a significant enrichment in genes that function in vessel formation, vascular regulation, and inflammatory response in SCZ endothelial cells. In line with these findings, data from 23 donors demonstrated that PECAM1+ microvascular vessel-like structures were increased in length and number in SCZ organoids in comparison to CTRL organoids. Furthermore, we report that patient-derived endothelial cells displayed higher paracellular permeability, implicating elevated vascular activity. Collectively, our data identified altered gene expression patterns, vessel-like structural changes, and enhanced permeability of endothelial cells in patient-derived models of SCZ. Hence, brain microvascular cells could play a role in the etiology of SCZ by modulating the permeability of the developing blood brain barrier (BBB).
Collapse
Affiliation(s)
- Isidora Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Paul Wolujewicz
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Tyler Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Raphael Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
12
|
Harry GJ. Developmental Associations between Neurovascularization and Microglia Colonization. Int J Mol Sci 2024; 25:1281. [PMID: 38279280 PMCID: PMC10816009 DOI: 10.3390/ijms25021281] [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: 12/30/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
The temporal and spatial pattern of microglia colonization and vascular infiltration of the nervous system implies critical associated roles in early stages of nervous system development. Adding to existing reviews that cover a broad spectrum of the various roles of microglia during brain development, the current review will focus on the developmental ontogeny and interdependency between the colonization of the nervous system with yolk sac derived macrophages and vascularization. Gaining a better understanding of the timing and the interdependency of these two processes will significantly contribute to the interpretation of data generated regarding alterations in either process during early development. Additionally, such knowledge should provide a framework for understanding the influence of the early gestational environmental and the impact of genetics, disease, disorders, or exposures on the early developing nervous system and the potential for long-term and life-time effects.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
13
|
Liu Z, Zhang J, Li X, Hu Q, Chen X, Luo L, Ai L, Ye J. Astrocytic expression of Yes-associated protein (YAP) regulates retinal neovascularization in a mouse model of oxygen-induced retinopathy. Microvasc Res 2024; 151:104611. [PMID: 37774941 DOI: 10.1016/j.mvr.2023.104611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Pathological neovascularization is the hallmark of many vascular oculopathies. There is still a great deal of uncertainty surrounding retinal neovascularization research. A working hypothesis that astrocytic Yes-associated protein (YAP) act as a key factor in retinal neovascularization was proposed. And our study was conducted to verified this hypothesis. In vivo, we successfully generated mice deficient in YAP in astrocytes (YAPf/f GFAP-Cre mice) and set up oxygen-induced retinopathy (OIR) model. Pathological neovascularization was evaluated by immunofluorescence staining and western blotting. In vitro, cultured retinal astrocytes were transfected with YAP siRNA. Enzyme-linked immunosorbent assay (ELISA) and western blot were used to determine the proteins in the supernatants and cells. The results showed that YAP was upregulated and activated in the OIR mice retinas. Conditional ablation of YAP aggravated pathological neovascularization, along with the upregulation of vascular endothelial growth factor A (VEGF-A) and monocyte chemoattractant protein-1 (MCP-1). Studies in vitro confirmed that the knockdown of YAP in astrocytes lead to increases in VEGF-A and MCP-1 levels, thus enhancing pro-angiogenic capability of YAP-deficit astrocytes. In conclusion, astrocytic YAP alleviates retinal pathological angiogenesis by inhibiting the over-activation of astrocytes, which suppresses excessive VEGF-A production and neuroinflammation.
Collapse
Affiliation(s)
- Zhifei Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jieqiong Zhang
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xue Li
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Qiumei Hu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Linlin Luo
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Liqianyu Ai
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Jian Ye
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| |
Collapse
|
14
|
Harry GJ. Microglia Colonization Associated with Angiogenesis and Neural Cell Development. ADVANCES IN NEUROBIOLOGY 2024; 37:163-178. [PMID: 39207692 DOI: 10.1007/978-3-031-55529-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The temporal and spatial pattern of microglia colonization of the nervous system implies a role in early stages of organ development including cell proliferation, differentiation, and neurovascularization. As microglia colonize and establish within the developing nervous system, they assume a neural-specific identity and contribute to key developmental events. Their association around blood vessels implicates them in development of the vascular system or vice versa. A similar association has been reported for neural cell proliferation and associated phenotypic shifts and for cell fate differentiation to neuronal or glial phenotypes. These processes are accomplished by phagocytic activities, cell-cell contact relationships, and secretion of various factors. This chapter will present data currently available from studies evaluating the dynamic and interactive nature of these processes throughout the progression of nervous system development.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
15
|
Deng H, Liu Y, Shi Z, Yang J, Liu C, Mei X. Zinc regulates a specific subpopulation of VEGFA + microglia to improve the hypoxic microenvironment for functional recovery after spinal cord injury. Int Immunopharmacol 2023; 125:111092. [PMID: 37883817 DOI: 10.1016/j.intimp.2023.111092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a central nervous system injury that is primarily traumatic and manifests as autonomic dysfunction below the level of injury. Our previous studies have found that zinc ions have important effects on the nervous system and nerve repair, promoting autophagy and reducing inflammatory responses. However, the role of zinc ions in vascular regeneration is unclear. AIMS We investigated the effect of zinc ions after spinal cord injury from the perspective of a hypoxic microenvironment, and elucidated the role of VEGF-A secreted by microglia for vascular regeneration after spinal cord injury, providing new ideas for the treatment of spinal cord injury. RESULTS Zinc promotes functional recovery after spinal cord injury by regulating VEGF-A secretion from microglia. On the one hand, VEGF-A secreted by microglia promotes angiogenesis through the PI3K/AKT/Bcl-2 pathway and improves the hypoxic microenvironment after spinal cord injury. On the other hand, VEGF-A secreted by microglia was positively correlated with platelet endothelial cell adhesion molecule-1 (CD31), and zinc could increase the association between microglia and blood vessels. CONCLUSION Zinc promoted microglia secretion of VEGF-A, increased vascular endothelial cell proliferation and migration through the PI3K/AKT/Bcl-2 pathway, and inhibited microglia apoptosis.
Collapse
Affiliation(s)
- Hao Deng
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Yu Liu
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Zuqiang Shi
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Jing Yang
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Chang Liu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China.
| | - Xifan Mei
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China.
| |
Collapse
|
16
|
Sun XY, Ju XC, Zhao HF, You ZW, Han RR, Luo ZG. Generation of Human Blood Vessel and Vascularized Cerebral Organoids. Bio Protoc 2023; 13:e4870. [PMID: 37969757 PMCID: PMC10632161 DOI: 10.21769/bioprotoc.4870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 11/17/2023] Open
Abstract
Brain organoids have been widely used to study diseases and the development of the nervous system. Many reports have investigated the application of brain organoids, but most of these models lack vascular structures, which play essential roles in brain development and neurological diseases. The brain and blood vessels originate from two different germ layers, making it difficult to induce vascularized brain organoids in vitro. We developed this protocol to generate brain-specific blood vessel and cerebral organoids and then fused them at a specific developmental time point. The fused cerebral organoids exhibited robust vascular network-like structures, which allows simulating the in vivo developmental processes of the brain for further applications in various neurological diseases. Key Features • Culturing vascularized brain organoids using human embryonic stem cells (hESCs). • The new approach generates not only neural cells and vessel-like networks but also brain-resident microglia immune cells in a single organoid.
Collapse
Affiliation(s)
- Xin-Yao Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiang-Chun Ju
- Okinawa Institute of Science and Technology, Onna-son, Japan
| | - Hong-Fang Zhao
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Wen You
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Run-Run Han
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
17
|
AL-Eitan LN, Alahmad SZ, ElMotasem MFM, Alghamdi MA. The synthetic cannabinoid 5F-MDMB-PICA enhances the metabolic activity and angiogenesis in human brain microvascular endothelial cells by upregulation of VEGF, ANG-1, and ANG-2. Toxicol Res (Camb) 2023; 12:796-806. [PMID: 37915478 PMCID: PMC10615825 DOI: 10.1093/toxres/tfad068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 11/03/2023] Open
Abstract
Brain angiogenesis, the formation of new blood vessels from existing brain vasculature, has been previously associated with neural plasticity and addictive behaviors related to substances. Synthetic cannabinoids (SCs) have become increasingly popular due to their ability to mimic the effects of cannabis, offering high potency and easy accessibility. In the current study, we reveal that the SC 5F-MDMB-PICA, the most common SC in the United States in 2019, increases cell metabolic activity and promotes angiogenesis in human brain microvascular endothelial cells (HBMECs). First, we performed an MTT assay to evaluate the effects of 5F-MDMB-PICA treatment at various concentrations (0.0001 μM, 0.001 μM, 0.01 μM, 0.1 μM, and 1 μM) on HBMECs metabolic activity. The results demonstrated higher concentrations of the SC improved cell metabolic activity. Furthermore, 5F-MDMB-PICA treatment enhanced tube formation and migration of HBMECs in a dosage-dependent manner. Additionally, the mRNA, secreted protein, and intracellular protein levels of vascular endothelial growth factor, angiopoietin-1, and angiopoietin-2, which are involved in the regulation of angiogenesis, as well as the protein levels of cannabinoid receptor type-1, were all increased following treatment with 5F-MDMB-PICA. Notably, the phosphorylation levels at Serine 9 residue of glycogen synthase kinase-3β were also increased in the 5F-MDMB-PICA treated HBMECs. Collectively, our findings demonstrate that 5F-MDMB-PICA can enhance angiogenesis in HBMECs, suggesting the significant role of angiogenesis in the response to SCs. Manipulating this interaction may pave the way for innovative treatments targeting SC addiction and angiogenesis-related conditions.
Collapse
Affiliation(s)
- Laith Naser AL-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Saif Zuhair Alahmad
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohd Fahmi Munib ElMotasem
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mansour Abdullah Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| |
Collapse
|
18
|
Liang JH, Akhanov V, Ho A, Tawfik M, D'Souza SP, Cameron MA, Lang RA, Samuel MA. Dopamine signaling from ganglion cells directs layer-specific angiogenesis in the retina. Curr Biol 2023; 33:3821-3834.e5. [PMID: 37572663 PMCID: PMC10529464 DOI: 10.1016/j.cub.2023.07.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/26/2023] [Accepted: 07/20/2023] [Indexed: 08/14/2023]
Abstract
During central nervous system (CNS) development, a precisely patterned vasculature emerges to support CNS function. How neurons control angiogenesis is not well understood. Here, we show that the neuromodulator dopamine restricts vascular development in the retina via temporally limited production by an unexpected neuron subset. Our genetic and pharmacological experiments demonstrate that elevating dopamine levels inhibits tip-cell sprouting and vessel growth, whereas reducing dopamine production by all retina neurons increases growth. Dopamine production by canonical dopaminergic amacrine interneurons is dispensable for these events. Instead, we found that temporally restricted dopamine production by retinal ganglion cells (RGCs) modulates vascular development. RGCs produce dopamine precisely during angiogenic periods. Genetically limiting dopamine production by ganglion cells, but not amacrines, decreases angiogenesis. Conversely, elevating ganglion-cell-derived dopamine production inhibits early vessel growth. These vasculature outcomes occur downstream of vascular endothelial growth factor receptor (VEGFR) activation and Notch-Jagged1 signaling. Jagged1 is increased and subsequently inhibits Notch signaling when ganglion cell dopamine production is reduced. Our findings demonstrate that dopaminergic neural activity from a small neuron subset functions upstream of VEGFR to serve as developmental timing cue that regulates vessel growth.
Collapse
Affiliation(s)
- Justine H Liang
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Viktor Akhanov
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Anthony Ho
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mohamed Tawfik
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shane P D'Souza
- Divisions of Pediatric Ophthalmology and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Morven A Cameron
- School of Medicine, Western Sydney University, Western Sydney University Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Richard A Lang
- Divisions of Pediatric Ophthalmology and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Ophthalmology, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Rejdak K, Sienkiewicz-Jarosz H, Bienkowski P, Alvarez A. Modulation of neurotrophic factors in the treatment of dementia, stroke and TBI: Effects of Cerebrolysin. Med Res Rev 2023; 43:1668-1700. [PMID: 37052231 DOI: 10.1002/med.21960] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophic factors (NTFs) are involved in the pathophysiology of neurological disorders such as dementia, stroke and traumatic brain injury (TBI), and constitute molecular targets of high interest for the therapy of these pathologies. In this review we provide an overview of current knowledge of the definition, discovery and mode of action of five NTFs, nerve growth factor, insulin-like growth factor 1, brain derived NTF, vascular endothelial growth factor and tumor necrosis factor alpha; as well as on their contribution to brain pathology and potential therapeutic use in dementia, stroke and TBI. Within the concept of NTFs in the treatment of these pathologies, we also review the neuropeptide preparation Cerebrolysin, which has been shown to resemble the activities of NTFs and to modulate the expression level of endogenous NTFs. Cerebrolysin has demonstrated beneficial treatment capabilities in vitro and in clinical studies, which are discussed within the context of the biochemistry of NTFs. The review focuses on the interactions of different NTFs, rather than addressing a single NTF, by outlining their signaling network and by reviewing their effect on clinical outcome in prevalent brain pathologies. The effects of the interactions of these NTFs and Cerebrolysin on neuroplasticity, neurogenesis, angiogenesis and inflammation, and their relevance for the treatment of dementia, stroke and TBI are summarized.
Collapse
Affiliation(s)
- Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | | | | | - Anton Alvarez
- Medinova Institute of Neurosciences, Clinica RehaSalud, Coruña, Spain
| |
Collapse
|
20
|
Tskitishvili E, Palazzo C, Foidart JM, Piel G, Pequeux C. Use of Liposome-encapsulated estetrol for treatment of Neonatal Hypoxic-Ischemic encephalopathy. Brain Res 2023; 1809:148369. [PMID: 37061081 DOI: 10.1016/j.brainres.2023.148369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/11/2022] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Estetrol (E4) is a natural estrogen synthesized only during pregnancy. It has strong neuroprotective and antioxidative activities. The aim of the present study was to define the neuroprotective potency of E4 encapsulated either in liposome (Lipo-E4) or in drug-in cyclodextrin (HP-β-CD) in liposome (DCL) system, and compare them with a single use of E4. In vitro studies were performed in an oxidative stress model of primary hippocampal neuronal cell cultures, followed by the lactate dehydrogenase activity and cell proliferation assays. In vivo studies were conducted by using a model of neonatal hypoxic-ischemic encephalopathy in immature rat pups. Brain samples were studied by (immuno)histochemistry for the detection of survived cells, expression of microtubule-associated protein-2, myelin basic protein, doublecortin and vascular-endothelial growth factor. Concentrations of glial fibrillary acidic protein in blood serum were studied by ELISA. In vitro, cell proliferation was significantly up-regulated in cultures treated either by DCL-E4 or E4 compared to the control cells, whereas DCL-E4 treated cells had significantly higher survival rate than the cells treated by E4 alone. Evaluation of brain samples showed that DCL-E4 and a high dose of E4 alone significantly preserve the grey and the white matter loses, and diminish GFAP expression in blood. Although DCL-E4 and E4 have similar effect on neurogenesis in the hippocampus and the cortex, DCL-E4 treatment significantly up-regulates angiogenesis in the hippocampus compared to a single use of E4. Present work reveals for the first time that liposome-encapsulated E4 might be a better alternative to a single use of E4.
Collapse
Affiliation(s)
- Ekaterine Tskitishvili
- Laboratory of Developmental Biology and Tumors, GIGA-Cancer, Department of Clinical Sciences, Faculty of Medicine, University of Liège, Belgium.
| | - Claudio Palazzo
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Belgium
| | - Jean-Michel Foidart
- Laboratory of Developmental Biology and Tumors, GIGA-Cancer, Department of Clinical Sciences, Faculty of Medicine, University of Liège, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Belgium
| | - Christel Pequeux
- Laboratory of Developmental Biology and Tumors, GIGA-Cancer, Department of Clinical Sciences, Faculty of Medicine, University of Liège, Belgium
| |
Collapse
|
21
|
Min W, Zhou JH. Endothelial Cell-Pericyte Interactions in the Pathogenesis of Cerebral Cavernous Malformations (CCMs). Cold Spring Harb Perspect Med 2023; 13:a041188. [PMID: 35667709 PMCID: PMC9760308 DOI: 10.1101/cshperspect.a041188] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cerebral cavernous malformations (CCMs), consisting of multiple, dilated capillary channels formed by a single layer of endothelium and lacking parenchymal cells, are exclusively to the brain. Patients with inherited autosomal-dominant CCMs carry loss-of-function mutations in one of three genes: CCM1, CCM2, and CCM3. It is not known why CCM lesions are confined to brain vasculature despite the ubiquitous expression of CCM proteins in all tissues, and whether cell types other than endothelial cells (ECs) contribute to CCM lesion formation. The prevailing view is that the primary defects in CCMs in humans are EC-intrinsic, such that EC-specific deletion of any one of the three genes in mice results in similar CCM lesions. An unexpected finding is that Ccm3 deletion in pericytes (PCs) also induces CCM lesions. CCM3 deletion in ECs or PCs destabilizes PC-EC associations, highlighting the importance of these interactions in CCM formation.
Collapse
Affiliation(s)
- Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Jenny Huanjiao Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
22
|
Hattori Y. The microglia-blood vessel interactions in the developing brain. Neurosci Res 2023; 187:58-66. [PMID: 36167249 DOI: 10.1016/j.neures.2022.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022]
Abstract
Microglia are the immune cells in the central nervous system (CNS). Once microglial progenitors are generated in the yolk sac, these cells enter the CNS and colonize its structures by migrating and proliferating during development. Although the microglial population in the CNS is still low in this stage compared to adults, these cells can associate with many surrounding cells, such as neural lineage cells and vascular-structure-composing cells, by extending their filopodia and with their broad migration capacity. Previous studies revealed multifaceted microglial actions on neural lineage cells, such as regulating the differentiation of neural progenitors and modulating neuronal positioning. Notably, microglia not only act on neural lineage cells but also interact with blood vessels, for example, by supporting vascular formation and integrity. On the other hand, blood vessels contribute to microglial colonization into the CNS and their migration at local tissues. Importantly, pericytes, the cells that encompass vascular endothelial cells, have been suggested to play a profound role in microglial function. This review summarizes recent advances in the understanding of the interaction of microglia and blood vessels, especially focusing on the significance of this interaction in CNS development, and discusses how microglial and blood vessel dysfunction leads to developmental disorders.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
23
|
Shaji M, Kitada A, Fujimoto K, Karsten SL, Yokokawa R. Long-term effect of sodium selenite on the integrity and permeability of on-chip microvasculature. APL Bioeng 2022; 6:046105. [PMID: 36397962 PMCID: PMC9665962 DOI: 10.1063/5.0122804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Development of the robust and functionally stable three-dimensional (3D) microvasculature remains challenging. One often-overlooked factor is the presence of potential anti-angiogenic agents in culture media. Sodium selenite, an antioxidant commonly used in serum-free media, demonstrates strong anti-angiogenic properties and has been proposed as an anticancer drug. However, its long-term effects on in vitro microvascular systems at the concentrations used in culture media have not been studied. In this study, we used a five-channel microfluidic device to investigate the concentration and temporal effects of sodium selenite on the morphology and functionality of on-chip preformed microvasculature. We found that high concentrations (∼3.0 μM) had adverse effects on microvasculature perfusion, permeability, and overall integrity within the first few days. Moreover, even at low concentrations (∼3.0 nM), a long-term culture effect was observed, resulting in an increase in vascular permeability without any noticeable changes in morphology. A further analysis suggested that vessel leakage may be due to vascular endothelial growth factor dysregulation, disruption of intracellular junctions, or both. This study provides important insight into the adverse effects caused by the routinely present sodium selenite on 3D microvasculature in long-term studies for its application in disease modeling and drug screening.
Collapse
Affiliation(s)
- Maneesha Shaji
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan
| | - Atsuya Kitada
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan
| | - Kazuya Fujimoto
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan
| | - Stanislav L. Karsten
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan
| |
Collapse
|
24
|
Hattori Y. The Multiple Roles of Pericytes in Vascular Formation and Microglial Functions in the Brain. Life (Basel) 2022; 12:1835. [PMID: 36362989 PMCID: PMC9699346 DOI: 10.3390/life12111835] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 10/15/2023] Open
Abstract
In the capillary walls, vascular endothelial cells are covered with mural cells, such as smooth muscle cells and pericytes. Although pericytes had been thought to play simply a structural role, emerging evidence has highlighted their multiple functions in the embryonic, postnatal, and adult brain. As the central nervous system (CNS) develops, the brain's vascular structure gradually matures into a hierarchical network, which is crucial for the proper development of neural lineage cells by providing oxygen and nutrients. Pericytes play an essential role in vascular formation and regulate blood‒brain barrier (BBB) integrity as a component of the neurovascular unit (NVU), in collaboration with other cells, such as vascular endothelial cells, astrocytes, neurons, and microglia. Microglia, the resident immune cells of the CNS, colonize the brain at embryonic day (E) 9.5 in mice. These cells not only support the development and maturation of neural lineage cells but also help in vascular formation through their extensive migration. Recent studies have demonstrated that pericytes directly contact microglia in the CNS, and their interactions have a profound effect on physiological and pathological aspects. This review summarizes the function of pericytes, focusing on the interplay between pericytes and microglia.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
25
|
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
|
26
|
Uwhangchungsimwon Inhibits Oxygen Glucose Deprivation/Re-Oxygenation-Induced Cell Death through Neuronal VEGF and IGF-1 Receptor Signaling and Synaptic Remodeling in Cortical Neurons. Antioxidants (Basel) 2022; 11:antiox11071388. [PMID: 35883879 PMCID: PMC9311511 DOI: 10.3390/antiox11071388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/10/2022] [Accepted: 07/10/2022] [Indexed: 11/17/2022] Open
Abstract
Uwhangchungsimwon (UCW), a multi-component herbal product, has long been used to treat vascular diseases such as headache, dizziness, high blood pressure, and stroke. Though the prophylactic actions of UCW are well known, insufficient experimental evidence exists on its effectiveness against stroke. Here, we investigated the mechanism underlying the efficacy of UCW in oxygen glucose deprivation/re-oxygenation (OGD/R)-injury to the primary cortical neurons using an in vitro ischemia model. Neurons secrete vascular endothelial growth factor (VEGF), which acts as a neurotrophic factor in response to an ischemic injury. VEGF modulates neuroprotection and axonal outgrowth by activating the VEGF receptors and plays a critical role in vascular diseases. In this study, cortical neurons were pretreated with UCW (2, 10, and 50 µg/mL) for 48 h, incubated in oxygen-glucose-deprived conditions for 2 h, and further reoxygenated for 24 h. UCW effectively protected neurons from OGD/R-induced degeneration and cell death. Moreover, the role of UCW in sustaining protection against OGD/R injury is associated with activation of VEGF-VEGFR and insulin-like growth factor 1 receptor expression. Therefore, UCW is a potential herbal supplement for the prevention of hypoxic-ischemic neuronal injury as it may occur after stroke.
Collapse
|
27
|
Li H. Physiologic and pathophysiologic roles of AKAP12. Sci Prog 2022; 105:368504221109212. [PMID: 35775596 PMCID: PMC10450473 DOI: 10.1177/00368504221109212] [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] [Indexed: 11/16/2022]
Abstract
A kinase anchoring protein (AKAP) 12 is a scaffolding protein that improves the specificity and efficiency of spatiotemporal signal through assembling intracellular signal proteins into a specific complex. AKAP12 is a negative mitogenic regulator that plays an important role in controlling cytoskeletal architecture, maintaining endothelial integrity, regulating glial function and forming blood-brain barrier (BBB) and blood retinal barrier (BRB). Moreover, elevated or reduced AKAP12 contributes to a variety of diseases. Complex connections between AKAP12 and various diseases including chronic liver diseases (CLDs), inflammatory diseases and a series of cancers will be tried to delineate in this paper. We first describe the expression, distribution and physiological function of AKAP12. Then we summarize the current knowledge of different connections between AKAP12 expression and various diseases. Some research groups have found paradoxical roles of AKAP12 in different diseases and further confirmation is needed. This paper aims to assess the role of AKAP12 in physiology and diseases to help lay the foundation for the design of small molecules for specific AKAP12 to correct the pathological signal defects.
Collapse
Affiliation(s)
- Hui Li
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| |
Collapse
|
28
|
Xu Y, Luo L, Chen J. Sulfamethoxazole induces brain capillaries toxicity in zebrafish by up-regulation of VEGF and chemokine signalling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113620. [PMID: 35561544 DOI: 10.1016/j.ecoenv.2022.113620] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/27/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
Sulfamethoxazole (SMX) is a widespread broad-spectrum bacteriostatic antibiotic. Its residual is frequently detected in the water and may therefore bioaccumulate in the brain of aquatic organisms via blood circulation. Brain capillaries toxicity is very important for brain development. However, little information is available in the literature to show the toxicity of SMX to brain development. To study the SMX's brain toxic effects and the related mechanisms, we exposed zebrafish embryos to SMX at different concentrations (0 ppm, 1 ppm, 25 ppm, 100 ppm and 250 ppm) and found that high concentration (250 ppm) of SMX would not only caused an abnormal in malformation rate, hatching rate, body length and survival rate of zebrafish embryos, but also lead to brain oedema. In addition, SMX also induced cerebral ischaemia, aggravates oxidative stress, and changes genes related to oxidative stress (sod1, cat, gpx4, and nrf2). Furthermore, ischaemia caused by SMX could promote ectopic angiogenesis in brain via activating the angiogenesis-related genes (vegfab, cxcr4a, cxcl12b) from 24 h to 53 h. Inhibition of VEGF signalling by SU5416, or inhibition of chemokine downstream PI3K signalling by LY294002, could rescue the brain capillaries toxicity and brain oedema induced by SMX. Our results provide new evidence for the brain toxicity of SMX and its residual danger in the environment and aquatic organisms.
Collapse
Affiliation(s)
- Yuhang Xu
- University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingfei Luo
- University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China; Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China
| | - Jingying Chen
- University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China.
| |
Collapse
|
29
|
Richter RP, Payne GA, Ambalavanan N, Gaggar A, Richter JR. The endothelial glycocalyx in critical illness: A pediatric perspective. Matrix Biol Plus 2022; 14:100106. [PMID: 35392182 PMCID: PMC8981764 DOI: 10.1016/j.mbplus.2022.100106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/18/2022] Open
Abstract
The vascular endothelium is the interface between circulating blood and end organs and thus has a critical role in preserving organ function. The endothelium is lined by a glycan-rich glycocalyx that uniquely contributes to endothelial function through its regulation of leukocyte and platelet interactions with the vessel wall, vascular permeability, coagulation, and vasoreactivity. Degradation of the endothelial glycocalyx can thus promote vascular dysfunction, inflammation propagation, and organ injury. The endothelial glycocalyx and its role in vascular pathophysiology has gained increasing attention over the last decade. While studies characterizing vascular glycocalyx injury and its downstream consequences in a host of adult human diseases and in animal models has burgeoned, studies evaluating glycocalyx damage in pediatric diseases are relatively few. As children have unique physiology that differs from adults, significant knowledge gaps remain in our understanding of the causes and effects of endothelial glycocalyx disintegrity in pediatric critical illness. In this narrative literature overview, we offer a unique perspective on the role of the endothelial glycocalyx in pediatric critical illness, drawing from adult and preclinical data in addition to pediatric clinical experience to elucidate how marked derangement of the endothelial surface layer may contribute to aberrant vascular biology in children. By calling attention to this nascent field, we hope to increase research efforts to address important knowledge gaps in pediatric vascular biology that may inform the development of novel therapeutic strategies.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- CD, cell differentiation marker
- COVID-19, coronavirus disease 2019
- CPB, cardiopulmonary bypass
- CT, component therapy
- Children
- Critical illness
- DENV NS1, dengue virus nonstructural protein 1
- DM, diabetes mellitus
- ECLS, extracorporeal life support
- ECMO, extracorporeal membrane oxygenation
- EG, endothelial glycocalyx
- Endothelial glycocalyx
- FFP, fresh frozen plasma
- GAG, glycosaminoglycan
- GPC, glypican
- HPSE, heparanase
- HSV, herpes simplex virus
- IV, intravenous
- MIS-C, multisystem inflammatory syndrome in children
- MMP, matrix metalloproteinase
- Pragmatic, Randomized Optimal Platelet and Plasma Ratios
- RHAMM, receptor for hyaluronan-mediated motility
- S protein, spike protein
- SAFE, Saline versus Albumin Fluid Evaluation
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SDC, syndecan
- SDF, sidestream darkfield
- SIRT1, sirtuin 1
- TBI, traumatic brain injury
- TBSA, total body surface area
- TMPRSS2, transmembrane protease serine 2
- Th2, type 2 helper T cell
- VSMC, vascular smooth muscle cell
- Vascular biology
- WB+CT, whole blood and component therapy
- eNOS, endothelial nitric oxide synthase
Collapse
Affiliation(s)
- Robert P. Richter
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory A. Payne
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namasivayam Ambalavanan
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Translational Research in Normal and Disordered Development Program, University of Alabama, Birmingham, AL, USA
| | - Amit Gaggar
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jillian R. Richter
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
30
|
Sun XY, Ju XC, Li Y, Zeng PM, Wu J, Zhou YY, Shen LB, Dong J, Chen Y, Luo ZG. Generation of vascularized brain organoids to study neurovascular interactions. eLife 2022; 11:76707. [PMID: 35506651 PMCID: PMC9246368 DOI: 10.7554/elife.76707] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/01/2022] [Indexed: 12/05/2022] Open
Abstract
Brain organoids have been used to recapitulate the processes of brain development and related diseases. However, the lack of vasculatures, which regulate neurogenesis and brain disorders, limits the utility of brain organoids. In this study, we induced vessel and brain organoids, respectively, and then fused two types of organoids together to obtain vascularized brain organoids. The fused brain organoids were engrafted with robust vascular network-like structures and exhibited increased number of neural progenitors, in line with the possibility that vessels regulate neural development. Fusion organoids also contained functional blood–brain barrier-like structures, as well as microglial cells, a specific population of immune cells in the brain. The incorporated microglia responded actively to immune stimuli to the fused brain organoids and showed ability of engulfing synapses. Thus, the fusion organoids established in this study allow modeling interactions between the neuronal and non-neuronal components in vitro, particularly the vasculature and microglia niche. Understanding how the organs form and how their cells behave is essential to finding the causes and treatment for developmental disorders, as well as understanding certain diseases. However, studying most organs in live animals or humans is technically difficult, expensive and invasive. To address this issue, scientists have developed models called ‘organoids’ that recapitulate the development of organs using stem cells in the lab. These models are easier to study and manipulate than the live organs. Brain organoids have been used to recapitulate brain formation as well as developmental, degenerative and psychiatric brain conditions such as microcephaly, autism and Alzheimer’s disease. However, these brain organoids lack the vasculature (the network of blood vessels) that supplies a live brain with nutrients and regulates its development, and which has important roles in brain disorders. Partly due to this lack of blood vessels, brain organoids also do not develop a blood brain barrier, the structure that prevents certain contents of the blood, including pathogens, toxins and even certain drugs from entering the brain. These characteristics limit the utility of existing brain organoids. To overcome these limitations, Sun, Ju et al. developed brain organoids and blood vessel organoids independently, and then fused them together to obtain vascularized brain organoids. These fusion organoids developed a robust network of blood vessels that was well integrated with the brain cells, and produced more neural cell precursors than brain organoids that had not been fused. This result is consistent with the idea that blood vessels can regulate brain development. Analyzing the fusion organoids revealed that they contain structures similar to the blood-brain barrier, as well as microglial cells (immune cells specific to the brain). When exposed to lipopolysaccharide – a component of the cell wall of certain bacteria – these cells responded by initiating an immune response in the fusion organoids. Notably, the microglial cells were also able to engulf connections between brain cells, a process necessary for the brain to develop the correct structures and work normally. Sun, Ju et al. have developed a new organoid system that will be of broad interest to researchers studying interactions between the brain and the circulatory system. The development of brain-blood-barrier-like structures in the fusion organoids could also facilitate the development of drugs that can cross this barrier, making it easier to treat certain conditions that affect the brain. Refining this model to allow the fusion organoids to grow for longer times in the lab, and adding blood flow to the system will be the next steps to establish this system.
Collapse
Affiliation(s)
- Xin-Yao Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiang-Chun Ju
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yang Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Peng-Ming Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jian Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ying-Ying Zhou
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Li-Bing Shen
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Jian Dong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuejun Chen
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
31
|
Mostajeran M, Edvinsson L, Ahnstedt H, Arkelius K, Ansar S. Repair-related molecular changes during recovery phase of ischemic stroke in female rats. BMC Neurosci 2022; 23:23. [PMID: 35413803 PMCID: PMC9004052 DOI: 10.1186/s12868-022-00696-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Some degree of spontaneous recovery is usually observed after stroke. Experimental studies have provided information about molecular mechanisms underlying this recovery. However, the majority of pre-clinical stroke studies are performed in male rodents, and females are not well studied. This is a clear discrepancy when considering the clinical situation. Thus, it is important to include females in the evaluation of recovery mechanisms for future therapeutic strategies. This study aimed to evaluate spontaneous recovery and molecular mechanisms involved in the recovery phase two weeks after stroke in female rats. METHODS Transient middle cerebral artery occlusion was induced in female Wistar rats using a filament model. Neurological functions were assessed up to day 14 after stroke. Protein expression of interleukin 10 (IL-10), transforming growth factor (TGF)-β, neuronal specific nuclei protein (NeuN), nestin, tyrosine-protein kinase receptor Tie-2, extracellular signal-regulated kinase (ERK) 1/2, and Akt were evaluated in the peri-infarct and ischemic core compared to contralateral side of the brain at day 14 by western blot. Expression of TGF-β in middle cerebral arteries was evaluated by immunohistochemistry. RESULTS Spontaneous recovery after stroke was observed from day 2 to day 14 and was accompanied by a significantly higher expression of nestin, p-Akt, p-ERK1/2 and TGF-β in ischemic regions compared to contralateral side at day 14. In addition, a significantly higher expression of TGF-β was observed in occluded versus non-occluded middle cerebral arteries. The expression of Tie-2 and IL-10 did not differ between the ischemic and contralateral sides. CONCLUSION Spontaneous recovery after ischemic stroke in female rats was coincided by a difference observed in the expression of molecular markers. The alteration of these markers might be of importance to address future therapeutic strategies.
Collapse
Affiliation(s)
- Maryam Mostajeran
- Division of Experimental Vascular Research, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lars Edvinsson
- Division of Experimental Vascular Research, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hilda Ahnstedt
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kajsa Arkelius
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Saema Ansar
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
32
|
Puebla M, Tapia PJ, Espinoza H. Key Role of Astrocytes in Postnatal Brain and Retinal Angiogenesis. Int J Mol Sci 2022; 23:ijms23052646. [PMID: 35269788 PMCID: PMC8910249 DOI: 10.3390/ijms23052646] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Angiogenesis is a key process in various physiological and pathological conditions in the nervous system and in the retina during postnatal life. Although an increasing number of studies have addressed the role of endothelial cells in this event, the astrocytes contribution in angiogenesis has received less attention. This review is focused on the role of astrocytes as a scaffold and in the stabilization of the new blood vessels, through different molecules release, which can modulate the angiogenesis process in the brain and in the retina. Further, differences in the astrocytes phenotype are addressed in glioblastoma, one of the most devastating types of brain cancer, in order to provide potential targets involved in the cross signaling between endothelial cells, astrocytes and glioma cells, that mediate tumor progression and pathological angiogenesis. Given the relevance of astrocytes in angiogenesis in physiological and pathological conditions, future studies are required to better understand the interrelation between endothelial and astrocyte signaling pathways during this process.
Collapse
Affiliation(s)
- Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Av. Plaza 680, Las Condes, Santiago 7550000, Chile;
| | - Pablo J. Tapia
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Av. Lota 2465, Providencia, Santiago 7500000, Chile;
- Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Av. República 71, Santiago 8320000, Chile
| | - Hilda Espinoza
- Facultad de Ciencias de la Salud, Universidad del Alba, Av. Ejército Libertador 171, Santiago 8320000, Chile
- Correspondence:
| |
Collapse
|
33
|
Liu W, Zhou Y, Zhang YX, Yang KL, Liu YL, Wu FH, Gao YR. Connexin 43 mediated the angiogenesis of buyang huanwu decoction via vascular endothelial growth factor and angiopoietin-1 after ischemic stroke. CHINESE J PHYSIOL 2022; 65:72-79. [DOI: 10.4103/cjp.cjp_94_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
34
|
Ahn JC, Hwang SJ, Lee HJ, Kim KW. Claudin-5a knockdown attenuates blood-neural barrier in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2021; 250:109176. [PMID: 34500089 DOI: 10.1016/j.cbpc.2021.109176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/12/2021] [Accepted: 08/25/2021] [Indexed: 12/31/2022]
Abstract
Mammalian claudin-5 (cldn5), a zebrafish cldn5a homolog, is essential to blood-brain barrier (BBB) integrity. Previously, the existence of an endothelial tight junction-based BBB with cldn5a expression in the cerebral microvessels was reported in zebrafish. However, the role of cldn5a in the cerebral microvessels of developing zebrafish has not been elucidated. Here, we further investigated the functional integrity of cldn5a in developing zebrafish by injecting cldn5a morpholinos. At 7 days post-fertilization, cldn5a immunoreactivity was detected on the brain surface, ventricular ependyma, and cerebral mircovessels but disappeared following cldna5a knockdown. Cldn5a morphants showed size-selective leakage of tracers through the BBB and downregulated expression of glucose transporter 1 (glut1) in the cerebral microvessels. In addition, leakiness in the blood-cerebrospinal fluid barrier was observed, implying the overall abnormal development of blood-neural barriers. The results of our study suggest that cldn5a is required for building and maintaining the blood-neural barrier during zebrafish development.
Collapse
Affiliation(s)
- Jong-Chan Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea
| | - Su Jung Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea.
| | - Kyu-Won Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
35
|
Sadanandan N, Shear A, Brooks B, Saft M, Cabantan DAG, Kingsbury C, Zhang H, Anthony S, Wang ZJ, Salazar FE, Lezama Toledo AR, Rivera Monroy G, Vega Gonzales-Portillo J, Moscatello A, Lee JY, Borlongan CV. Treating Metastatic Brain Cancers With Stem Cells. Front Mol Neurosci 2021; 14:749716. [PMID: 34899179 PMCID: PMC8651876 DOI: 10.3389/fnmol.2021.749716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cell therapy may present an effective treatment for metastatic brain cancer and glioblastoma. Here we posit the critical role of a leaky blood-brain barrier (BBB) as a key element for the development of brain metastases, specifically melanoma. By reviewing the immunological and inflammatory responses associated with BBB damage secondary to tumoral activity, we identify the involvement of this pathological process in the growth and formation of metastatic brain cancers. Likewise, we evaluate the hypothesis of regenerating impaired endothelial cells of the BBB and alleviating the damaged neurovascular unit to attenuate brain metastasis, using the endothelial progenitor cell (EPC) phenotype of bone marrow-derived mesenchymal stem cells. Specifically, there is a need to evaluate the efficacy for stem cell therapy to repair disruptions in the BBB and reduce inflammation in the brain, thereby causing attenuation of metastatic brain cancers. To establish the viability of stem cell therapy for the prevention and treatment of metastatic brain tumors, it is crucial to demonstrate BBB repair through augmentation of vasculogenesis and angiogenesis. BBB disruption is strongly linked to metastatic melanoma, worsens neuroinflammation during metastasis, and negatively influences the prognosis of metastatic brain cancer. Using stem cell therapy to interrupt inflammation secondary to this leaky BBB represents a paradigm-shifting approach for brain cancer treatment. In this review article, we critically assess the advantages and disadvantages of using stem cell therapy for brain metastases and glioblastoma.
Collapse
Affiliation(s)
| | - Alex Shear
- University of Florida, Gainesville, FL, United States
| | - Beverly Brooks
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Madeline Saft
- University of Michigan, Ann Arbor, MI, United States
| | | | - Chase Kingsbury
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Henry Zhang
- University of Florida, Gainesville, FL, United States
| | - Stefan Anthony
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Zhen-Jie Wang
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Felipe Esparza Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud (FCS), Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Alma R. Lezama Toledo
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud (FCS), Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud (FCS), Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | | | - Alexa Moscatello
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Cesario V. Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
36
|
Beura SK, Panigrahi AR, Yadav P, Agrawal S, Singh SK. Role of Neurons and Glia Cells in Wound Healing as a Novel Perspective Considering Platelet as a Conventional Player. Mol Neurobiol 2021; 59:137-160. [PMID: 34633653 DOI: 10.1007/s12035-021-02587-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Wound healing is a complex physiological process in which the damaged or injured tissue is replaced or regenerated by new cells or existing cells respectively in their synthesized and secreted matrices. Several cells modulate the process of wound healing including macrophages, fibroblasts, and keratinocytes. Apart from these cells, platelet has been considered as a major cellular fragment to be involved in wound healing at several stages by secreting its granular contents including growth factors, thus resulting in coagulation, inflammation, and angiogenesis. A distant cell, which is gaining significant attention nowadays due to its resemblance with platelet in several aspects, is the neuron. Not only neurons but also glia cells are also confirmed to regulate wound healing at different stages in an orchestrated manner. Furthermore, these neurons and glia cells mediate wound healing inducing tissue repair and regeneration apart from hemostasis, angiogenesis, and inflammation by secreting various growth factors, coagulation molecules, immunomodulatory molecules as well as neurohormones, neuropeptides, and neurotrophins. Therefore, in wound healing platelets, neurons and glia cells not only contribute to tissue repair but are also responsible for establishing the wound microenvironment, thus affecting the proliferation of immune cells, fibroblast, and keratinocytes. Here in this review, we will enlighten the physiological roles of neurons and glia cells in coordination with platelets to understand various cellular and molecular mechanism in brain injury and associated neurocognitive impairments.
Collapse
Affiliation(s)
- Samir K Beura
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Pooja Yadav
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Siwani Agrawal
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sunil K Singh
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India.
| |
Collapse
|
37
|
van Lanen RH, Melchers S, Hoogland G, Schijns OE, Zandvoort MAV, Haeren RH, Rijkers K. Microvascular changes associated with epilepsy: A narrative review. J Cereb Blood Flow Metab 2021; 41:2492-2509. [PMID: 33866850 PMCID: PMC8504411 DOI: 10.1177/0271678x211010388] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) is dysfunctional in temporal lobe epilepsy (TLE). In this regard, microvascular changes are likely present. The aim of this review is to provide an overview of the current knowledge on microvascular changes in epilepsy, and includes clinical and preclinical evidence of seizure induced angiogenesis, barriergenesis and microcirculatory alterations. Anatomical studies show increased microvascular density in the hippocampus, amygdala, and neocortex accompanied by BBB leakage in various rodent epilepsy models. In human TLE, a decrease in afferent vessels, morphologically abnormal vessels, and an increase in endothelial basement membranes have been observed. Both clinical and experimental evidence suggests that basement membrane changes, such as string vessels and protrusions, indicate and visualize a misbalance between endothelial cell proliferation and barriergenesis. Vascular endothelial growth factor (VEGF) appears to play a crucial role. Following an altered vascular anatomy, its physiological functioning is affected as expressed by neurovascular decoupling that subsequently leads to hypoperfusion, disrupted parenchymal homeostasis and potentially to seizures". Thus, epilepsy might be a condition characterized by disturbed cerebral microvasculature in which VEGF plays a pivotal role. Additional physiological data from patients is however required to validate findings from models and histological studies on patient biopsies.
Collapse
Affiliation(s)
- Rick Hgj van Lanen
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stan Melchers
- Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Olaf Emg Schijns
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marc Amj van Zandvoort
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Molecular Cell Biology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Roel Hl Haeren
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Kim Rijkers
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
38
|
Do PT, Wu CC, Chiang YH, Hu CJ, Chen KY. Mesenchymal Stem/Stromal Cell Therapy in Blood-Brain Barrier Preservation Following Ischemia: Molecular Mechanisms and Prospects. Int J Mol Sci 2021; 22:ijms221810045. [PMID: 34576209 PMCID: PMC8468469 DOI: 10.3390/ijms221810045] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is the leading cause of mortality and long-term disability worldwide. Disruption of the blood-brain barrier (BBB) is a prominent pathophysiological mechanism, responsible for a series of subsequent inflammatory cascades that exacerbate the damage to brain tissue. However, the benefit of recanalization is limited in most patients because of the narrow therapeutic time window. Recently, mesenchymal stem cells (MSCs) have been assessed as excellent candidates for cell-based therapy in cerebral ischemia, including neuroinflammatory alleviation, angiogenesis and neurogenesis promotion through their paracrine actions. In addition, accumulating evidence on how MSC therapy preserves BBB integrity after stroke may open up novel therapeutic targets for treating cerebrovascular diseases. In this review, we focus on the molecular mechanisms of MSC-based therapy in the ischemia-induced prevention of BBB compromise. Currently, therapeutic effects of MSCs for stroke are primarily based on the fundamental pathogenesis of BBB breakdown, such as attenuating leukocyte infiltration, matrix metalloproteinase (MMP) regulation, antioxidant, anti-inflammation, stabilizing morphology and crosstalk between cellular components of the BBB. We also discuss prospective studies to improve the effectiveness of MSC therapy through enhanced migration into defined brain regions of stem cells. Targeted therapy is a promising new direction and is being prioritized for extensive research.
Collapse
Affiliation(s)
- Phuong Thao Do
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Pediatrics, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Chung-Che Wu
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan; (C.-C.W.); (Y.-H.C.)
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan; (C.-C.W.); (Y.-H.C.)
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- TMU Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan
| | - Chaur-Jong Hu
- TMU Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Neurology and Stroke Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Correspondence: (C.-J.H.); (K.-Y.C.); Tel.: +886-227361661 (ext. 3032) (C.-J.H.); +886-227361661 (ext. 7602) (K.-Y.C.)
| | - Kai-Yun Chen
- TMU Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
- The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (C.-J.H.); (K.-Y.C.); Tel.: +886-227361661 (ext. 3032) (C.-J.H.); +886-227361661 (ext. 7602) (K.-Y.C.)
| |
Collapse
|
39
|
Valentino M, Dejana E, Malinverno M. The multifaceted PDCD10/CCM3 gene. Genes Dis 2021; 8:798-813. [PMID: 34522709 PMCID: PMC8427250 DOI: 10.1016/j.gendis.2020.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
The programmed cell death 10 (PDCD10) gene was originally identified as an apoptosis-related gene, although it is now usually known as CCM3, as the third causative gene of cerebral cavernous malformation (CCM). CCM is a neurovascular disease that is characterized by vascular malformations and is associated with headaches, seizures, focal neurological deficits, and cerebral hemorrhage. The PDCD10/CCM3 protein has multiple subcellular localizations and interacts with several multi-protein complexes and signaling pathways. Thus PDCD10/CCM3 governs many cellular functions, which include cell-to-cell junctions and cytoskeleton organization, cell proliferation and apoptosis, and exocytosis and angiogenesis. Given its central role in the maintenance of homeostasis of the cell, dysregulation of PDCD10/CCM3 can result in a wide range of altered cell functions. This can lead to severe diseases, including CCM, cognitive disability, and several types of cancers. Here, we review the multifaceted roles of PDCD10/CCM3 in physiology and pathology, with a focus on its functions beyond CCM.
Collapse
Affiliation(s)
| | - Elisabetta Dejana
- The FIRC Institute of Molecular Oncology (IFOM), Milan, 16 20139, Italy.,Department of Oncology and Haemato-Oncology, University of Milan, Milan, 7 20122, Italy.,Vascular Biology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, SE-751 05, Sweden
| | - Matteo Malinverno
- The FIRC Institute of Molecular Oncology (IFOM), Milan, 16 20139, Italy
| |
Collapse
|
40
|
Milich LM, Choi JS, Ryan C, Cerqueira SR, Benavides S, Yahn SL, Tsoulfas P, Lee JK. Single-cell analysis of the cellular heterogeneity and interactions in the injured mouse spinal cord. J Exp Med 2021; 218:e20210040. [PMID: 34132743 PMCID: PMC8212781 DOI: 10.1084/jem.20210040] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/09/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
The wound healing process that occurs after spinal cord injury is critical for maintaining tissue homeostasis and limiting tissue damage, but eventually results in a scar-like environment that is not conducive to regeneration and repair. A better understanding of this dichotomy is critical to developing effective therapeutics that target the appropriate pathobiology, but a major challenge has been the large cellular heterogeneity that results in immensely complex cellular interactions. In this study, we used single-cell RNA sequencing to assess virtually all cell types that comprise the mouse spinal cord injury site. In addition to discovering novel subpopulations, we used expression values of receptor-ligand pairs to identify signaling pathways that are predicted to regulate specific cellular interactions during angiogenesis, gliosis, and fibrosis. Our dataset is a valuable resource that provides novel mechanistic insight into the pathobiology of not only spinal cord injury but also other traumatic disorders of the CNS.
Collapse
Affiliation(s)
- Lindsay M. Milich
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
- University of Miami Neuroscience Graduate Program, Miami, FL
| | - James S. Choi
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
| | - Christine Ryan
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
- University of Miami Neuroscience Graduate Program, Miami, FL
| | - Susana R. Cerqueira
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
| | - Sofia Benavides
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
| | - Stephanie L. Yahn
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
- University of Miami Neuroscience Graduate Program, Miami, FL
| | - Pantelis Tsoulfas
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
| | - Jae K. Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL
| |
Collapse
|
41
|
Cheng C, Wang X, Jiang Y, Li Y, Liao Z, Li W, Yu Z, Whalen MJ, Lok J, Dumont AS, Liu N, Wang X. Recombinant Annexin A2 Administration Improves Neurological Outcomes After Traumatic Brain Injury in Mice. Front Pharmacol 2021; 12:708469. [PMID: 34400908 PMCID: PMC8363504 DOI: 10.3389/fphar.2021.708469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Microvascular failure is one of the key pathogenic factors in the dynamic pathological evolution after traumatic brain injury (TBI). Our laboratory and others previously reported that Annexin A2 functions in blood-brain barrier (BBB) development and cerebral angiogenesis, and recombinant human Annexin A2 (rA2) protected against hypoxia plus IL-1β-induced cerebral trans-endothelial permeability in vitro, and cerebral angiogenesis impairment of AXNA2 knock-out mice in vivo. We thereby hypothesized that ANXA2 might be a cerebrovascular therapy candidate that targets early BBB integrity disruption, and subacute/delayed cerebrovascular remodeling after TBI, ultimately improve neurological outcomes. In a controlled cortex impact (CCI) mice model, we found rA2 treatment (1 mg/kg) significantly reduced early BBB disruption at 24 h after TBI; and rA2 daily treatment for 7 days augmented TBI-induced mRNA levels of pro-angiogenic and endothelial-derived trophic factors in cerebral microvessels. In cultured human brain microvascular endothelial cells (HBMEC), through MAPKs array, we identified that rA2 significantly activated Akt, ERK, and CREB, and the activated CREB might be responsible for the rA2-induced VEGF and BDNF expression. Moreover, rA2 administration significantly increased cerebral angiogenesis examined at 14 days and vessel density at 28 days after TBI in mice. Consistently, our results validated that rA2 significantly induced angiogenesis in vitro, evidenced by tube formation and scratched migration assays in HBMEC. Lastly, we demonstrated that rA2 improved long-term sensorimotor and cognitive function, and reduced brain tissue loss at 28 days after TBI. Our findings suggest that rA2 might be a novel vascular targeting approach for treating TBI.
Collapse
Affiliation(s)
- Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Xiaoshu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Yinghua Jiang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Yadan Li
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Zhengbu Liao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Wenlu Li
- Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Zhanyang Yu
- Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Michael J Whalen
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Josephine Lok
- Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ning Liu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
42
|
Galectin-1 Contributes to Vascular Remodeling and Blood Flow Recovery After Cerebral Ischemia in Mice. Transl Stroke Res 2021; 13:160-170. [PMID: 33973144 DOI: 10.1007/s12975-021-00913-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/28/2020] [Accepted: 04/26/2021] [Indexed: 12/19/2022]
Abstract
Galectin-1 is found in the vasculature and has been confirmed to promote angiogenesis in several cancer models. Furthermore, galectin-1 has been demonstrated to improve the recovery of cerebral ischemia. However, whether vascular remodeling contributes to this improvement is still unknown. In the present study, photochemical cerebral ischemia was induced in both galectin-1-treated (2 μg/day, i.c.v, 3 days) and galectin-1 knockout mice. Laser speckle imaging and immunofluorescent staining demonstrated that circulation and vascular remodeling in the ischemic cortex were improved by galectin-1 treatment but disrupted in galectin-1 knockout mice. Western blot analysis showed that the expression of matrix metallopeptidase-9 and vascular endothelial growth factor (VEGF) was regulated by galectin-1 in vivo. To determine how galectin-1 influences endothelial cells, the expression of galectin-1 in bEnd.3 cells was increased by transfection with an expression plasmid and knocked down by siRNA. As demonstrated by quantitative RT-PCR and western blot analysis, the expression of metallopeptidase-9, VEGF, and VEGF receptors was upregulated by galectin-1 overexpression but downregulated after galectin-1 knockdown. Flow cytometry, Transwell assay, and capillary-like tube formation assay were performed on cells after gene manipulation as well as cells treated by exogenous galectin-1 after anoxia. It demonstrated that galectin-1 potentiated the cell proliferation, migration capacity, and tube formation ability. Taken together, these data suggest that by targeting vascular remodeling, galectin-1 contributes to the restoration of blood flow, which promotes the recovery of mice after cerebral ischemic insults.
Collapse
|
43
|
Gama Sosa MA, De Gasperi R, Perez GM, Hof PR, Elder GA. Hemovasculogenic origin of blood vessels in the developing mouse brain. J Comp Neurol 2021; 529:340-366. [PMID: 32415669 DOI: 10.1002/cne.24951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 01/20/2023]
Abstract
Vascular structures in the developing brain are thought to form via angiogenesis from preformed blood vessels in the cephalic mesenchyme. Immunohistochemical studies of developing mouse brain from E10.5 to E13.5 revealed the presence of avascular blood islands of primitive erythroid cells expressing hemangioblast markers (Flk1, Tal1/Scl1, platelet endothelial cell adhesion molecule 1, vascular endothelial-cadherin, and CD34) and an endothelial marker recognized by Griffonia simplicifolia isolectin B4 (IB4) in the cephalic mesenchyme. These cells formed a perineural vascular plexus from which angiogenic sprouts originated and penetrated the neuroepithelium. In addition, avascular isolated cells expressing primitive erythroid, hemangioblast and endothelial makers were visible in the neuroepithelium where they generated vasculogenic and hemogenic foci. From E10.5 to E13.5, these vasculogenic foci were a source of new blood vessel formation in the developing brain. In vitro, cultured E13.5 brain endothelial cells contained hemogenic endothelial cells capable of generating erythroid cells. Similar cells were present in primary cultures of dissociated cells from E10.5 embryonic head. Our results provide new evidence that the brain vasculature, like that of the yolk sac and the eye choriocapillaris and hyaloid vascular systems, develops at least in part via hemovasculogenesis, a process in which vasculogenesis and hematopoiesis occur simultaneously.
Collapse
Affiliation(s)
- Miguel A Gama Sosa
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Gissel M Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Patrick R Hof
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gregory A Elder
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
44
|
Treatment with Atorvastatin During Vascular Remodeling Promotes Pericyte-Mediated Blood-Brain Barrier Maturation Following Ischemic Stroke. Transl Stroke Res 2021; 12:905-922. [PMID: 33423214 DOI: 10.1007/s12975-020-00883-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
We previously showed that newly formed vessels in ischemic rat brain have high blood-brain barrier (BBB) permeability at 3 weeks after stroke due to a lack of major endothelial tight junction proteins (TJPs), which may exacerbate edema in stroke patients. Atorvastatin was suggested a dose-dependent pro-angiogenic effect and ameliorating BBB permeability beyond its cholesterol-lowering effects. This study examined our hypothesis that, during vascular remodeling after stroke, treatment with atorvastatin could facilitate BBB maturation in remodeling vasculature in ischemic brain. Adult spontaneously hypertensive rats underwent middle cerebral artery occlusion with reperfusion (MCAO/RP). Atorvastatin, at dose of 3 mg/kg, was delivered daily starting at 14 days after MCAO/RP onset for 7 days. The rats were studied at multiple time points up to 8 weeks with multimodal-MRI, behavior tests, immunohistochemistry, and biochemistry. The delayed treatment of atorvastatin significantly reduced infarct size and BBB permeability, restored cerebral blood flow, and improved the neurological outcome at 8 weeks after MCAO/RP. Postmortem studies showed that atorvastatin promoted angiogenesis and stabilized the newly formed vessels in peri-infarct areas. Importantly, atorvastatin facilitated maturation of BBB properties in the new vessels by promoting endothelial tight junction (TJ) formation. Further in vivo and in vitro studies demonstrated that proliferating peri-vascular pericytes expressing neural-glial antigen 2 (NG2) mediated the role of atorvastatin on BBB maturation through regulating endothelial TJ strand formations. Our results suggested a therapeutic potential of atorvastatin in facilitating a full BBB integrity and functional stroke recovery, and an essential role for pericyte-mediated endothelial TJ formation in remodeling vasculature.
Collapse
|
45
|
Zhan Y, Li MZ, Yang L, Feng XF, Lei JF, Zhang N, Zhao YY, Zhao H. The three-phase enriched environment paradigm promotes neurovascular restorative and prevents learning impairment after ischemic stroke in rats. Neurobiol Dis 2020; 146:105091. [DOI: 10.1016/j.nbd.2020.105091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/30/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
|
46
|
Yang Y, Torbey MT. Angiogenesis and Blood-Brain Barrier Permeability in Vascular Remodeling after Stroke. Curr Neuropharmacol 2020; 18:1250-1265. [PMID: 32691713 PMCID: PMC7770645 DOI: 10.2174/1570159x18666200720173316] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/27/2020] [Accepted: 07/11/2020] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis, the growth of new blood vessels, is a natural defense mechanism helping to restore oxygen and nutrient supply to the affected brain tissue following an ischemic stroke. By stimulating vessel growth, angiogenesis may stabilize brain perfusion, thereby promoting neuronal survival, brain plasticity, and neurologic recovery. However, therapeutic angiogenesis after stroke faces challenges: new angiogenesis-induced vessels have a higher than normal permeability, and treatment to promote angiogenesis may exacerbate outcomes in stroke patients. The development of therapies requires elucidation of the precise cellular and molecular basis of the disease. Microenvironment homeostasis of the central nervous system is essential for its normal function and is maintained by the blood-brain barrier (BBB). Tight junction proteins (TJP) form the tight junction (TJ) between vascular endothelial cells (ECs) and play a key role in regulating the BBB permeability. We demonstrated that after stroke, new angiogenesis-induced vessels in peri-infarct areas have abnormally high BBB permeability due to a lack of major TJPs in ECs. Therefore, promoting TJ formation and BBB integrity in the new vessels coupled with speedy angiogenesis will provide a promising and safer treatment strategy for improving recovery from stroke. Pericyte is a central neurovascular unite component in vascular barriergenesis and are vital to BBB integrity. We found that pericytes also play a key role in stroke-induced angiogenesis and TJ formation in the newly formed vessels. Based on these findings, in this article, we focus on regulation aspects of the BBB functions and describe cellular and molecular special features of TJ formation with an emphasis on role of pericytes in BBB integrity during angiogenesis after stroke.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| | - Michel T Torbey
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| |
Collapse
|
47
|
Jiang W, Liu C, Deng M, Wang F, Ren X, Fan Y, Du J, Wang Y. H 2S promotes developmental brain angiogenesis via the NOS/NO pathway in zebrafish. Stroke Vasc Neurol 2020; 6:244-251. [PMID: 33246971 PMCID: PMC8258041 DOI: 10.1136/svn-2020-000584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/19/2020] [Accepted: 10/28/2020] [Indexed: 01/04/2023] Open
Abstract
Background Hydrogen sulphide (H2S) is considered as the third member of the gasotransmitter family, along with nitric oxide (NO) and carbon monoxide. H2S has been reported to induce angiogenesis by promoting the growth, migration and tube-like structure formation of endothelial cells. Those studies were conducted in conditions of cell culture, mouse Matrigel plug assay model, rat wound healing model or rat hindlimb ischaemia model. Recent in vivo studies showed the physiological importance of H2S in muscle angiogenesis. However, the importance of endogenous H2S for brain angiogenesis during development remains unknown. We therefore aimed at determining the role of H2S in brain vascular development. Methods and results Both knockdown and knockout of H2S-producing enzymes, cystathionine β-synthase (cbs) and cystathionine γ-lyase (cth), using morpholino oligonucleotides and clustered regularly interspaced short palindromic repeats/Cas9-mediated mutation, impaired brain vascular development of larval zebrafish. Incubation with the slow-releasing H2S donor GYY4137 alleviated the defects of brain vascular development in cbs and cth morphants. Quantitative analysis of the midbrain vascular network showed that H2S enhances angiogenesis without affecting the topological structure of the brain vasculature. Mechanically, nitric oxide synthase 2a (nos2a) expression and NO production were decreased in both cbs and cth morphants. Overexpression of nos2a by coinjection of cbs or cth MO with full-length zebrafish nos2a mRNA alleviated the brain vascular developmental defects in cbs and cth morphants. Conclusion We conclude that H2S promotes brain developmental angiogenesis via the NOS/NO pathway in zebrafish.
Collapse
Affiliation(s)
- Weiqing Jiang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Chen Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingzhu Deng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fei Wang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Xiao Ren
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Yilin Fan
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yonggang Wang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
48
|
Elibol B, Beker M, Jakubowska-Dogru E, Kilic U. Fetal alcohol and maternal stress modify the expression of proteins controlling postnatal development of the male rat hippocampus. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2020; 46:718-730. [PMID: 32915069 DOI: 10.1080/00952990.2020.1780601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Developing brains can partially get over prenatal alcohol exposure-related detrimental conditions by activating some mechanisms involved in survival. Objectives: This study aimed to shed light on the molecular correlates of compensatory mechanisms by examining temporal profiles in the expression of proteins controlling postnatal development in the rat hippocampus prenatally exposed to intubation stress/ethanol. Methods: Male pups were randomly assigned to age subgroups (n = 21/age) which were sacrificed on postnatal day (PD)1, PD10, PD30, and PD60. Ethanol (6 g/kg/day) were intragastrically intubated to the dams throughout 7-21 gestation days. The expression of neurogenesis and angiogenesis markers, extracellular matrix proteins, and growth-promoting ligands were examined by western blot. Results: The most rapid increase in the index of neuronal maturation was noted between PD10-PD30 (p < .05). Prenatal stress caused a decrease of neurogenesis markers at birth and an increase of their expressions at PD10 and PD30 to reach control levels (p < .001). The impact of fetal-alcohol was observed as a decrease in the expression of synaptic plasticity protein versican at birth (p < .001), an increase in the synaptic repulsion protein ephrin-B2 at PD10 (p < .001), and a decrease in the maturation of BDNF at PD30 (p < .001) with a decrease in the mature neuron markers at PD30 (p < .001) and PD60 (p = .005) which were compensated with upregulation of angiogenesis and increasing brevican expression, a neuronal maturation protein (p < .001). Conclusion: These data provide in vivo evidence for the potential therapeutic factors related to neurogenesis, angiogenesis, and neurite remodeling which may tolerate the alcohol/stress dependent teratogenicity in the developing hippocampus.
Collapse
Affiliation(s)
- Birsen Elibol
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University , Istanbul, Turkey
| | - Merve Beker
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University , Istanbul, Turkey.,Department of Medical Biology, School of Medicine, University of Health Sciences , Istanbul, Turkey
| | - Ewa Jakubowska-Dogru
- Department of Biological Sciences, Faculty of Science and Arts, Middle East Technical University , Ankara, Turkey
| | - Ulkan Kilic
- Department of Medical Biology, School of Medicine, University of Health Sciences , Istanbul, Turkey
| |
Collapse
|
49
|
GKT136901 protects primary human brain microvascular endothelial cells against methamphetamine-induced blood-brain barrier dysfunction. Life Sci 2020; 256:117917. [DOI: 10.1016/j.lfs.2020.117917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 01/01/2023]
|
50
|
Santander N, Lizama CO, Meky E, McKinsey GL, Jung B, Sheppard D, Betsholtz C, Arnold TD. Lack of Flvcr2 impairs brain angiogenesis without affecting the blood-brain barrier. J Clin Invest 2020; 130:4055-4068. [PMID: 32369453 PMCID: PMC7410045 DOI: 10.1172/jci136578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022] Open
Abstract
Fowler syndrome is a rare autosomal recessive brain vascular disorder caused by mutation in FLVCR2 in humans. The disease occurs during a critical period of brain vascular development, is characterized by glomeruloid vasculopathy and hydrocephalus, and is almost invariably prenatally fatal. Here, we sought to gain insights into the process of brain vascularization and the pathogenesis of Fowler syndrome by inactivating Flvcr2 in mice. We showed that Flvcr2 was necessary for angiogenic sprouting in the brain, but surprisingly dispensable for maintaining the blood-brain barrier. Endothelial cells lacking Flvcr2 had altered expression of angiogenic factors, failed to adopt tip cell properties, and displayed reduced sprouting, leading to vascular malformations similar to those seen in humans with Fowler syndrome. Brain hypovascularization was associated with hypoxia and tissue infarction, ultimately causing hydrocephalus and death of mutant animals. Strikingly, despite severe vascular anomalies and brain tissue infarction, the blood-brain barrier was maintained in Flvcr2 mutant mice. Our Fowler syndrome model therefore defined the pathobiology of this disease and provided new insights into brain angiogenesis by showing uncoupling of vessel morphogenesis and blood-brain barrier formation.
Collapse
Affiliation(s)
| | - Carlos O. Lizama
- Cardiovascular Research Institute, UCSF, San Francisco, California, USA
| | | | | | - Bongnam Jung
- Integrated Cardiometabolic Center, Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
| | - Dean Sheppard
- Department of Cell Biology, UCSF, San Francisco, California, USA
| | - Christer Betsholtz
- Integrated Cardiometabolic Center, Department of Medicine, Huddinge, Karolinska Institutet, Solna, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|