1
|
Fernezelian D, Rondeau P, Gence L, Diotel N. Telencephalic stab wound injury induces regenerative angiogenesis and neurogenesis in zebrafish: unveiling the role of vascular endothelial growth factor signaling and microglia. Neural Regen Res 2025; 20:2938-2954. [PMID: 39248179 PMCID: PMC11826465 DOI: 10.4103/nrr.nrr-d-23-01881] [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: 11/15/2023] [Revised: 07/13/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00025/figure1/v/2024-11-26T163120Z/r/image-tiff After brain damage, regenerative angiogenesis and neurogenesis have been shown to occur simultaneously in mammals, suggesting a close link between these processes. However, the mechanisms by which these processes interact are not well understood. In this work, we aimed to study the correlation between angiogenesis and neurogenesis after a telencephalic stab wound injury. To this end, we used zebrafish as a relevant model of neuroplasticity and brain repair mechanisms. First, using the Tg( fli1:EGFP × mpeg1.1:mCherry ) zebrafish line, which enables visualization of blood vessels and microglia respectively, we analyzed regenerative angiogenesis from 1 to 21 days post-lesion. In parallel, we monitored brain cell proliferation in neurogenic niches localized in the ventricular zone by using immunohistochemistry. We found that after brain damage, the blood vessel area and width as well as expression of the fli1 transgene and vascular endothelial growth factor ( vegfaa and vegfbb ) were increased. At the same time, neural stem cell proliferation was also increased, peaking between 3 and 5 days post-lesion in a manner similar to angiogenesis, along with the recruitment of microglia. Then, through pharmacological manipulation by injecting an anti-angiogenic drug (Tivozanib) or Vegf at the lesion site, we demonstrated that blocking or activating Vegf signaling modulated both angiogenic and neurogenic processes, as well as microglial recruitment. Finally, we showed that inhibition of microglia by clodronate-containing liposome injection or dexamethasone treatment impairs regenerative neurogenesis, as previously described, as well as injury-induced angiogenesis. In conclusion, we have described regenerative angiogenesis in zebrafish for the first time and have highlighted the role of inflammation in this process. In addition, we have shown that both angiogenesis and neurogenesis are involved in brain repair and that microglia and inflammation-dependent mechanisms activated by Vegf signaling are important contributors to these processes. This study paves the way for a better understanding of the effect of Vegf on microglia and for studies aimed at promoting angiogenesis to improve brain plasticity after brain injury.
Collapse
Affiliation(s)
- Danielle Fernezelian
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint Pierre, La Réunion, France
| | - Philippe Rondeau
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint Pierre, La Réunion, France
| | - Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint Pierre, La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint Pierre, La Réunion, France
| |
Collapse
|
2
|
Lin H, He J, Ren J, Chen X, Wang T, Zhang H, Wang S, Wang M, Chen T, Duan S, He N. Targeted plasma proteomics reveals organ damage signatures of AIDS- and noncommunicable disease-related deaths in people with HIV. Nat Commun 2025; 16:3877. [PMID: 40274826 PMCID: PMC12022165 DOI: 10.1038/s41467-025-59242-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Antiretroviral therapy (ART) is shifting the primary driver of mortality for people with HIV (PWH) from opportunistic infections to noncommunicable diseases (NCDs). Protein biomarkers differentiating both AIDS-related and NCDs-related deaths from PWH may help early and precise risk prediction and intervention. We conduct a nested case-control study where 126 HIV deaths, 162 age-sex-matched HIV survivors and 152 HIV-negative controls are analyzed with 92 protein biomarkers of the Olink Organ Damage panel by proximity extension assays (PEA). Using LASSO regression, logistic regression, and ROC analysis, twelve proteins are significantly associated with HIV death, of which six (SIRT5, PPM1B, PSMA1, GALNT10, VEGFC, PTN) are specifically associated with NCDs-related death, two (RCOR1, SERPINA9) are specifically associated with AIDS-related death, and four (CA12, CA14, RARRES1, EDIL3) are associated with both. Three of these proteins are replicable in the external validation sample. The adjusted protein panels consisting of significantly associated proteins selected through both LASSO and logistic regression model well predicted NCDs-related death (AUC = 0.970) and AIDS-related death (AUC = 0.960) in PWH. The selected proteins also displayed a significant correlation with traditional biomarkers of NCDs among PWH (P < 0.05). The potential clinical utility of these biomarkers could shed light on pathogenesis of end-stage organ dysfunction in PWH.
Collapse
Affiliation(s)
- Haijiang Lin
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Jiayu He
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
- Yi-Wu Research Institute, Fudan University, Shanghai, China
| | - Jiyuan Ren
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
- Yi-Wu Research Institute, Fudan University, Shanghai, China
| | - Xiaoxiao Chen
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Tingting Wang
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Haijun Zhang
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Shanling Wang
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Miaochen Wang
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
- Yi-Wu Research Institute, Fudan University, Shanghai, China
| | - Tailin Chen
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
- Yi-Wu Research Institute, Fudan University, Shanghai, China
| | - Song Duan
- Dehong Prefecture Center for Disease Control and Prevention, Dehong Dai and Jingpo Autonomous Prefecture, Yunnan Province, China
| | - Na He
- School of Public Health, and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China.
- Yi-Wu Research Institute, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Fernezelian D, Pfitsch S, Rastegar S, Diotel N. Mapping the cellular expression patterns of vascular endothelial growth factor aa and bb genes and their receptors in the adult zebrafish brain during constitutive and regenerative neurogenesis. Neural Dev 2024; 19:17. [PMID: 39267104 PMCID: PMC11396322 DOI: 10.1186/s13064-024-00195-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
The complex interplay between vascular signaling and neurogenesis in the adult brain remains a subject of intense research. By exploiting the unique advantages of the zebrafish model, in particular the persistent activity of neural stem cells (NSCs) and the remarkable ability to repair brain lesions, we investigated the links between NSCs and cerebral blood vessels. In this study, we first examined the gene expression profiles of vascular endothelial growth factors aa and bb (vegfaa and vegfbb), under physiological and regenerative conditions. Employing fluorescence in situ hybridization combined with immunostaining and histology techniques, we demonstrated the widespread expression of vegfaa and vegfbb across the brain, and showed their presence in neurons, microglia/immune cells, endothelial cells and NSCs. At 1 day post-lesion (dpl), both vegfaa and vegfbb were up-regulated in neurons and microglia/peripheral immune cells (macrophages). Analysis of vegf receptors (vegfr) revealed high expression throughout the brain under homeostatic conditions, with vegfr predominantly expressed in neurons and NSCs and to a lower extent in microglia/immune cells and endothelial cells. These findings were further validated by Vegfr3 and Vegfr4 immunostainings, which showed significant expression in neurogenic radial glial cells.Following brain lesion (1 dpl), while vegfr gene expression remained stable, vegfr transcripts were detected in proliferative cells within the injured parenchyma. Collectively, our results provide a first overview of Vegf/Vegfr signaling in the brain and suggest important roles for Vegf in neurogenesis and regenerative processes.
Collapse
Affiliation(s)
- Danielle Fernezelian
- UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, Saint-Pierre, La Réunion, France
| | - Sabrina Pfitsch
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Nicolas Diotel
- UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, Saint-Pierre, La Réunion, France.
| |
Collapse
|
4
|
Mandal A, Moneme C, Tewari BP, Goldstein AM, Sontheimer H, Cheng L, Moore SR, Levin D. A novel method for culturing enteric neurons generates neurospheres containing functional myenteric neuronal subtypes. J Neurosci Methods 2024; 407:110144. [PMID: 38670535 PMCID: PMC11144385 DOI: 10.1016/j.jneumeth.2024.110144] [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: 12/23/2023] [Revised: 03/04/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND The enteric nervous system (ENS) is comprised of neurons, glia, and neural progenitor cells that regulate essential gastrointestinal functions. Advances in high-efficiency enteric neuron culture would facilitate discoveries surrounding ENS regulatory processes, pathophysiology, and therapeutics. NEW METHOD Development of a simple, robust, one-step method to culture murine enteric neurospheres in a 3D matrix that supports neural growth and differentiation. RESULTS Myenteric plexus cells isolated from the entire length of adult murine small intestine formed ≥3000 neurospheres within 7 days. Matrigel-embedded neurospheres exhibited abundant neural stem and progenitor cells expressing Sox2, Sox10 and Msi1 by day 4. By day 5, neural progenitor cell marker Nestin appeared in the periphery of neurospheres prior to differentiation. Neurospheres produced extensive neurons and neurites, confirmed by Tubulin beta III, PGP9.5, HuD/C, and NeuN immunofluorescence, including neural subtypes Calretinin, ChAT, and nNOS following 8 days of differentiation. Individual neurons within and external to neurospheres generated depolarization induced action potentials which were inhibited in the presence of sodium channel blocker, Tetrodotoxin. Differentiated neurospheres also contained a limited number of glia and endothelial cells. COMPARISON WITH EXISTING METHODS This novel one-step neurosphere growth and differentiation culture system, in 3D format (in the presence of GDNF, EGF, and FGF2), allows for ∼2-fold increase in neurosphere count in the derivation of enteric neurons with measurable action potentials. CONCLUSION Our method describes a novel, robust 3D culture of electrophysiologically active enteric neurons from adult myenteric neural stem and progenitor cells.
Collapse
Affiliation(s)
- Arabinda Mandal
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Chioma Moneme
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Bhanu P Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Lily Cheng
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sean R Moore
- Department of Pediatrics, Division of Pediatric Gastroenterology Hepatology, and Nutrition, University of Virginia, Charlottesville, VA, USA.
| | - Daniel Levin
- Department of Surgery, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
5
|
Luo J, Luo Y, Zhao M, Liu Y, Liu J, Du Z, Gong H, Wang L, Zhao J, Wang X, Gu Z, Zhao W, Liu T, Fan X. Fullerenols Ameliorate Social Deficiency and Rescue Cognitive Dysfunction of BTBR T +Itpr3 tf/J Autistic-Like Mice. Int J Nanomedicine 2024; 19:6035-6055. [PMID: 38911505 PMCID: PMC11192297 DOI: 10.2147/ijn.s459511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a neurodevelopmental condition that affects social interaction and communication and can cause stereotypic behavior. Fullerenols, a type of carbon nanomaterial known for its neuroprotective properties, have not yet been studied for their potential in treating ASD. We aimed to investigate its role in improving autistic behaviors in BTBR T+Itpr3tf/J (BTBR) mice and its underlying mechanism, which could provide reliable clues for future ASD treatments. METHODS Our research involved treating C57BL/6J (C57) and BTBR mice with either 0.9% NaCl or fullerenols (10 mg/kg) daily for one week at seven weeks of age. We then conducted ASD-related behavioral tests in the eighth week and used RNA-seq to screen for vital pathways in the mouse hippocampus. Additionally, we used real-time quantitative PCR (RT-qPCR) to verify related pathway genes and evaluated the number of stem cells in the hippocampal dentate gyrus (DG) by Immunofluorescence staining. RESULTS Our findings revealed that fullerenols treatment significantly improved the related ASD-like behaviors of BTBR mice, manifested by enhanced social ability and improved cognitive deficits. Immunofluorescence results showed that fullerenols treatment increased the number of DCX+ and SOX2+/GFAP+ cells in the DG region of BTBR mice, indicating an expanded neural progenitor cell (NPC) pool of BTBR mice. RNA-seq analysis of the mouse hippocampus showed that VEGFA was involved in the rescued hippocampal neurogenesis by fullerenols treatment. CONCLUSION In conclusion, our findings suggest that fullerenols treatment improves ASD-like behavior in BTBR mice by upregulating VEGFA, making nanoparticle- fullerenols a promising drug for ASD treatment.
Collapse
Affiliation(s)
- Jing Luo
- School of Life Sciences, Chongqing University, Chongqing, 401331, People’s Republic of China
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Maoru Zhao
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Yulong Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Zhulin Du
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Jinghui Zhao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Xiaqing Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Zhanjun Gu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, People’s Republic of China
| | - Wenhui Zhao
- School of Life Sciences, Chongqing University, Chongqing, 401331, People’s Republic of China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| |
Collapse
|
6
|
Chai YC, To SK, Simorgh S, Zaunz S, Zhu Y, Ahuja K, Lemaitre A, Ramezankhani R, van der Veer BK, Wierda K, Verhulst S, van Grunsven LA, Pasque V, Verfaillie C. Spatially Self-Organized Three-Dimensional Neural Concentroid as a Novel Reductionist Humanized Model to Study Neurovascular Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304421. [PMID: 38037510 PMCID: PMC10837345 DOI: 10.1002/advs.202304421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/15/2023] [Indexed: 12/02/2023]
Abstract
Although human pluripotent stem cell (PSC)-derived brain organoids have enabled researchers to gain insight into human brain development and disease, these organoids contain solely ectodermal cells and are not vascularized as occurs during brain development. Here it is created less complex and more homogenous large neural constructs starting from PSC-derived neuroprogenitor cells (NPC), by fusing small NPC spheroids into so-called concentroids. Such concentroids consisted of a pro-angiogenic core, containing neuronal and outer radial glia cells, surrounded by an astroglia-dense outer layer. Incorporating PSC-derived endothelial cells (EC) around and/or in the concentroids promoted vascularization, accompanied by differential outgrowth and differentiation of neuronal and astroglia cells, as well as the development of ectodermal-derived pericyte-like mural cells co-localizing with EC networks. Single nucleus transcriptomic analysis revealed an enhanced neural cell subtype maturation and diversity in EC-containing concentroids, which better resemble the fetal human brain compared to classical organoids or NPC-only concentroids. This PSC-derived "vascularized" concentroid brain model will facilitate the study of neurovascular/blood-brain barrier development, neural cell migration, and the development of effective in vitro vascularization strategies of brain mimics.
Collapse
Affiliation(s)
- Yoke Chin Chai
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - San Kit To
- Stem Cell Institute LeuvenDepartment of Development and RegenerationLeuven Institute for Single Cell Omics (LISCO)KU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Susan Simorgh
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Samantha Zaunz
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - YingLi Zhu
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Karan Ahuja
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Alix Lemaitre
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Roya Ramezankhani
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Bernard K. van der Veer
- Laboratory for Stem Cell and Developmental EpigeneticsDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Keimpe Wierda
- Electrophysiology Expert UnitVIB‐KU Leuven Center for Brain & Disease ResearchLeuven3000Belgium
| | - Stefaan Verhulst
- Liver Cell Biology Research GroupVrije Universiteit Brussel (VUB)Brussels1090Belgium
| | - Leo A. van Grunsven
- Liver Cell Biology Research GroupVrije Universiteit Brussel (VUB)Brussels1090Belgium
| | - Vincent Pasque
- Stem Cell Institute LeuvenDepartment of Development and RegenerationLeuven Institute for Single Cell Omics (LISCO)KU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| | - Catherine Verfaillie
- Stem Cell Institute LeuvenDepartment of Development and RegenerationKU Leuven, O&N4, Herestraat 49Leuven3000Belgium
| |
Collapse
|
7
|
Pekacar S, Özüpek B, Akkol EK, Taştan H, Ersan H, Orhan DD. Identification of bioactive components on antihemorrhoidal activity of Cistus laurifolius L. using RP-HPLC and LC-QTOF-MS. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117122. [PMID: 37660958 DOI: 10.1016/j.jep.2023.117122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cistus laurifolius is widely used in folk medicine in Anatolia for the treatment of many ailments. The leaves of the plant are used in the form of tea in the treatment of hemorrhoids in the Western Black Sea Region and Central Anatolia. AIMS OF THE STUDY It was aimed at evaluating the anti-hemorrhoidal effects of C. laurifolus leaves in croton oil-induced hemorrhoid model in rats. MATERIALS AND METHODS The methanolic and aqueous extracts of C. laurifolius were tested for in vivo anti-hemorrhoidal efficacy using an experimental hemorrhoid model, followed by histological and biochemical analysis. Hemorrhoid was created by using croton oil on the anal region of the rats. TNF-α and VEGF mRNA expression levels were assessed using real-time PCR detections. The extract was also tested for anti-inflammatory properties, which are based on the suppression of an increase in capillary permeability caused by acetic acid. LC-QTOF-MS and RP-HPLC were used for the phytochemical analysis. RESULTS In comparison to the control, histological and biochemical assessment showed that the methanolic extract of C. laurifolius is particularly effective against hemorrhoids. The same extract group's TNF-α mRNA expression was found to be the lowest. Additionally, the methanolic extract showed a strong inhibitory effect on the increase in capillary permeability resulted on by acetic acid. Three phenolic compounds were discovered in the extracts by phytochemical analyses, while more than eighteen compounds were found by LC-QTOF-MS analysis. Five of these compounds are phenolic acid derivatives, and flavonoids constitute the majority of the group. CONCLUSION This is the first evidence from the research that C. laurifolius possesses strong anti-inflammatory and anti-hemorrhoidal properties.
Collapse
Affiliation(s)
- Sultan Pekacar
- Faculty of Pharmacy, Department of Pharmacognosy, Gazi University, 06330, Ankara, Turkey.
| | - Burçin Özüpek
- Faculty of Pharmacy, Department of Pharmacognosy, Gazi University, 06330, Ankara, Turkey.
| | - Esra Küpeli Akkol
- Faculty of Pharmacy, Department of Pharmacognosy, Gazi University, 06330, Ankara, Turkey.
| | - Hakkı Taştan
- Department of Biology, Faculty of Science, Gazi University, Etiler, 06330, Ankara, Turkey.
| | - Halil Ersan
- Hükümet Konağı Çocuk Büro Amirliği, Kapaklı, 59510, Tekirdağ, Turkey.
| | - Didem Deliorman Orhan
- Faculty of Pharmacy, Department of Pharmacognosy, Gazi University, 06330, Ankara, Turkey.
| |
Collapse
|
8
|
You Z, Gao X, Kang X, Yang W, Xiong T, Li Y, Wei F, Zhuang Y, Zhang T, Sun Y, Shen H, Dai J. Microvascular endothelial cells derived from spinal cord promote spinal cord injury repair. Bioact Mater 2023; 29:36-49. [PMID: 37621772 PMCID: PMC10444976 DOI: 10.1016/j.bioactmat.2023.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/06/2023] [Accepted: 06/23/2023] [Indexed: 08/26/2023] Open
Abstract
Neural regeneration after spinal cord injury (SCI) closely relates to the microvascular endothelial cell (MEC)-mediated neurovascular unit formation. However, the effects of central nerve system-derived MECs on neovascularization and neurogenesis, and potential signaling involved therein, are unclear. Here, we established a primary spinal cord-derived MECs (SCMECs) isolation with high cell yield and purity to describe the differences with brain-derived MECs (BMECs) and their therapeutic effects on SCI. Transcriptomics and proteomics revealed differentially expressed genes and proteins in SCMECs were involved in angiogenesis, immunity, metabolism, and cell adhesion molecular signaling was the only signaling pathway enriched of top 10 in differentially expressed genes and proteins KEGG analysis. SCMECs and BMECs could be induced angiogenesis by different stiffness stimulation of PEG hydrogels with elastic modulus 50-1650 Pa for SCMECs and 50-300 Pa for BMECs, respectively. Moreover, SCMECs and BMECs promoted spinal cord or brain-derived NSC (SNSC/BNSC) proliferation, migration, and differentiation at different levels. At certain dose, SCMECs in combination with the NeuroRegen scaffold, showed higher effectiveness in the promotion of vascular reconstruction. The potential underlying mechanism of this phenomenon may through VEGF/AKT/eNOS- signaling pathway, and consequently accelerated neuronal regeneration and functional recovery of SCI rats compared to BMECs. Our findings suggested a promising role of SCMECs in restoring vascularization and neural regeneration.
Collapse
Affiliation(s)
- Zhifeng You
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xu Gao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xinyi Kang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Wen Yang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tiandi Xiong
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yue Li
- i-Lab, Key Laboratory of Multifunction Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Feng Wei
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Ting Zhang
- i-Lab, Key Laboratory of Multifunction Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yifu Sun
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
9
|
Winkelman MA, Dai G. Bioengineered perfused human brain microvascular networks enhance neural progenitor cell survival, neurogenesis, and maturation. SCIENCE ADVANCES 2023; 9:eaaz9499. [PMID: 37163593 PMCID: PMC10171804 DOI: 10.1126/sciadv.aaz9499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/10/2023] [Indexed: 05/12/2023]
Abstract
Neural progenitor cells (NPCs) have the capability to self-renew and differentiate into neurons and glial cells. In the adult brain, NPCs are found near brain microvascular networks (BMVNs) in specialized microenvironments called the neurovascular niche (NVN). Although several in vitro NVN models have been previously reported, most do not properly recapitulate the intimate cellular interactions between NPCs and perfused brain microvessels. Here, we developed perfused BMVNs composed of primary human brain endothelial cells, pericytes, and astrocytes within microfluidic devices. When induced pluripotent stem cell-derived NPCs were introduced into BMVNs, we found that NPC survival, neurogenesis, and maturation were enhanced. The application of flow during BMVN coculture was also beneficial for neuron differentiation. Collectively, our work highlighted the important role of BMVNs and flow in NPC self-renewal and neurogenesis, as well as demonstrated our model's potential to study the biological and physical interactions of human NVN in vitro.
Collapse
Affiliation(s)
- Max A. Winkelman
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | | |
Collapse
|
10
|
Karakatsani A, Álvarez-Vergara MI, de Almodóvar CR. The vasculature of neurogenic niches: Properties and function. Cells Dev 2023; 174:203841. [PMID: 37060947 DOI: 10.1016/j.cdev.2023.203841] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
In the adult rodent brain, neural stem cells (NSCs) reside in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the hippocampus. In these areas, NSCs and their progeny integrate intrinsic signals and extrinsic cues provided by their microenvironment that control their behavior. The vasculature in the SVZ and SGZ, and the choroid plexus (ChP) in the SVZ, have emerged as critical compartments of the neurogenic niches as they provide a rich repertoire of cues to regulate NSC quiescence, proliferation, self-renewal and differentiation. Physical contact between NSCs and blood vessels is also a feature within the niches and supports different processes such as quiescence, migration and vesicle transport. In this review, we provide a description of the brain and choroid plexus vasculature in both stem cell niches, highlighting the main properties and role of the vasculature in each niche. We also summarize the current understanding of how blood vessel- and ChP-derived signals influence the behavior of NSCs in physiological adulthood, as well as upon aging.
Collapse
Affiliation(s)
- Andromachi Karakatsani
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - María I Álvarez-Vergara
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Carmen Ruiz de Almodóvar
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, University of Bonn, Bonn, Germany; Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Yazdani N, Willits RK. Mimicking the neural stem cell niche: An engineer’s view of cell: material interactions. FRONTIERS IN CHEMICAL ENGINEERING 2023. [DOI: 10.3389/fceng.2022.1086099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells have attracted attention in recent years to treat neurodegeneration. There are two neurogenic regions in the brain where neural stem cells reside, one of which is called the subventricular zone (SVZ). The SVZ niche is a complicated microenvironment providing cues to regulate self-renewal and differentiation while maintaining the neural stem cell’s pool. Many scientists have spent years understanding the cellular and structural characteristics of the SVZ niche, both in homeostasis and pathological conditions. On the other hand, engineers focus primarily on designing platforms using the knowledge they acquire to understand the effect of individual factors on neural stem cell fate decisions. This review provides a general overview of what we know about the components of the SVZ niche, including the residing cells, extracellular matrix (ECM), growth factors, their interactions, and SVZ niche changes during aging and neurodegenerative diseases. Furthermore, an overview will be given on the biomaterials used to mimic neurogenic niche microenvironments and the design considerations applied to add bioactivity while meeting the structural requirements. Finally, it will discuss the potential gaps in mimicking the microenvironment.
Collapse
|
12
|
Li M, Gao L, Zhao L, Zou T, Xu H. Toward the next generation of vascularized human neural organoids. Med Res Rev 2023; 43:31-54. [PMID: 35993813 DOI: 10.1002/med.21922] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/22/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Thanks to progress in the development of three-dimensional (3D) culture technologies, human central nervous system (CNS) development and diseases have been gradually deciphered by using organoids derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs). Selforganized neural organoids (NOs) have been used to mimic morphogenesis and functions of specific organs in vitro. Many NOs have been reproduced in vitro, such as those mimicking the human brain, retina, and spinal cord. However, NOs fail to capitulate to the maturation and complexity of in vivo neural tissues. The persistent issues with current NO cultivation protocols are inadequate oxygen supply and nutrient diffusion due to the absence of vascular networks. In vivo, the developing CNS is interpenetrated by vasculature that not only supplies oxygen and nutrients but also provides a structural template for neuronal growth. To address these deficiencies, recent studies have begun to couple NO culture with bioengineering techniques and methodologies, including genetic engineering, coculture, multidifferentiation, microfluidics and 3D bioprinting, and transplantation, which might promote NO maturation and create more functional NOs. These cutting-edge methods could generate an ever more reliable NO model in vitro for deciphering the codes of human CNS development, disease progression, and translational application. In this review, we will summarize recent technological advances in culture strategies to generate vascularized NOs (vNOs), with a special focus on cerebral- and retinal-organoid models.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing, China
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
13
|
Malheiro A, Seijas-Gamardo A, Harichandan A, Mota C, Wieringa P, Moroni L. Development of an In Vitro Biomimetic Peripheral Neurovascular Platform. ACS APPLIED MATERIALS & INTERFACES 2022; 14:31567-31585. [PMID: 35815638 PMCID: PMC9305708 DOI: 10.1021/acsami.2c03861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Nerves and blood vessels are present in most organs and are indispensable for their function and homeostasis. Within these organs, neurovascular (NV) tissue forms congruent patterns and establishes vital interactions. Several human pathologies, including diabetes type II, produce NV disruptions with serious consequences that are complicated to study using animal models. Complex in vitro organ platforms, with neural and vascular supply, allow the investigation of such interactions, whether in a normal or pathological context, in an affordable, simple, and direct manner. To date, a few in vitro models contain NV tissue, and most strategies report models with nonbiomimetic representations of the native environment. To this end, we have established here an NV platform that contains mature vasculature and neural tissue, composed of human microvascular endothelial cells (HMVECs), induced pluripotent stem cell (iPSCs)-derived sensory neurons, and primary rat Schwann cells (SCs) within a fibrin-embedded polymeric scaffold. First, we show that SCs can induce the formation of and stabilize vascular networks to the same degree as the traditional and more thoroughly studied human dermal fibroblasts (HDFs). We also show that through SC prepatterning, we are able to control vessel orientation. Using our NV platform, we demonstrate the concomitant formation of three-dimensional neural and vascular tissue, and the influence of different medium formulations and cell types on the NV tissue outcome. Finally, we propose a protocol to form mature NV tissue, via the integration of independent neural and vascular constituents. The platform described here provides a versatile and advanced model for in vitro research of the NV axis.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Adrián Seijas-Gamardo
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Abhishek Harichandan
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Carlos Mota
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| |
Collapse
|
14
|
Li Y, Schor J, Bartko J, Albert G, Halterman MW. The transcription factor C/EBPβ promotes vascular endothelial growth factor A expression and neural stem cell expansion. FEBS Lett 2022; 596:1661-1671. [PMID: 35593120 DOI: 10.1002/1873-3468.14405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/28/2022] [Accepted: 05/03/2022] [Indexed: 11/11/2022]
Abstract
The bZIP transcription factor CCAAT enhancer-binding protein β (C/EBPβ) exhibits neurogenic, neuritogenic, and pro-survival effects in the central nervous system. Here, we show that C/EBPβ regulates neural stem cell (NSC) expansion and vascular endothelial growth factor A (VEGF-A) level by acting on a C/EBPβ-responsive element within the Vegf-a promoter. As predicted, C/EBPβ depletion reduced VEGF-A production, NSC number, and average neurosphere size in proliferating cultures. Conversely, deletion of the C/EBPβ repressor CHOP-10 induced C/EBPβ and VEGF-A expression, while stimulating NSC expansion. These data highlight the role of C/EBPβ in regulating VEGF-A production and the growth of NSCs and suggest CHOP-dependent antagonism of C/EBPβ may function as a transcriptional rheostat linking stress-associated cues with stem cell quiescence among other pathological responses affecting the neurogenic niche.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794
| | - Jonathan Schor
- Department of Neurology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, 14642
| | - Jonathan Bartko
- Department of Neurology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, 14642
| | - George Albert
- Department of Neurology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, 14642
| | - Marc W Halterman
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794
| |
Collapse
|
15
|
Vogenstahl J, Parrilla M, Acker-Palmer A, Segarra M. Vascular Regulation of Developmental Neurogenesis. Front Cell Dev Biol 2022; 10:890852. [PMID: 35573692 PMCID: PMC9099230 DOI: 10.3389/fcell.2022.890852] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Evolutionary studies indicate that the nervous system evolved prior to the vascular system, but the increasing complexity of organisms prompted the vascular system to emerge in order to meet the growing demand for oxygen and nutrient supply. In recent years, it has become apparent that the symbiotic communication between the nervous and the vascular systems goes beyond the exclusive covering of the demands on nutrients and oxygen carried by blood vessels. Indeed, this active interplay between both systems is crucial during the development of the central nervous system (CNS). Several neural-derived signals that initiate and regulate the vascularization of the CNS have been described, however less is known about the vascular signals that orchestrate the development of the CNS cytoarchitecture. Here, we focus on reviewing the effects of blood vessels in the process of neurogenesis during CNS development in vertebrates. In mammals, we describe the spatiotemporal features of vascular-driven neurogenesis in two brain regions that exhibit different neurogenic complexity in their germinal zone, the hindbrain and the forebrain.
Collapse
Affiliation(s)
- Johanna Vogenstahl
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Marta Parrilla
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
- *Correspondence: Amparo Acker-Palmer, ; Marta Segarra,
| | - Marta Segarra
- Neuro and Vascular Guidance Group, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
- *Correspondence: Amparo Acker-Palmer, ; Marta Segarra,
| |
Collapse
|
16
|
Regulating Endogenous Neural Stem Cell Activation to Promote Spinal Cord Injury Repair. Cells 2022; 11:cells11050846. [PMID: 35269466 PMCID: PMC8909806 DOI: 10.3390/cells11050846] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects millions of individuals worldwide. Currently, there is no cure, and treatment options to promote neural recovery are limited. An innovative approach to improve outcomes following SCI involves the recruitment of endogenous populations of neural stem cells (NSCs). NSCs can be isolated from the neuroaxis of the central nervous system (CNS), with brain and spinal cord populations sharing common characteristics (as well as regionally distinct phenotypes). Within the spinal cord, a number of NSC sub-populations have been identified which display unique protein expression profiles and proliferation kinetics. Collectively, the potential for NSCs to impact regenerative medicine strategies hinges on their cardinal properties, including self-renewal and multipotency (the ability to generate de novo neurons, astrocytes, and oligodendrocytes). Accordingly, endogenous NSCs could be harnessed to replace lost cells and promote structural repair following SCI. While studies exploring the efficacy of this approach continue to suggest its potential, many questions remain including those related to heterogeneity within the NSC pool, the interaction of NSCs with their environment, and the identification of factors that can enhance their response. We discuss the current state of knowledge regarding populations of endogenous spinal cord NSCs, their niche, and the factors that regulate their behavior. In an attempt to move towards the goal of enhancing neural repair, we highlight approaches that promote NSC activation following injury including the modulation of the microenvironment and parenchymal cells, pharmaceuticals, and applied electrical stimulation.
Collapse
|
17
|
Hongjin W, Han C, Baoxiang J, Shiqi Y, Xiaoyu X. Reconstituting neurovascular unit based on the close relations between neural stem cells and endothelial cells: an effective method to explore neurogenesis and angiogenesis. Rev Neurosci 2021; 31:143-159. [PMID: 31539363 DOI: 10.1515/revneuro-2019-0023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
The discovery of neural stem cells (NSCs) and their microenvironment, the NSC niche, brought new therapeutic strategies through neurogenesis and angiogenesis for stroke and most neurodegenerative diseases, including Alzheimer's disease. Based on the close links between NSCs and endothelial cells, the integration of neurogenesis and angiogenesis of the NSC niche is also a promising area to the neurovascular unit (NVU) modeling and is now offering a powerful tool to advance our understanding of the brain. In this review, critical aspects of the NVU and model systems are discussed. First, we briefly describe the interaction of each part in the NSC niche. Second, we introduce the co-culture system, microfluidic platforms, and stem cell-derived 3D reconstitution used in NVU modeling based on the close relations between NSCs and endothelial cells, and various characteristics of cell interactions in these systems are also described. Finally, we address the challenges in modeling the NVU that can potentially be overcome by employing strategies for advanced biomaterials and stem cell co-culture use. Based on these approaches, researchers will continue to develop predictable technologies to control the fate of stem cells, achieve accurate screening of drugs for the nervous system, and advance the clinical application of NVU models.
Collapse
Affiliation(s)
- Wang Hongjin
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.,Chongqing Key Laboratory of New Drug Screening From Traditional Chinese Medicine, Chongqing 400715, China.,Pharmacology of Chinese Materia Medica-Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Chen Han
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.,Chongqing Key Laboratory of New Drug Screening From Traditional Chinese Medicine, Chongqing 400715, China.,Pharmacology of Chinese Materia Medica-Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Jiang Baoxiang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.,Chongqing Key Laboratory of New Drug Screening From Traditional Chinese Medicine, Chongqing 400715, China.,Pharmacology of Chinese Materia Medica-Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Yu Shiqi
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.,Chongqing Key Laboratory of New Drug Screening From Traditional Chinese Medicine, Chongqing 400715, China.,Pharmacology of Chinese Materia Medica-Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Xu Xiaoyu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.,Chongqing Key Laboratory of New Drug Screening From Traditional Chinese Medicine, Chongqing 400715, China.,Pharmacology of Chinese Materia Medica-Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| |
Collapse
|
18
|
Tejeda G, Ciciriello AJ, Dumont CM. Biomaterial Strategies to Bolster Neural Stem Cell-Mediated Repair of the Central Nervous System. Cells Tissues Organs 2021; 211:655-669. [PMID: 34120118 DOI: 10.1159/000515351] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/12/2021] [Indexed: 01/25/2023] Open
Abstract
Stem cell therapies have the potential to not only repair, but to regenerate tissue of the central nervous system (CNS). Recent studies demonstrate that transplanted stem cells can differentiate into neurons and integrate with the intact circuitry after traumatic injury. Unfortunately, the positive findings described in rodent models have not been replicated in clinical trials, where the burden to maintain the cell viability necessary for tissue repair becomes more challenging. Low transplant survival remains the greatest barrier to stem cell-mediated repair of the CNS, often with fewer than 1-2% of the transplanted cells remaining after 1 week. Strategic transplantation parameters, such as injection location, cell concentration, and transplant timing achieve only modest improvements in stem cell transplant survival and appear inconsistent across studies. Biomaterials provide researchers with a means to significantly improve stem cell transplant survival through two mechanisms: (1) a vehicle to deliver and protect the stem cells and (2) a substrate to control the cytotoxic injury environment. These biomaterial strategies can alleviate cell death associated with delivery to the injury and can be used to limit cell death after transplantation by limiting cell exposure to cytotoxic signals. Moreover, it is likely that control of the injury environment with biomaterials will lead to a more reliable support for transplanted cell populations. This review will highlight the challenges associated with cell delivery in the CNS and the advances in biomaterial development and deployment for stem cell therapies necessary to bolster stem cell-mediated repair.
Collapse
Affiliation(s)
- Giancarlo Tejeda
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| |
Collapse
|
19
|
Winkelman MA, Koppes AN, Koppes RA, Dai G. Bioengineering the neurovascular niche to study the interaction of neural stem cells and endothelial cells. APL Bioeng 2021; 5:011507. [PMID: 33688617 PMCID: PMC7932757 DOI: 10.1063/5.0027211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
The ability of mammalian neural stem cells (NSCs) to self-renew and differentiate throughout adulthood has made them ideal to study neurogenesis and attractive candidates for neurodegenerative disease therapies. In the adult mammalian brain, NSCs are maintained in the neurovascular niche (NVN) where they are found near the specialized blood vessels, suggesting that brain endothelial cells (BECs) are prominent orchestrators of NSC fate. However, most of the current knowledge of the mammalian NVN has been deduced from nonhuman studies. To circumvent the challenges of in vivo studies, in vitro models have been developed to better understand the reciprocal cellular mechanisms of human NSCs and BECs. This review will cover the current understanding of mammalian NVN biology, the effects of endothelial cell-derived signals on NSC fate, and the in vitro models developed to study the interactions between NSCs and BECs.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
20
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
21
|
Okabe K, Fukada H, Tai-Nagara I, Ando T, Honda T, Nakajima K, Takeda N, Fong GH, Ema M, Kubota Y. Neuron-derived VEGF contributes to cortical and hippocampal development independently of VEGFR1/2-mediated neurotrophism. Dev Biol 2020; 459:65-71. [PMID: 31790655 DOI: 10.1016/j.ydbio.2019.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/07/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a potent mitogen critical for angiogenesis and organogenesis. Deletion or inhibition of VEGF during development not only profoundly suppresses vascular outgrowth, but significantly affects the development and function of various organs. In the brain, VEGF is thought to not only promote vascular growth, but also directly act on neurons as a neurotrophic factor by activating VEGF receptors. In the present study, we demonstrated that deletion of VEGF using hGfap-Cre line, which recombines genes specifically in cortical and hippocampal neurons, severely impaired brain organization and vascularization of these regions. The mutant mice had motor deficits, with lethality around the time of weaning. Multiple reporter lines indicated that VEGF was highly expressed in neurons, but that its cognate receptors, VEGFR1 and 2 were exclusive to endothelial cells in the brain. In accordance, mice lacking neuronal VEGFR1 and VEGFR2 did not exhibit neuronal deformities or lethality. Taken together, our data suggest that neuron-derived VEGF contributes to cortical and hippocampal development likely through angiogenesis independently of direct neurotrophic effects mediated by VEGFR1 and 2.
Collapse
Affiliation(s)
- Keisuke Okabe
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Plastic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Hugh Fukada
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ikue Tai-Nagara
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Tomofumi Ando
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takao Honda
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06032, USA; Department of Cell Biology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06032, USA
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
22
|
Adult Neurogenesis in the Subventricular Zone and Its Regulation After Ischemic Stroke: Implications for Therapeutic Approaches. Transl Stroke Res 2019; 11:60-79. [DOI: 10.1007/s12975-019-00717-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
23
|
Angiogenic potential of co-spheroids of neural stem cells and endothelial cells in injectable gelatin-based hydrogel. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:140-149. [DOI: 10.1016/j.msec.2019.01.089] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/12/2018] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
|
24
|
Deng S, Zeng Y, Wu L, Hu Z, Shen J, Shen Y, Shen Y, Zhou Y, Chen J, Lin S. The regulatory roles of VEGF-Notch signaling pathway on aplastic anemia with kidney deficiency and blood stasis. J Cell Biochem 2019; 120:2078-2089. [PMID: 30230583 DOI: 10.1002/jcb.27516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/27/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF)-Notch signaling pathway plays an important role in aplastic anemia (AA). This study aimed to evaluate the regulatory roles of VEGF-Notch signaling pathway on mesenchymal stem cells (MSCs) isolated from AA patients with kidney deficiency and blood stasis (KB) (AA MSCs). METHODS Expression of VEGF-Notch signaling related factors, including VEGF, VEGFR, Notch-1, Jagged1, Delta-like1, and hes1 was detected in bone marrow (BM) tissues and AA MSCs by Western blot analysis. VEGF (100 ng/mL) and γ-secretase inhibitor (DAPT) (10 μM) was used to active and inhibit VEGF-Notch signaling in AA MSCs, respectively. After treatment, the proliferation, apoptosis, and adipogenic differentiation of AA MSCs was detected by Cell Counting Kit-8, flow cytometry, and Oil red O staining, respectively. Lentivirus short hairpin RNA (shRNA) were constructed to downregulate Notch-1 and VEGF in normal bone marrow mesenchymal stem cells (BMSCs), and the effects of VEGF/Notch-1 shRNA transfected BMSCs on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were evaluated. RESULTS Significantly lower expression of VEGF, VEGFR, Notch-1, Jagged1, Delta-like1, and hes1 was revealed in AA BM tissues and AA MSCs when compared with the normal control (P < 0.05). The intervention of DAPT significantly inhibited the proliferation, and promoted the apoptosis and adipogenic differentiation of AA MSCs, while VEGF intervention exhibited opposite results (P < 0.05). Meanwhile, the proliferation, migration, and angiogenesis of HUVECs were significantly promoted by normal BMSCs, while inhibited by VEGF/Notch-1 shRNA transfected BMSCs (P < 0.05). CONCLUSION The activation of VEGF-Notch signaling pathway may be a potential therapeutic target for AA with KB.
Collapse
Affiliation(s)
- Shu Deng
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuqing Zeng
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Liqiang Wu
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiping Hu
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianping Shen
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiping Shen
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingying Shen
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuhong Zhou
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian Chen
- Department of Scientific Research, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Shengyun Lin
- Department of Hematology, First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
25
|
Lin R, Cai J, Kenyon L, Iozzo R, Rosenwasser R, Iacovitti L. Systemic Factors Trigger Vasculature Cells to Drive Notch Signaling and Neurogenesis in Neural Stem Cells in the Adult Brain. Stem Cells 2018; 37:395-406. [PMID: 30431198 PMCID: PMC7028145 DOI: 10.1002/stem.2947] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 10/19/2018] [Accepted: 10/25/2018] [Indexed: 01/10/2023]
Abstract
It is well documented that adult neural stem cells (NSCs) residing in the subventricular zone (SVZ) and the subgranular zone (SGZ) are induced to proliferate and differentiate into new neurons after injury such as stroke and hypoxia. However, the role of injury‐related cues in driving this process and the means by which they communicate with NSCs remains largely unknown. Recently, the coupling of neurogenesis and angiogenesis and the extensive close contact between vascular cells and other niche cells, known as the neurovascular unit (NVU), has attracted interest. Further facilitating communication between blood and NSCs is a permeable blood‐brain‐barrier (BBB) present in most niches, making vascular cells a potential conduit between systemic signals, such as vascular endothelial growth factor (VEGF), and NSCs in the niche, which could play an important role in regulating neurogenesis. We show that the leaky BBB in stem cell niches of the intact and stroke brain can respond to circulating VEGF165 to drive induction of the Notch ligand DLL4 (one of the most important cues in angiogenesis) in endothelial cells (ECs), pericytes, and further induce significant proliferation and neurogenesis of stem cells. Stem Cells2019;37:395–406
Collapse
Affiliation(s)
- Ruihe Lin
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jingli Cai
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lawrence Kenyon
- Department of Pathology, Anatomy, & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato Iozzo
- Department of Pathology, Anatomy, & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert Rosenwasser
- The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Jia C, Keasey MP, Malone HM, Lovins C, Sante RR, Razskazovskiy V, Hagg T. Vitronectin from brain pericytes promotes adult forebrain neurogenesis by stimulating CNTF. Exp Neurol 2018; 312:20-32. [PMID: 30408465 DOI: 10.1016/j.expneurol.2018.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/17/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022]
Abstract
Vitronectin (VTN) is a glycoprotein in the blood and affects hemostasis. VTN is also present in the extracellular matrix of various organs but little is known about its function in healthy adult tissues. We show, in adult mice, that VTN is uniquely expressed by approximately half of the pericytes of subventricular zone (SVZ) where neurogenesis continues throughout life. Intracerebral VTN antibody injection or VTN knockout reduced neurogenesis as well as expression of pro-neurogenic CNTF, and anti-neurogenic LIF and IL-6. Conversely, injections of VTN, or plasma from VTN+/+, but not VTN-/- mice, increased these cytokines. VTN promoted SVZ neurogenesis when LIF and IL-6 were suppressed by co-administration of a gp130 inhibitor. Unexpectedly, VTN inhibited FAK signaling and VTN-/- mice had increased FAK signaling in the SVZ. Further, an FAK inhibitor or VTN increased CNTF expression, but not in conditional astrocytic FAK knockout mice, suggesting that VTN increases CNTF through FAK inhibition in astrocytes. These results identify a novel role of pericyte-derived VTN in the brain, where it regulates SVZ neurogenesis through co-expression of CNTF, LIF and IL-6. VTN-integrin-FAK and gp130 signaling may provide novel targets to induce neurogenesis for cell replacement therapies.
Collapse
Affiliation(s)
- Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Matthew P Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Hannah M Malone
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Chiharu Lovins
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Richard R Sante
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Vlad Razskazovskiy
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Theo Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.
| |
Collapse
|
27
|
Xue L, Huang J, Zhang T, Wang X, Fu J, Geng Z, Zhao Y, Chen H. PTEN inhibition enhances angiogenesis in an in vitro model of ischemic injury by promoting Akt phosphorylation and subsequent hypoxia inducible factor-1α upregulation. Metab Brain Dis 2018; 33:1679-1688. [PMID: 29936638 DOI: 10.1007/s11011-018-0276-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/19/2018] [Indexed: 11/26/2022]
Abstract
Angiogenesis is an important pathophysiological response to cerebral ischemia. PTEN is a lipid phosphatase whose loss activates PI3K/Akt signaling, which is related to HIF-1α upregulation and enhanced angiogenesis in human cancer cells. However, the specific roles of PTEN in endothelial cell functions and angiogenesis after cerebral ischemia remain unknown. Therefore, we sought to examine the potential effects of PTEN inhibition on post-ischemic angiogenesis in human blood vessel cells and to determine the underlying mechanism. In this present study, human umbilical vein endothelial cells (HUVECs) were exposed to oxygen-glucose deprivation (OGD), cell proliferation, migration and apoptosis, in vitro tube formation and expression of PTEN/Akt pathway and angiogenic factors were examined in HUVECs after treatment with PTEN inhibitor bisperoxovanadium (bpV) at different doses. The results showed that bpV significantly increased the cell proliferation and reduced cell apoptosis indicating that the drug exerts a cytoprotective effect on HUVECs with OGD exposure. bpV also enhanced cell migration and tube formation in HUVECs following OGD, and upregulated HIF-1α and VEGF expressions, but attenuated endostatin expression. Additionally, western blotting analysis demonstrated that Akt phosphorylation in HUVECs was significantly increased after bpV treatment. These findings suggest that PTEN inhibition promotes post-ischemic angiogenesis in HUVECs after exposure to OGD and this enhancing effect might be achieved through activation of the Akt signal cascade.
Collapse
Affiliation(s)
- Lixia Xue
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiankang Huang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ting Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiuzhe Wang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jianliang Fu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhi Geng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hao Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Xuhui District, Shanghai, 20033, China.
| |
Collapse
|
28
|
Paredes I, Himmels P, Ruiz de Almodóvar C. Neurovascular Communication during CNS Development. Dev Cell 2018; 45:10-32. [PMID: 29634931 DOI: 10.1016/j.devcel.2018.01.023] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/22/2017] [Accepted: 01/08/2018] [Indexed: 12/11/2022]
Abstract
A precise communication between the nervous and the vascular systems is crucial for proper formation and function of the central nervous system (CNS). Interestingly, this communication does not only occur by neural cells regulating the growth and properties of the vasculature, but new studies show that blood vessels actively control different neurodevelopmental processes. Here, we review the current knowledge on how neurons in particular influence growing blood vessels during CNS development and on how vessels participate in shaping the neural compartment. We also review the identified molecular mechanisms of this bidirectional communication.
Collapse
Affiliation(s)
- Isidora Paredes
- Biochemistry Center, Heidelberg University, 69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Patricia Himmels
- Biochemistry Center, Heidelberg University, 69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Carmen Ruiz de Almodóvar
- Biochemistry Center, Heidelberg University, 69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
29
|
Parry SM, Peeples ES. The impact of hypoxic-ischemic brain injury on stem cell mobilization, migration, adhesion, and proliferation. Neural Regen Res 2018; 13:1125-1135. [PMID: 30028311 PMCID: PMC6065219 DOI: 10.4103/1673-5374.235012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy continues to be a significant cause of death or neurodevelopmental delays despite standard use of therapeutic hypothermia. The use of stem cell transplantation has recently emerged as a promising supplemental therapy to further improve the outcomes of infants with hypoxic-ischemic encephalopathy. After the injury, the brain releases several chemical mediators, many of which communicate directly with stem cells to encourage mobilization, migration, cell adhesion and differentiation. This manuscript reviews the biomarkers that are released from the injured brain and their interactions with stem cells, providing insight regarding how their upregulation could improve stem cell therapy by maximizing cell delivery to the injured tissue.
Collapse
Affiliation(s)
- Stephanie M Parry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
30
|
Huang L, Wang G. The Effects of Different Factors on the Behavior of Neural Stem Cells. Stem Cells Int 2017; 2017:9497325. [PMID: 29358957 PMCID: PMC5735681 DOI: 10.1155/2017/9497325] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/18/2017] [Indexed: 02/07/2023] Open
Abstract
The repair of central nervous system (CNS) injury has been a worldwide problem in the biomedical field. How to reduce the damage to the CNS and promote the reconstruction of the damaged nervous system structure and function recovery has always been the concern of nerve tissue engineering. Multiple differentiation potentials of neural stem cell (NSC) determine the application value for the repair of the CNS injury. Thus, how to regulate the behavior of NSCs becomes the key to treating the CNS injury. So far, a large number of researchers have devoted themselves to searching for a better way to regulate the behavior of NSCs. This paper summarizes the effects of different factors on the behavior of NSCs in the past 10 years, especially on the proliferation and differentiation of NSCs. The final purpose of this review is to provide a more detailed theoretical basis for the clinical repair of the CNS injury by nerve tissue engineering.
Collapse
Affiliation(s)
- Lixiang Huang
- Department of Chemistry and Biology, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Gan Wang
- Department of Chemistry and Biology, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China
| |
Collapse
|
31
|
Tang X, Gao J, Jia X, Zhao W, Zhang Y, Pan W, He J. Bipotent progenitors as embryonic origin of retinal stem cells. J Cell Biol 2017; 216:1833-1847. [PMID: 28465291 PMCID: PMC5461025 DOI: 10.1083/jcb.201611057] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/01/2017] [Accepted: 04/03/2017] [Indexed: 01/24/2023] Open
Abstract
In lower vertebrates, retinal stem cells (RSCs) capable of producing all retinal cell types are a resource for retinal tissue growth throughout life. However, the embryonic origin of RSCs remains largely elusive. Using a Zebrabow-based clonal analysis, we characterized the RSC niche in the ciliary marginal zone of zebrafish retina and illustrate that blood vessels associated with RSCs are required for the maintenance of actively proliferating RSCs. Full lineage analysis of RSC progenitors reveals lineage patterns of RSC production. Moreover, in vivo lineage analysis demonstrates that these RSC progenitors are the direct descendants of a set of bipotent progenitors in the medial epithelial layer of developing optic vesicles, suggesting the involvement of the mixed-lineage states in the RSC lineage specification.
Collapse
Affiliation(s)
- Xia Tang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianan Gao
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinling Jia
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wencao Zhao
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Yijie Zhang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weijun Pan
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
32
|
Rosa AI, Grade S, Santos SD, Bernardino L, Chen TC, Relvas J, Hofman FM, Agasse F. Heterocellular Contacts with Mouse Brain Endothelial Cells Via Laminin and α6β1 Integrin Sustain Subventricular Zone (SVZ) Stem/Progenitor Cells Properties. Front Cell Neurosci 2016; 10:284. [PMID: 28018177 PMCID: PMC5156690 DOI: 10.3389/fncel.2016.00284] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023] Open
Abstract
Neurogenesis in the subventricular zone (SVZ) is regulated by diffusible factors and cell–cell contacts. In vivo, SVZ stem cells are associated with the abluminal surface of blood vessels and such interactions are thought to regulate their neurogenic capacity. SVZ neural stem cells (NSCs) have been described to contact endothelial-derived laminin via α6β1 integrin. To elucidate whether heterocellular contacts with brain endothelial cells (BEC) regulate SVZ cells neurogenic capacities, cocultures of SVZ neurospheres and primary BEC, both obtained from C57BL/6 mice, were performed. The involvement of laminin-integrin interactions in SVZ homeostasis was tested in three ways. Firstly, SVZ cells were analyzed following incubation of BEC with the protein synthesis inhibitor cycloheximide (CHX) prior to coculture, a treatment expected to decrease membrane proteins. Secondly, SVZ cells were cocultured with BEC in the presence of an anti-α6 integrin neutralizing antibody. Thirdly, BEC were cultured with β1−/− SVZ cells. We showed that contact with BEC supports, at least in part, proliferation and stemness of SVZ cells, as evaluated by the number of BrdU positive (+) and Sox2+ cells in contact with BEC. These effects are dependent on BEC-derived laminin binding to α6β1 integrin and are decreased in cocultures incubated with anti-α6 integrin neutralizing antibody and in cocultures with SVZ β1−/− cells. Moreover, BEC-derived laminin sustains stemness in SVZ cell cultures via activation of the Notch and mTOR signaling pathways. Our results show that BEC/SVZ interactions involving α6β1 integrin binding to laminin, contribute to SVZ cell proliferation and stemness.
Collapse
Affiliation(s)
- Alexandra I Rosa
- Centre for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Department of Pathology, University of Southern CaliforniaLos Angeles, CA, USA; Department of Neurological Surgery, University of Southern CaliforniaLos Angeles, CA, USA
| | - Sofia Grade
- Centre for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Sofia D Santos
- Glial Cell Biology Group, Institute for Molecular and Cell Biology - IBMCPorto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal
| | - Liliana Bernardino
- Centre for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Health Sciences Research Centre, Faculty of Health Sciences, University of Beira InteriorCovilhã, Portugal
| | - Thomas C Chen
- Department of Neurological Surgery, University of Southern California Los Angeles, CA, USA
| | - João Relvas
- Glial Cell Biology Group, Institute for Molecular and Cell Biology - IBMCPorto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do PortoPorto, Portugal
| | - Florence M Hofman
- Department of Pathology, University of Southern California Los Angeles, CA, USA
| | - Fabienne Agasse
- Centre for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Department of Pathology, University of Southern CaliforniaLos Angeles, CA, USA; Department of Neurological Surgery, University of Southern CaliforniaLos Angeles, CA, USA
| |
Collapse
|
33
|
Arulmoli J, Wright HJ, Phan DTT, Sheth U, Que RA, Botten GA, Keating M, Botvinick EL, Pathak MM, Zarembinski TI, Yanni DS, Razorenova OV, Hughes CCW, Flanagan LA. Combination scaffolds of salmon fibrin, hyaluronic acid, and laminin for human neural stem cell and vascular tissue engineering. Acta Biomater 2016; 43:122-138. [PMID: 27475528 PMCID: PMC5386322 DOI: 10.1016/j.actbio.2016.07.043] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/29/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVβ1 and α5β1, and several laminin binding integrins (α7β1, α6β1, α3β1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.
Collapse
Affiliation(s)
- Janahan Arulmoli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Heather J Wright
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Duc T T Phan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Urmi Sheth
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard A Que
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Giovanni A Botten
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark Keating
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Medha M Pathak
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | | | - Daniel S Yanni
- Disc Comfort, Inc., 351 Hospital Road, Suite 202, Newport Beach, CA 92663, USA
| | - Olga V Razorenova
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lisa A Flanagan
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
34
|
Abstract
Immunomodulators regulate stem cell activity at all stages of development as well as during adulthood. Embryonic stem cell (ESC) proliferation as well as neurogenic processes during embryonic development are controlled by factors of the immune system. We review here immunophenotypic expression patterns of different stem cell types, including ESC, neural (NSC) and tissue-specific mesenchymal stem cells (MSC), and focus on immunodulatory properties of these cells. Immune and inflammatory responses, involving actions of cytokines as well as toll-like receptor (TLR) signaling are known to affect the differentiation capacity of NSC and MSC. Secretion of pro- and anti-inflammatory messengers by MSC have been observed as the consequence of TLR and cytokine activation and promotion of differentiation into specified phenotypes. As result of augmented differentiation capacity, stem cells secrete angiogenic factors including vascular endothelial growth factor, resulting in multifactorial actions in tissue repair. Immunoregulatory properties of tissue specific adult stem cells are put into the context of possible clinical applications.
Collapse
|
35
|
Kim DH, Moon EY, Yi JH, Lee HE, Park SJ, Ryu YK, Kim HC, Lee S, Ryu JH. Peptide fragment of thymosin β4 increases hippocampal neurogenesis and facilitates spatial memory. Neuroscience 2015; 310:51-62. [PMID: 26363149 DOI: 10.1016/j.neuroscience.2015.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/01/2015] [Accepted: 09/01/2015] [Indexed: 12/16/2022]
Abstract
Although several studies have suggested the neuroprotective effect of thymosin β4 (TB4), a major actin-sequestering protein, on the central nervous system, little is understood regarding the action of N-acetyl-serylaspartyl-lysyl-proline (Ac-SDKP), a peptide fragment of TB4 on brain function. Here, we examined neurogenesis-stimulative effect of Ac-SDKP. Intrahippocampal infusion of Ac-SDKP facilitated the generation of new neurons in the hippocampus. Ac-SDKP-treated mouse hippocampus showed an increase in β-catenin stability with reduction of glycogen synthase kinase-3β (GSK-3β) activity. Moreover, inhibition of vascular endothelial growth factor (VEGF) signaling blocked Ac-SDKP-facilitated neural proliferation. Subchronic intrahippocampal infusion of Ac-SDKP also increased spatial memory. Taken together, these data demonstrate that Ac-SDKP functions as a regulator of neural proliferation and indicate that Ac-SDKP may be a therapeutic candidate for diseases characterized by neuronal loss.
Collapse
Affiliation(s)
- D H Kim
- Department of Medicinal Biotechnology, College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea; Dong-A Anti-Aging Research Center, Dong-A University, Busan 604-714, Republic of Korea
| | - E-Y Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of Korea.
| | - J H Yi
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK
| | - H E Lee
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea; Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - S J Park
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea; Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Y-K Ryu
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of Korea
| | - H-C Kim
- Biomedical Mouse Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Taejeon 305-806, Republic of Korea
| | - S Lee
- Faculty of Marine Biomedical Science, Cheju National University, Jeju 690-756, Republic of Korea
| | - J H Ryu
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea; Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea.
| |
Collapse
|
36
|
Regional and stage-specific effects of prospectively purified vascular cells on the adult V-SVZ neural stem cell lineage. J Neurosci 2015; 35:4528-39. [PMID: 25788671 DOI: 10.1523/jneurosci.1188-14.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adult neural stem cells reside in specialized niches. In the ventricular-subventricular zone (V-SVZ), quiescent neural stem cells (qNSCs) become activated (aNSCs), and generate transit amplifying cells (TACs), which give rise to neuroblasts that migrate to the olfactory bulb. The vasculature is an important component of the adult neural stem cell niche, but whether vascular cells in neurogenic areas are intrinsically different from those elsewhere in the brain is unknown. Moreover, the contribution of pericytes to the neural stem cell niche has not been defined. Here, we describe a rapid FACS purification strategy to simultaneously isolate primary endothelial cells and pericytes from brain microregions of nontransgenic mice using CD31 and CD13 as surface markers. We compared the effect of purified vascular cells from a neurogenic (V-SVZ) and non-neurogenic brain region (cortex) on the V-SVZ stem cell lineage in vitro. Endothelial and pericyte diffusible signals from both regions differentially promote the proliferation and neuronal differentiation of qNSCs, aNSCs, and TACs. Unexpectedly, diffusible cortical signals had the most potent effects on V-SVZ proliferation and neurogenesis, highlighting the intrinsic capacity of non-neurogenic vasculature to support stem cell behavior. Finally, we identify PlGF-2 as an endothelial-derived mitogen that promotes V-SVZ cell proliferation. This purification strategy provides a platform to define the functional and molecular contribution of vascular cells to stem cell niches and other brain regions under different physiological and pathological states.
Collapse
|
37
|
Geloso MC, Corvino V, Di Maria V, Marchese E, Michetti F. Cellular targets for neuropeptide Y-mediated control of adult neurogenesis. Front Cell Neurosci 2015; 9:85. [PMID: 25852477 PMCID: PMC4360818 DOI: 10.3389/fncel.2015.00085] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/23/2015] [Indexed: 12/14/2022] Open
Abstract
Neuropeptides are emerging as key regulators of stem cell niche activities in health and disease, both inside and outside the central nervous system (CNS). Among them, neuropeptide Y (NPY), one of the most abundant neuropeptides both in the nervous system and in non-neural districts, has become the focus of much attention for its involvement in a wide range of physiological and pathological conditions, including the modulation of different stem cell activities. In particular, a pro-neurogenic role of NPY has been evidenced in the neurogenic niche, where a direct effect on neural progenitors has been demonstrated, while different cellular types, including astrocytes, microglia and endothelial cells, also appear to be responsive to the peptide. The marked modulation of the NPY system during several pathological conditions that affect neurogenesis, including stress, seizures and neurodegeneration, further highlights the relevance of this peptide in the regulation of adult neurogenesis. In view of the considerable interest in understanding the mechanisms controlling neural cell fate, this review aims to summarize and discuss current data on NPY signaling in the different cellular components of the neurogenic niche in order to elucidate the complexity of the mechanisms underlying the modulatory properties of this peptide.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| |
Collapse
|
38
|
Liu F, Ni JJ, Huang JJ, Kou ZW, Sun FY. VEGF overexpression enhances the accumulation of phospho-S292 MeCP2 in reactive astrocytes in the adult rat striatum following cerebral ischemia. Brain Res 2015; 1599:32-43. [DOI: 10.1016/j.brainres.2014.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 12/12/2022]
|
39
|
Michailidou I, de Vries HE, Hol EM, van Strien ME. Activation of endogenous neural stem cells for multiple sclerosis therapy. Front Neurosci 2015; 8:454. [PMID: 25653584 PMCID: PMC4299409 DOI: 10.3389/fnins.2014.00454] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/22/2014] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system, leading to severe neurological deficits. Current MS treatment regimens, consist of immunomodulatory agents aiming to reduce the rate of relapses. However, these agents are usually insufficient to treat chronic neurological disability. A promising perspective for future therapy of MS is the regeneration of lesions with replacement of the damaged oligodendrocytes or neurons. Therapies targeting to the enhancement of endogenous remyelination, aim to promote the activation of either the parenchymal oligodendrocyte progenitor cells or the subventricular zone-derived neural stem cells (NSCs). Less studied but highly potent, is the strategy of neuronal regeneration with endogenous NSCs that although being linked to numerous limitations, is anticipated to ameliorate cognitive disability in MS. Focusing on the forebrain, this review highlights the role of NSCs in the regeneration of MS lesions.
Collapse
Affiliation(s)
- Iliana Michailidou
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Netherlands
| | - Elly M Hol
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands ; Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands ; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Miriam E van Strien
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands ; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
40
|
Hypoxic conditioned medium from rat cerebral cortical cells enhances the proliferation and differentiation of neural stem cells mainly through PI3-K/Akt pathways. PLoS One 2014; 9:e111938. [PMID: 25386685 PMCID: PMC4227679 DOI: 10.1371/journal.pone.0111938] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 10/01/2014] [Indexed: 01/01/2023] Open
Abstract
Purpose To investigate the effects of hypoxic conditioned media from rat cerebral cortical cells on the proliferation and differentiation of neural stem cells (NSCs) in vitro, and to study the roles of PI3-K/Akt and JNK signal transduction pathways in these processes. Methods Cerebral cortical cells from neonatal Sprague–Dawley rat were cultured under hypoxic and normoxic conditions; the supernatant was collected and named ‘hypoxic conditioned medium’ (HCM) and ‘normoxic conditioned medium’ (NCM), respectively. We detected the protein levels (by ELISA) of VEGF and BDNF in the conditioned media and mRNA levels (by RT-PCR) in cerebral cortical cells. The proliferation (number and size of neurospheres) and differentiation (proportion of neurons and astrocytes over total cells) of NSCs was assessed. LY294002 and SP600125, inhibitors of PI3-K/Akt and JNK, respectively, were applied, and the phosphorylation levels of PI3-K, Akt and JNK were measured by western blot. Results The protein levels and mRNA expressions of VEGF and BDNF in 4% HCM and 1% HCM were both higher than that of those in NCM. The efficiency and speed of NSCs proliferation was enhanced in 4% HCM compared with 1% HCM. The highest percentage of neurons and lowest percentage of astrocytes was found in 4% HCM. However, the enhancement of NSCs proliferation and differentiation into neurons accelerated by 4% HCM was inhibited by LY294002 and SP600125, with LY294002 having a stronger inhibitory effect. The increased phosphorylation levels of PI3-K, Akt and JNK in 4% HCM were blocked by LY294002 and SP600125. Conclusions 4%HCM could promote NSCs proliferation and differentiation into high percentage of neurons, these processes may be mainly through PI3-K/Akt pathways.
Collapse
|
41
|
Ren J, Zhou X, Wang J, Zhao J, Zhang P. Poxue Huayu and Tianjing Busui Decoction for cerebral hemorrhage (Upregulation of neurotrophic factor expression): Upregulation of neurotrophic factor expression. Neural Regen Res 2014; 8:2039-49. [PMID: 25206512 PMCID: PMC4146063 DOI: 10.3969/j.issn.1673-5374.2013.22.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 07/08/2013] [Indexed: 11/18/2022] Open
Abstract
This study established a rat model of cerebral hemorrhage by injecting autologous anticoagulated blood. Rat models were intragastrically administered 5, 10, 20 g/kg Poxue Huayu and Tianjing Busui Decoction, supplemented with Hirudo, raw rhubarb, raw Pollen Typhae, gadfly, Fructrs Trichosanthis, Radix Notoginseng, Rhizoma Acori Talarinowii, and glue of tortoise plastron, once a day, for 14 consecutive days. Results demonstrated that brain water content significantly reduced in rats with cerebral hemorrhage, and intracerebral hematoma volume markedly reduced after treatment. Immunohistochemical staining revealed that brain-derived neurotrophic factor, tyrosine kinase B and vascular endothelial growth factor expression noticeably increased around the surrounding hematoma. Reverse transcription-PCR revealed that brain-derived neurotrophic factor and tyrosine kinase B mRNA expression significantly increased around the surrounding hematoma. Neurologic impairment obviously reduced. These results indicated that Poxue Huayu and Tianjing Busui Decoction exert therapeutic effects on cerebral hemorrhage by upregulating the expression of brain-derived neurotrophic factor.
Collapse
Affiliation(s)
- Jixiang Ren
- Department of Encephalopathy, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Xiangyu Zhou
- Grade 2010 Clinical Medicine Major, School of Clinical Medicine, Yanbian University, Yanji 133002, Jilin Province, China
| | - Jian Wang
- Department of Encephalopathy, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Jianjun Zhao
- Department of Encephalopathy, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Pengguo Zhang
- Department of Imaging, School of Second Clinical Medicine, Jilin University, Changchun 130041, Jilin Province, China
| |
Collapse
|
42
|
Vissapragada R, Contreras MA, da Silva CG, Kumar VA, Ochoa A, Vasudevan A, Selim MH, Ferran C, Thomas AJ. Bidirectional crosstalk between periventricular endothelial cells and neural progenitor cells promotes the formation of a neurovascular unit. Brain Res 2014; 1565:8-17. [PMID: 24675025 DOI: 10.1016/j.brainres.2014.03.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 02/17/2014] [Accepted: 03/15/2014] [Indexed: 12/17/2022]
Abstract
Interactions between neural progenitor cells (NPC) and endothelial cells (EC) from adult vascular beds have been well explored previously. However, the factors and signaling mechanisms that regulate neurogenesis and angiogenesis are most prevalent during embryonic development. This study aimed to determine whether embryonic brain endothelial cells from the periventricular region (PVEC) present an advantage over adult brain EC in supporting NPC growth and differentiation. PVEC were isolated from E15 mouse brains, processed, and sorted with immunomagnetic beads using antibodies against CD31/PECAM. On immunofluorescence (IF) staining, nearly all cells were positive for EC markers CD31 and CD144/VE-Cadherin. In proliferation studies, NPC proliferation was highest in transwell co-culture with PVEC, approximately 2.3 fold increase compared to baseline versus 1.4 fold increase when co-cultured with adult brain endothelial cells (ABEC). These results correlated with the PVEC mediated delay in NPC differentiation, evidenced by high expression of progenitor marker Nestin evaluated by IF staining. Upon further characterization of PVEC in an angiogenesis assay measuring cord length, PVEC exhibited a high capacity to form cords in basal conditions compared to ABEC. This was enhanced in the presence of NPC, with both cell types displaying a preferential structural alignment resembling neurovascular networks. PVEC also expressed high Vegfa levels at baseline in comparison to NPC and ABEC. Vegfa levels increased when co-cultured with NPC. We demonstrate that PVEC and NPC co-cultures act synergistically to promote the formation of a neurovascular unit through dynamic and reciprocal communication. Our results suggest that PVEC/NPC could provide promising neuro-regenerative therapies for patients suffering brain injuries.
Collapse
Affiliation(s)
- Ravi Vissapragada
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Mauricio A Contreras
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America; Division of Vascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Cleide G da Silva
- The Division of Vascular and Endovascular Surgery and the Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Vivek A Kumar
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Angelica Ochoa
- College of Engineering, Boston University, Boston, MA, United States of America
| | - Anju Vasudevan
- Department of Psychiatry, Harvard Medical School and Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Belmont, MA, United States of America
| | - Magdy H Selim
- Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Christiane Ferran
- Division of Vascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America; The Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America; Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Ajith J Thomas
- Division of Neurosurgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States of America.
| |
Collapse
|
43
|
Zhou Y, Guan X, Yu M, Wang X, Zhu W, Wang C, Yu M, Wang H. Angiogenic/osteogenic response of BMMSCs on bone-derived scaffold: effect of hypoxia and role of PI3K/Akt-mediated VEGF-VEGFR pathway. Biotechnol J 2014; 9:944-53. [PMID: 24421279 DOI: 10.1002/biot.201300310] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 10/15/2013] [Accepted: 01/09/2014] [Indexed: 11/05/2022]
Abstract
Bone tissue deficiency is a common clinical challenge. Tissue-engineered bone constructs are an effective approach for the repair of orthopedic bone defects. Mimicking the essential components of the in vivo microenvironment is an efficient way to develop functional constructs. In this study, bone marrow-derived mesenchymal stromal cells (BMMSCs) were seeded into bone-derived scaffolds, a material with similar structure to natural bone. This was done under hypoxic conditions, an environment that imitates that experienced by BMMSCs in vivo. Our data indicate that hypoxia (5% O2 ) significantly increases the proliferation of BMMSCs seeded in scaffolds. As reflected by highly expressed osteogenesis- and angiogenesis-associated biomarkers, including vascular endothelial growth factor (VEGF), RUNX2, bone morphogenetic protein-2/4 and osteopontin, hypoxia also significantly increases the osteogenic and angiogenic responses of BMMSCs seeded into bone-derived scaffold composites. PI3K/Akt-mediated regulation of VEGF-activated VEGFR1/2 signaling is important for hypoxia-induced proliferative/osteogenic/angiogenic responses in BMMSC cellular scaffolds. The combination of bone-derived scaffolds and hypoxia is conducive to the differentiation of BMMSCs into functional tissue-engineered scaffold composites.
Collapse
Affiliation(s)
- Yi Zhou
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Wei R, Yang J, Hong TP. Relationship between vascular endothelial cells and pancreatic islet development and stem cell differentiation. Shijie Huaren Xiaohua Zazhi 2013; 21:2493-2499. [DOI: 10.11569/wcjd.v21.i25.2493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As the main components of the pancreatic islet niche, endothelial cells participate in many processes of pancreatic development, including pancreatic cell fate decision, endocrine pancreatic cell differentiation and proliferation, and spatial distribution of the pancreas. On different occasions, endothelial cells play disparate roles by cross-talking with islet cells to influence endocrine pancreatic cell differentiation and islet morphology and function. Cytokines such as hepatocyte growth factor and sphingosine-1-phosphate as well as the extracellular matrixes such as laminin and collagen Ⅳ, which are produced and/or secreted by endothelial cells, play important roles in the regulation of islet development and function. Furthermore, endothelial cells are involved in the balance between self-renewal and differentiation of stem cells. Application of endothelial cells to induce the differentiation of stem cells into functional islet cells may be one of the most promising approaches to cell replacement therapy for diabetes.
Collapse
|
45
|
Interactions between VEGFR and Notch signaling pathways in endothelial and neural cells. Cell Mol Life Sci 2013; 70:1779-92. [PMID: 23479133 DOI: 10.1007/s00018-013-1312-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 02/25/2013] [Accepted: 02/25/2013] [Indexed: 12/25/2022]
Abstract
Notch cell interaction mechanism governs cell fate decisions in many different cell contexts throughout the lifetime of all Metazoan species. It links the fate of one cell to that of its neighbors through cell-to-cell contacts, and binding of Notch receptors expressed on one cell to their membrane bound ligands on an adjacent cell. Environmental cues, such as growth factors and extracellular matrix molecules, superimpose a dynamic regulation on this canonical Notch signaling pathway. In this review, we will focus on Notch signaling in the vertebrate vascular and nervous systems and examine its role in angiogenesis, neurogenesis, and neurovascular interactions. We will also highlight the molecular relationships of the Notch pathway with vascular endothelial growth factors (VEGFs) and their high-affinity tyrosine kinase VEGF receptors, key regulators of both angiogenesis and neurogenesis.
Collapse
|
46
|
Horst OV, Chavez MG, Jheon AH, Desai T, Klein OD. Stem cell and biomaterials research in dental tissue engineering and regeneration. Dent Clin North Am 2012; 56:495-520. [PMID: 22835534 PMCID: PMC3494412 DOI: 10.1016/j.cden.2012.05.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
This review summarizes approaches used in tissue engineering and regenerative medicine, with a focus on dental applications. Dental caries and periodontal disease are the most common diseases resulting in tissue loss. To replace or regenerate new tissues, various sources of stem cells have been identified such as somatic stem cells from teeth and peridontium. Advances in biomaterial sciences including microfabrication, self-assembled biomimetic peptides, and 3-dimensional printing hold great promise for whole-organ or partial tissue regeneration to replace teeth and periodontium.
Collapse
Affiliation(s)
- Orapin V. Horst
- Division of Endodontics, Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, Box 0758, 521 Parnassus Avenue, Clinical Science Building 627, San Francisco, CA 94143-0758, USA
| | - Miquella G. Chavez
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Box 2330, 1700 4th Street, San Francisco, CA 94158-2330, USA
- Department of Orofacial Sciences, University of California, San Francisco, Box 0442, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| | - Andrew H. Jheon
- Department of Orofacial Sciences, University of California, San Francisco, Box 0442, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
| | - Tejal Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Box 2330, 1700 4th Street, San Francisco, CA 94158-2330, USA
- Department of Physiology, University of California, San Francisco, Byers Hall Room 203C, MC 2520, 1700 4th Street, San Francisco, CA 94158-2330, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences, University of California, San Francisco, Box 0442, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
- Department of Pediatrics, University of California, San Francisco, Box 0442, 513 Parnassus Avenue, San Francisco, CA 94143-0442, USA
- Corresponding author. Department of Orofacial Sciences, University of California, San Francisco, Box 0442, 513 Parnassus Avenue, San Francisco, CA 94143-0442.
| |
Collapse
|
47
|
Elias PZ, Spector M. Implantation of a collagen scaffold seeded with adult rat hippocampal progenitors in a rat model of penetrating brain injury. J Neurosci Methods 2012; 209:199-211. [PMID: 22698665 DOI: 10.1016/j.jneumeth.2012.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 04/24/2012] [Accepted: 06/05/2012] [Indexed: 10/28/2022]
Abstract
Penetrating brain injury (PBI) is a complex central nervous system injury in which mechanical damage to brain parenchyma results in hemorrhage, ischemia, broad areas of necrosis, and eventually cavitation. The permanent loss of brain tissue affords the possibility of treatment using a biomaterial scaffold to fill the lesion site and potentially deliver pharmacological or cellular therapeutic agents. The administration of cellular therapy may be of benefit in both mitigating the secondary injury process and promoting regeneration through replacement of certain cell populations. This study investigated the survival and differentiation of adult rat hippocampal neural progenitor cells delivered by a collagen scaffold in a rat model of PBI. The cell-scaffold construct was implanted 1 week after injury and was observed to remain intact with open pores upon analysis 4 weeks later. Implanted neural progenitors were found to have survived within the scaffold, and also to have migrated into the surrounding brain. Differentiated phenotypes included astrocytes, oligodendrocytes, vascular endothelial cells, and possibly macrophages. The demonstrated multipotency of this cell population in vivo in the context of traumatic brain injury has implications for regenerative therapies, but additional stimulation appears necessary to promote neuronal differentiation outside normally neurogenic regions.
Collapse
Affiliation(s)
- Paul Z Elias
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | |
Collapse
|
48
|
Muffley LA, Pan SC, Smith AN, Ga M, Hocking AM, Gibran NS. Differentiation state determines neural effects on microvascular endothelial cells. Exp Cell Res 2012; 318:2085-93. [PMID: 22683922 DOI: 10.1016/j.yexcr.2012.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/17/2012] [Accepted: 06/01/2012] [Indexed: 12/25/2022]
Abstract
Growing evidence indicates that nerves and capillaries interact paracrinely in uninjured skin and cutaneous wounds. Although mature neurons are the predominant neural cell in the skin, neural progenitor cells have also been detected in uninjured adult skin. The aim of this study was to characterize differential paracrine effects of neural progenitor cells and mature sensory neurons on dermal microvascular endothelial cells. Our results suggest that neural progenitor cells and mature sensory neurons have unique secretory profiles and distinct effects on dermal microvascular endothelial cell proliferation, migration, and nitric oxide production. Neural progenitor cells and dorsal root ganglion neurons secrete different proteins related to angiogenesis. Specific to neural progenitor cells were dipeptidyl peptidase-4, IGFBP-2, pentraxin-3, serpin f1, TIMP-1, TIMP-4 and VEGF. In contrast, endostatin, FGF-1, MCP-1 and thrombospondin-2 were specific to dorsal root ganglion neurons. Microvascular endothelial cell proliferation was inhibited by dorsal root ganglion neurons but unaffected by neural progenitor cells. In contrast, microvascular endothelial cell migration in a scratch wound assay was inhibited by neural progenitor cells and unaffected by dorsal root ganglion neurons. In addition, nitric oxide production by microvascular endothelial cells was increased by dorsal root ganglion neurons but unaffected by neural progenitor cells.
Collapse
Affiliation(s)
- Lara A Muffley
- University of Washington, Campus Box 359796, 300 9th Avenue, Seattle, WA 98104, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Vascular endothelial growth factor A (VEGF-A) is best known for its essential roles in blood vessel growth. However, evidence has emerged that VEGF-A also promotes a wide range of neuronal functions, both in vitro and in vivo, including neurogenesis, neuronal migration, neuronal survival and axon guidance. Recent studies have employed mouse models to distinguish the direct effects of VEGF on neurons from its indirect, vessel-mediated effects. Ultimately, refining our knowledge of VEGF signalling pathways in neurons should help us to understand how the current use of therapeutics targeting the VEGF pathway in cancer and eye disease might be expanded to promote neuronal health and nerve repair.
Collapse
Affiliation(s)
- Francesca Mackenzie
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, UK
| | | |
Collapse
|
50
|
Kim H, Cooke MJ, Shoichet MS. Creating permissive microenvironments for stem cell transplantation into the central nervous system. Trends Biotechnol 2011; 30:55-63. [PMID: 21831464 DOI: 10.1016/j.tibtech.2011.07.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 10/17/2022]
Abstract
Traumatic injury to the central nervous system (CNS) is highly debilitating, with the clinical need for regenerative therapies apparent. Neural stem/progenitor cells (NSPCs) are promising because they can repopulate lost or damaged cells and tissues. However, the adult CNS does not provide an optimal milieu for exogenous NSPCs to survive, engraft, differentiate, and integrate with host tissues. This review provides an overview of tissue engineering strategies to improve stem cell therapies by providing a defined microenvironment during transplantation. The use of biomaterials for physical support, growth factor delivery, and cellular co-transplantation are discussed. Providing the proper environment for stem cell survival and host tissue integration is crucial in realizing the full potential of these cells in CNS repair strategies.
Collapse
Affiliation(s)
- Howard Kim
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|