1
|
Aksan B, Mauceri D. Beyond vessels: unraveling the impact of VEGFs on neuronal functions and structure. J Biomed Sci 2025; 32:33. [PMID: 40050849 PMCID: PMC11884128 DOI: 10.1186/s12929-025-01128-8] [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: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025] Open
Abstract
Neurons rely on the bloodstream for essential nutrients and oxygen, which is facilitated by an intricate coupling of the neuronal and vascular systems. Central to this neurovascular interaction is the vascular endothelial growth factor (VEGF) family, a group of secreted growth factors traditionally known for their roles in promoting endothelial cell proliferation, migration, and survival in the cardiovascular and lymphatic systems. However, emerging evidence shows that VEGFs also play indispensable roles in the nervous system, extending beyond their canonical angiogenic and lymphangiogenic functions. Over the past two decades, VEGFs have been found to exert direct effects on neurons, influencing key aspects of neuronal function independently of their actions on vascular cells. In particular, it has become increasingly evident that VEGFs also play crucial functions in the development, regulation, and maintenance of neuronal morphology. Understanding the roles of VEGFs in neuronal development is of high scientific and clinical interest because of the significance of precise neuronal morphology for neural connectivity and network function, as well as the association of morphological abnormalities with neurological and neurodegenerative disorders. This review begins with an overview of the VEGF family members, their structural characteristics, receptors, and established roles in vasculature. However, it then highlights and focuses on the exciting variety of neuronal functions of VEGFs, especially their crucial role in the development, regulation, and maintenance of neuronal morphology.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Institute of Anatomy and Cell Biology, Dept. Molecular and Cellular Neuroscience, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
2
|
González-Hernández S, Mukouyama YS. Lymphatic vasculature in the central nervous system. Front Cell Dev Biol 2023; 11:1150775. [PMID: 37091974 PMCID: PMC10119411 DOI: 10.3389/fcell.2023.1150775] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The central nervous system (CNS) is considered as an immune privilege organ, based on experiments in the mid 20th century showing that the brain fails to mount an efficient immune response against an allogeneic graft. This suggests that in addition to the presence of the blood-brain barrier (BBB), the apparent absence of classical lymphatic vasculature in the CNS parenchyma limits the capacity for an immune response. Although this view is partially overturned by the recent discovery of the lymphatic-like hybrid vessels in the Schlemm's canal in the eye and the lymphatic vasculature in the outmost layer of the meninges, the existence of lymphatic vessels in the CNS parenchyma has not been reported. Two potential mechanisms by which lymphatic vasculature may arise in the organs are: 1) sprouting and invasion of lymphatic vessels from the surrounding tissues into the parenchyma and 2) differentiation of blood endothelial cells into lymphatic endothelial cells in the parenchyma. Considering these mechanisms, we here discuss what causes the dearth of lymphatic vessels specifically in the CNS parenchyma.
Collapse
|
3
|
Shen K, Duan Q, Duan W, Xu S, An N, Ke Y, Wang L, Liu S, Yang H, Zhang C. Vascular endothelial growth factor-C modulates cortical NMDA receptor activity in cortical lesions of young patients and rat model with focal cortical dysplasia. Brain Pathol 2022; 32:e13065. [PMID: 35259773 PMCID: PMC9425019 DOI: 10.1111/bpa.13065] [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: 01/22/2022] [Revised: 02/20/2022] [Accepted: 02/25/2022] [Indexed: 12/01/2022] Open
Abstract
Emergence of dysmorphic neurons is the primary pathology in focal cortical dysplasia (FCD) associated pediatric intractable epilepsy; however, the etiologies related to the development and function of dysmorphic neurons are not fully understood. Our previous studies revealed that the expression of vascular endothelial growth factor-C (VEGF-C) and corresponding receptors VEGFR-2, VEGFR-3 was increased in the epileptic lesions of patients with tuberous sclerosis complex or mesial temporal lobe epilepsy. Here, we showed that the expression of VEGF-C, VEGFR-2, and VEGFR-3 was increased at both mRNA and protein levels in patients with cortical lesions of type I, IIa, and IIb FCD. The immunoreactivity of VEGF-C, VEGFR-2 and VEGFR-3 was located in the micro-columnar neurons in FCD type I lesions, dysplastic neurons (DNs) in FCD type IIa lesions, balloon cells (BCs) and astrocytes in FCD type IIb lesions. Additionally, the amplitude of evoked-EPSCs (eEPSC) mediated by NMDA receptor, the ratio of NMDA receptor- and AMPA receptor-mediated eEPSC were increased in the dysmorphic neurons of FCD rats established by prenatal X-ray radiation. Furthermore, NMDA receptor mediated current in dysmorphic neurons was further potentiated by exogenous administration of VEGF-C, however, could be antagonized by ki8751, the blocker of VEGFR-2. These results suggest that VEGF-C system participate in the pathogenesis of cortical lesions in patients with FCD in association with modulating NMDA receptor-mediated currents.
Collapse
Affiliation(s)
- Kai‐Feng Shen
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Qing‐Tian Duan
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Wei Duan
- Department of NeurologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Sen‐Lin Xu
- Institute of PathologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Ning An
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Yan‐Yan Ke
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Li‐Ting Wang
- Biomedical Analysis CenterArmy Medical UniversityChongqingChina
| | - Shi‐Yong Liu
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Hui Yang
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
| | - Chun‐Qing Zhang
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
| |
Collapse
|
4
|
Brinks J, van Dijk EHC, Klaassen I, Schlingemann RO, Kielbasa SM, Emri E, Quax PHA, Bergen AA, Meijer OC, Boon CJF. Exploring the choroidal vascular labyrinth and its molecular and structural roles in health and disease. Prog Retin Eye Res 2021; 87:100994. [PMID: 34280556 DOI: 10.1016/j.preteyeres.2021.100994] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022]
Abstract
The choroid is a key player in maintaining ocular homeostasis and plays a role in a variety of chorioretinal diseases, many of which are poorly understood. Recent advances in the field of single-cell RNA sequencing have yielded valuable insights into the properties of choroidal endothelial cells (CECs). Here, we review the role of the choroid in various physiological and pathophysiological mechanisms, focusing on the role of CECs. We also discuss new insights regarding the phenotypic properties of CECs, CEC subpopulations, and the value of measuring transcriptomics in primary CEC cultures derived from post-mortem eyes. In addition, we discuss key phenotypic, structural, and functional differences that distinguish CECs from other endothelial cells such as retinal vascular endothelial cells. Understanding the specific clinical and molecular properties of the choroid will shed new light on the pathogenesis of the broad clinical range of chorioretinal diseases such as age-related macular degeneration, central serous chorioretinopathy and other diseases within the pachychoroid spectrum, uveitis, and diabetic choroidopathy. Although our knowledge is still relatively limited with respect to the clinical features and molecular pathways that underlie these chorioretinal diseases, we summarise new approaches and discuss future directions for gaining new insights into these sight-threatening diseases and highlight new therapeutic strategies such as pluripotent stem cell‒based technologies and gene therapy.
Collapse
Affiliation(s)
- J Brinks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - E H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - I Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - R O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - S M Kielbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - E Emri
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - P H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - A A Bergen
- Department of Clinical Genetics, Section of Ophthalmogenetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - O C Meijer
- Department of Medicine, Division of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands
| | - C J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Glaesel K, May C, Marcus K, Matschke V, Theiss C, Theis V. miR-129-5p and miR-130a-3p Regulate VEGFR-2 Expression in Sensory and Motor Neurons during Development. Int J Mol Sci 2020; 21:ijms21113839. [PMID: 32481647 PMCID: PMC7312753 DOI: 10.3390/ijms21113839] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 01/23/2023] Open
Abstract
The wide-ranging influence of vascular endothelial growth factor (VEGF) within the central (CNS) and peripheral nervous system (PNS), for example through effects on axonal growth or neuronal cell survival, is mainly mediated by VEGF receptor 2 (VEGFR-2). However, the regulation of VEGFR-2 expression during development is not yet well understood. As microRNAs are considered to be key players during neuronal maturation and regenerative processes, we identified the two microRNAs (miRNAs)-miR-129-5p and miR-130a-3p-that may have an impact on VEGFR-2 expression in young and mature sensory and lower motor neurons. The expression level of VEGFR-2 was analyzed by using in situ hybridization, RT-qPCR, Western blot, and immunohistochemistry in developing rats. microRNAs were validated within the spinal cord and dorsal root ganglia. To unveil the molecular impact of our candidate microRNAs, dissociated cell cultures of sensory and lower motor neurons were transfected with mimics and inhibitors. We depicted age-dependent VEGFR-2 expression in sensory and lower motor neurons. In detail, in lower motor neurons, VEGFR-2 expression was significantly reduced during maturation, in conjunction with an increased level of miR-129-5p. In sensory dorsal root ganglia, VEGFR-2 expression increased during maturation and was accompanied by an overexpression of miR-130a-3p. In a second step, the functional significance of these microRNAs with respect to VEGFR-2 expression was proven. Whereas miR-129-5p seems to decrease VEGFR-2 expression in a direct manner in the CNS, miR-130a-3p might indirectly control VEGFR-2 expression in the PNS. A detailed understanding of genetic VEGFR-2 expression control might promote new strategies for the treatment of severe neurological diseases like ischemia or peripheral nerve injury.
Collapse
Affiliation(s)
- Kevin Glaesel
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, 44780 Bochum, Germany; (K.G.); (V.M.); (V.T.)
| | - Caroline May
- Medical Proteom-Center, Ruhr University Bochum, 44780 Bochum, NRW, Germany; (C.M.); (K.M.)
| | - Katrin Marcus
- Medical Proteom-Center, Ruhr University Bochum, 44780 Bochum, NRW, Germany; (C.M.); (K.M.)
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, 44780 Bochum, Germany; (K.G.); (V.M.); (V.T.)
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, 44780 Bochum, Germany; (K.G.); (V.M.); (V.T.)
- Correspondence: ; Tel.: +49-234-32-25018
| | - Verena Theis
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, 44780 Bochum, Germany; (K.G.); (V.M.); (V.T.)
| |
Collapse
|
6
|
Anosmin-1 activates vascular endothelial growth factor receptor and its related signaling pathway for olfactory bulb angiogenesis. Sci Rep 2020; 10:188. [PMID: 31932617 PMCID: PMC6957483 DOI: 10.1038/s41598-019-57040-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Anosmin-1 is a secreted glycoprotein encoded by the ANOS1 gene, and its loss of function causes Kallmann syndrome (KS), which is characterized by anosmia and hypogonadism due to olfactory bulb (OB) dysfunction. However, the physiological function of anosmin-1 remains to be elucidated. In KS, disordered angiogenesis is observed in OB, resulting in its hypoplasia. In this study, we examined the involvement of anosmin-1 in angiogenic processes. Anosmin-1 was detected on the vessel-like structure in OB of chick embryos, and promoted the outgrowth of vascular sprouts as shown by assays of OB tissue culture. Cell migration, proliferation, and tube formation of endothelial cells were induced by treatment with anosmin-1 as well as vascular endothelial growth factor-A (VEGF-A), and further enhanced by treatment with both of them. We newly identified that anosmin-1 activated VEGF receptor-2 (VEGFR2) by binding directly to it, and its downstream signaling molecules, phospholipase Cγ1 (PLCγ1) and protein kinase C (PKC). These results suggest that anosmin-1 plays a key role in the angiogenesis of developing OB through the VEGFR2–PLCγ1–PKC axis by enhancing the VEGF function.
Collapse
|
7
|
Liang Q, Zhang L, Wood RW, Ji RC, Boyce BF, Schwarz EM, Wang Y, Xing L. Avian Reticuloendotheliosis Viral Oncogene Related B Regulates Lymphatic Endothelial Cells during Vessel Maturation and Is Required for Lymphatic Vessel Function in Adult Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2516-2530. [PMID: 31539516 DOI: 10.1016/j.ajpath.2019.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/05/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
Abstract
NF-κB signals through canonical transcription factor p65 (RelA)/p50 and noncanonical avian reticuloendotheliosis viral oncogene related B (RelB)/p52 pathways. The RelA/p50 is involved in basal and inflammatory lymphangiogenesis. However, the role of RelB/p52 in lymphatic vessel biology is unknown. Herein, we investigated changes in lymphatic vessels (LVs) in mice deficient in noncanonical NF-κB signaling and the function of RelB in lymphatic endothelial cells (LECs). LVs were examined in Relb-/-, p52-/-, or control mice, and the gene expression profiles in LECs with RelB knockdown. Relb-/-, but not p52-/-, mice exhibited multiple LV abnormalities. They include the following: i) increased capillary vessel diameter, ii) reduced smooth muscle cell (SMC) coverage of mature vessels, iii) leakage, and iv) loss of active and passive lymphatic flow. Relb-/- mature LVs had thinner vessel walls, more apoptotic LECs and SMCs, and fewer LEC junctions. RelB knockdown LECs had decreased growth, survival, and adhesion, and dysregulated signaling pathways involving these cellular events. These results suggest that Relb-/- mice have abnormal LVs, mainly in mature vessels with reduced SMC coverage, leakage, and loss of contractions. RelB knockdown in LECs leads to reduced growth, survival, and adhesion. RelB plays a vital role in LEC-mediated LV maturation and function.
Collapse
Affiliation(s)
- Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Li Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Ronald W Wood
- Department of Obstetrics and Gynecology, Urology, and Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York
| | | | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
8
|
Han X, Zhao X, Lan X, Li Q, Gao Y, Liu X, Wan J, Yang Z, Chen X, Zang W, Guo AM, Falck JR, Koehler RC, Wang J. 20-HETE synthesis inhibition promotes cerebral protection after intracerebral hemorrhage without inhibiting angiogenesis. J Cereb Blood Flow Metab 2019; 39:1531-1543. [PMID: 29485354 PMCID: PMC6681539 DOI: 10.1177/0271678x18762645] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
20-HETE, an arachidonic acid metabolite synthesized by cytochrome P450 4A, plays an important role in acute brain damage from ischemic stroke or subarachnoid hemorrhage. We tested the hypothesis that 20-HETE inhibition has a protective effect after intracerebral hemorrhage (ICH) and then investigated its effect on angiogenesis. We exposed hippocampal slice cultures to hemoglobin and induced ICH in mouse brains by intrastriatal collagenase injection to investigate the protective effect of 20-HETE synthesis inhibitor N-hydroxy-N'-(4-n-butyl-2-methylphenyl)-formamidine (HET0016). Hemoglobin-induced neuronal death was assessed by propidium iodide after 18 h in vitro. Lesion volume, neurologic deficits, cell death, reactive oxygen species (ROS), neuroinflammation, and angiogenesis were evaluated at different time points after ICH. In cultured mouse hippocampal slices, HET0016 attenuated hemoglobin-induced neuronal death and decreased levels of proinflammatory cytokines and ROS. In vivo, HET0016 reduced brain lesion volume and neurologic deficits, and decreased neuronal death, ROS production, gelatinolytic activity, and the inflammatory response at three days after ICH. However, HET0016 did not inhibit angiogenesis, as levels of CD31, VEGF, and VEGFR2 were unchanged on day 28. We conclude that 20-HETE is involved in ICH-induced brain damage. Inhibition of 20-HETE synthesis may provide a viable means to mitigate ICH injury without inhibition of angiogenesis.
Collapse
Affiliation(s)
- Xiaoning Han
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xiaochun Zhao
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xi Lan
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Qian Li
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yufeng Gao
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xi Liu
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jieru Wan
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zengjin Yang
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xuemei Chen
- 2 Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Weidong Zang
- 2 Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Austin M Guo
- 3 Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | - John R Falck
- 4 Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Raymond C Koehler
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jian Wang
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Cho KO, Kim JY, Jeong KH, Lee MY, Kim SY. Increased expression of vascular endothelial growth factor-C and vascular endothelial growth factor receptor-3 after pilocarpine-induced status epilepticus in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:281-289. [PMID: 31297012 PMCID: PMC6609264 DOI: 10.4196/kjpp.2019.23.4.281] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
Abstract
Vascular endothelial growth factor (VEGF)-C and its receptor, vascular endothelial growth factor receptor (VEGFR)-3, are responsible for lymphangiogenesis in both embryos and adults. In epilepsy, the expression of VEGF-C and VEGFR-3 was significantly upregulated in the human brains affected with temporal lobe epilepsy. Moreover, pharmacologic inhibition of VEGF receptors after acute seizures could suppress the generation of spontaneous recurrent seizures, suggesting a critical role of VEGF-related signaling in epilepsy. Therefore, in the present study, the spatiotemporal expression of VEGF-C and VEGFR-3 against pilocarpine-induced status epilepticus (SE) was investigated in C57BL/6N mice using immunohistochemistry. At 1 day after SE, hippocampal astrocytes and microglia were activated. Pyramidal neuronal death was observed at 4 days after SE. In the subpyramidal zone, VEGF-C expression gradually increased and peaked at 7 days after SE, while VEGFR-3 was significantly upregulated at 4 days after SE and began to decrease at 7 days after SE. Most VEGF-C/VEGFR-3-expressing cells were pyramidal neurons, but VEGF-C was also observed in some astrocytes in sham-manipulated animals. However, at 4 days and 7 days after SE, both VEGFR-3 and VEGF-C immunoreactivities were observed mainly in astrocytes and in some microglia of the stratum radiatum and lacunosum-moleculare of the hippocampus, respectively. These data indicate that VEGF-C and VEGFR-3 can be upregulated in hippocampal astrocytes and microglia after pilocarpine-induced SE, providing basic information about VEGF-C and VEGFR-3 expression patterns following acute seizures.
Collapse
Affiliation(s)
- Kyung-Ok Cho
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Institute of Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Joo Youn Kim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Kyoung Hoon Jeong
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Mun-Yong Lee
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seong Yun Kim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
10
|
Ghazale H, Ripoll C, Leventoux N, Jacob L, Azar S, Mamaeva D, Glasson Y, Calvo CF, Thomas JL, Meneceur S, Lallemand Y, Rigau V, Perrin FE, Noristani HN, Rocamonde B, Huillard E, Bauchet L, Hugnot JP. RNA Profiling of the Human and Mouse Spinal Cord Stem Cell Niches Reveals an Embryonic-like Regionalization with MSX1 + Roof-Plate-Derived Cells. Stem Cell Reports 2019; 12:1159-1177. [PMID: 31031189 PMCID: PMC6524006 DOI: 10.1016/j.stemcr.2019.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 11/30/2022] Open
Abstract
Anamniotes, rodents, and young humans maintain neural stem cells in the ependymal zone (EZ) around the central canal of the spinal cord, representing a possible endogenous source for repair in mammalian lesions. Cell diversity and genes specific for this region are ill defined. A cellular and molecular resource is provided here for the mouse and human EZ based on RNA profiling, immunostaining, and fluorescent transgenic mice. This uncovered the conserved expression of 1,200 genes including 120 transcription factors. Unexpectedly the EZ maintains an embryonic-like dorsal-ventral pattern of expression of spinal cord developmental transcription factors (ARX, FOXA2, MSX1, and PAX6). In mice, dorsal and ventral EZ cells express Vegfr3 and are derived from the embryonic roof and floor plates. The dorsal EZ expresses a high level of Bmp6 and Gdf10 genes and harbors a subpopulation of radial quiescent cells expressing MSX1 and ID4 transcription factors. A molecular resource for the human and mouse spinal cord ependymal zone Identification of 120 transcription factors in the human and mouse ependymal zone Embryonic-like organization of the adult spinal cord ependymal zone Dorsal ependymal cells expressing Msx1 are derived from the embryonic roof plate
Collapse
Affiliation(s)
- Hussein Ghazale
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Chantal Ripoll
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Nicolas Leventoux
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Laurent Jacob
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013, Paris, France
| | - Safa Azar
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Daria Mamaeva
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Yael Glasson
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Charles-Felix Calvo
- Neuroglial Interactions in Cerebral Physiopathology. CIRB, CNRS UMR 7241/INSERM U1050 Collège de France 11, Place Marcelin Berthelot, 75005 Paris, France
| | - Jean-Leon Thomas
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013, Paris, France; Department of Neurology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Sarah Meneceur
- Institut Pasteur, Department of Developmental and Stem Cell Biology, UMR3738 CNRS, Paris 75015, France
| | - Yvan Lallemand
- Institut Pasteur, Department of Developmental and Stem Cell Biology, UMR3738 CNRS, Paris 75015, France
| | - Valérie Rigau
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France; CHU of Montpellier, Hopital Gui de Chaulliac, Pathology Department, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Florence E Perrin
- University of Montpellier, Faculté des Sciences, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| | - Harun N Noristani
- University of Montpellier, Faculté des Sciences, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| | - Brenda Rocamonde
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013, Paris, France
| | - Emmanuelle Huillard
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013, Paris, France
| | - Luc Bauchet
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France; CHU of Montpellier, Hopital Gui de Chaulliac, Neurosurgery Department, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Jean-Philippe Hugnot
- INSERM U1051, INM, Hopital Saint Eloi, 80 Avenue Augustin Fliche, 34091 Montpellier, France; University of Montpellier, Faculté des Sciences, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France.
| |
Collapse
|
11
|
Tan KZ, Cunningham AM, Joshi A, Oei JL, Ward MC. Expression of kappa opioid receptors in developing rat brain - Implications for perinatal buprenorphine exposure. Reprod Toxicol 2018; 78:81-89. [PMID: 29635048 DOI: 10.1016/j.reprotox.2018.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 12/19/2022]
Abstract
Buprenorphine, a mu opioid receptor partial agonist and kappa opioid receptor (KOR) antagonist, is an emerging therapeutic agent for maternal opioid dependence in pregnancy and neonatal abstinence syndrome. However, the endogenous opioid system plays a critical role in modulating neurodevelopment and perinatal buprenorphine exposure may detrimentally influence this. To identify aspects of neurodevelopment vulnerable to perinatal buprenorphine exposure, we defined KOR protein expression and its cellular associations in normal rat brain from embryonic day 16 to postnatal day 23 with double-labelling immunohistochemistry. KOR was expressed on neural stem and progenitor cells (NSPCs), choroid plexus epithelium, subpopulations of cortical neurones and oligodendrocytes, and NSPCs and subpopulations of neurones in postnatal hippocampus. These distinct patterns of KOR expression suggest several pathways vulnerable to perinatal buprenorphine exposure, including proliferation, neurogenesis and neurotransmission. We thus suggest the cautious use of buprenorphine in both mothers and infants until its impact on neurodevelopment is better defined.
Collapse
Affiliation(s)
- Kathleen Z Tan
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia
| | - Anne M Cunningham
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; Westfield Research Laboratories, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia.
| | - Anjali Joshi
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; Westfield Research Laboratories, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia
| | - Ju Lee Oei
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; The Royal Hospital for Women, Barker Street, Randwick, NSW 2031, Australia
| | - Meredith C Ward
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; The Royal Hospital for Women, Barker Street, Randwick, NSW 2031, Australia; Westfield Research Laboratories, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia.
| |
Collapse
|
12
|
Harder DR, Rarick KR, Gebremedhin D, Cohen SS. Regulation of Cerebral Blood Flow: Response to Cytochrome P450 Lipid Metabolites. Compr Physiol 2018; 8:801-821. [PMID: 29687906 DOI: 10.1002/cphy.c170025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There have been numerous reviews related to the cerebral circulation. Most of these reviews are similar in many ways. In the present review, we thought it important to provide an overview of function with specific attention to details of cerebral arterial control related to brain homeostasis, maintenance of neuronal energy demands, and a unique perspective related to the role of astrocytes. A coming review in this series will discuss cerebral vascular development and unique properties of the neonatal circulation and developing brain, thus, many aspects of development are missing here. Similarly, a review of the response of the brain and cerebral circulation to heat stress has recently appeared in this series (8). By trying to make this review unique, some obvious topics were not discussed in lieu of others, which are from recent and provocative research such as endothelium-derived hyperpolarizing factor, circadian regulation of proteins effecting cerebral blood flow, and unique properties of the neurovascular unit. © 2018 American Physiological Society. Compr Physiol 8:801-821, 2018.
Collapse
Affiliation(s)
- David R Harder
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin, USA
| | - Kevin R Rarick
- Department of Pediatrics, Division of Critical Care, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Debebe Gebremedhin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Susan S Cohen
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
13
|
Durán-Carabali LE, Arcego DM, Odorcyk FK, Reichert L, Cordeiro JL, Sanches EF, Freitas LD, Dalmaz C, Pagnussat A, Netto CA. Prenatal and Early Postnatal Environmental Enrichment Reduce Acute Cell Death and Prevent Neurodevelopment and Memory Impairments in Rats Submitted to Neonatal Hypoxia Ischemia. Mol Neurobiol 2017; 55:3627-3641. [DOI: 10.1007/s12035-017-0604-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
|
14
|
Pike JA, Styles IB, Rappoport JZ, Heath JK. Quantifying receptor trafficking and colocalization with confocal microscopy. Methods 2017; 115:42-54. [PMID: 28131869 DOI: 10.1016/j.ymeth.2017.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 01/08/2023] Open
Abstract
Confocal microscopy is a powerful tool for the study of cellular receptor trafficking and endocytosis. Unbiased and robust image analysis workflows are required for the identification, and study, of aberrant trafficking. After a brief review of related strategies, identifying both good and bad practice, custom workflows for the analysis of live cell 3D time-lapse data are presented. Strategies for data pre-processing, including denoising and background subtraction are considered. We use a 3D level set protocol to accurately segment cells using only the signal from fluorescently labelled receptor. A protocol for the quantification of changes to subcellular receptor distribution over time is then presented. As an example, ligand stimulated trafficking of epidermal growth factor receptor (EGFR) is shown to be significantly reduced in both AG1478 and Dynasore treated cells. Protocols for the quantitative analysis of colocalization between receptor and endosomes are also introduced, including strategies for signal isolation and statistical testing. By calculating the Manders and Pearson coefficients, both co-occurrence and correlation can be assessed. A statistically significant decrease in the level of ligand induced co-occurrence between EGFR and rab5 positive endosomes is demonstrated for both the AG1478 and Dynasore treated cells relative to a control. Finally, a strategy for the visualisation of co-occurrence is presented, which provides an unbiased alternative to colour overlays.
Collapse
Affiliation(s)
- Jeremy A Pike
- PSIBS Doctoral Training Centre, School of Chemistry, University of Birmingham, Birmingham B15 2TT, UK; School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK.
| | - Iain B Styles
- School of Computer Science, University of Birmingham, Birmingham B15 2TT, UK
| | - Joshua Z Rappoport
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; Center for Advanced Microscopy and the Nikon Imaging Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John K Heath
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
15
|
Herrfurth L, Theis V, Matschke V, May C, Marcus K, Theiss C. Morphological Plasticity of Emerging Purkinje Cells in Response to Exogenous VEGF. Front Mol Neurosci 2017; 10:2. [PMID: 28194096 PMCID: PMC5276996 DOI: 10.3389/fnmol.2017.00002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is well known as the growth factor with wide-ranging functions even in the central nervous system (CNS). Presently, most attention is given to the investigation of its role in neuronal protection, growth and maturation processes, whereby most effects are mediated through VEGF receptor 2 (VEGFR-2). The purpose of our current study is to provide new insights into the impact of VEGF on immature and mature Purkinje cells (PCs) in accordance with maturity and related receptor expression. Therefore, to expand our knowledge of VEGF effects in PCs development and associated VEGFR-2 expression, we used cultivated organotypic cerebellar slice cultures in immunohistochemical or microinjection studies, followed by confocal laser scanning microscopy (CLSM) and morphometric analysis. Additionally, we incorporated in our study the method of laser microdissection, followed by quantitative polymerase chain reaction (qPCR). For the first time we could show the age-dependent VEGF sensitivity of PCs with the largest promoting effects being on dendritic length and cell soma size in neonatal and juvenile stages. Once mature, PCs were no longer susceptible to VEGF stimulation. Analysis of VEGFR-2 expression revealed its presence in PCs throughout development, which underlined its mediating functions in neuronal cells.
Collapse
Affiliation(s)
- Leonard Herrfurth
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| | - Verena Theis
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| | - Veronika Matschke
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| | - Caroline May
- Abteilung für Medizinische Proteomik/Bioanalytik, Medizinisches Proteom-Center, Ruhr-University Bochum Bochum, Germany
| | - Katrin Marcus
- Abteilung für Medizinische Proteomik/Bioanalytik, Medizinisches Proteom-Center, Ruhr-University Bochum Bochum, Germany
| | - Carsten Theiss
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| |
Collapse
|
16
|
Kobeissy FH, Hansen K, Neumann M, Fu S, Jin K, Liu J. Deciphering the Role of Emx1 in Neurogenesis: A Neuroproteomics Approach. Front Mol Neurosci 2016; 9:98. [PMID: 27799894 PMCID: PMC5065984 DOI: 10.3389/fnmol.2016.00098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022] Open
Abstract
Emx1 has long been implicated in embryonic brain development. Previously we found that mice null of Emx1 gene had smaller dentate gyri and reduced neurogenesis, although the molecular mechanisms underlying this defect was not well understood. To decipher the role of Emx1 gene in neural regeneration and the timing of its involvement, we determine the frequency of neural stem cells (NSCs) in embryonic and adult forebrains of Emx1 wild type (WT) and knock out (KO) mice in the neurosphere assay. Emx1 gene deletion reduced the frequency and self-renewal capacity of NSCs of the embryonic brain but did not affect neuronal or glial differentiation. Emx1 KO NSCs also exhibited a reduced migratory capacity in response to serum or vascular endothelial growth factor (VEGF) in the Boyden chamber migration assay compared to their WT counterparts. A thorough comparison between NSC lysates from Emx1 WT and KO mice utilizing 2D-PAGE coupled with tandem mass spectrometry revealed 38 proteins differentially expressed between genotypes, including the F-actin depolymerization factor Cofilin. A global systems biology and cluster analysis identified several potential mechanisms and cellular pathways implicated in altered neurogenesis, all involving Cofilin1. Protein interaction network maps with functional enrichment analysis further indicated that the differentially expressed proteins participated in neural-specific functions including brain development, axonal guidance, synaptic transmission, neurogenesis, and hippocampal morphology, with VEGF as the upstream regulator intertwined with Cofilin1 and Emx1. Functional validation analysis indicated that apart from the overall reduced level of phosphorylated Cofilin1 (p-Cofilin1) in the Emx1 KO NSCs compared to WT NSCs as demonstrated in the western blot analysis, VEGF was able to induce more Cofilin1 phosphorylation and FLK expression only in the latter. Our results suggest that a defect in Cofilin1 phosphorylation induced by VEGF or other growth factors might contribute to the reduced neurogenesis in the Emx1 null mice during brain development.
Collapse
Affiliation(s)
- Firas H Kobeissy
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research, University of Florida Gainesville, FL, USA
| | - Katharina Hansen
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| | - Melanie Neumann
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| | - Shuping Fu
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA; Key Laboratory of Acupuncture and Medicine Research of Minister of Education, Nanjing University of Chinese MedicineNanjing, China
| | - Kulin Jin
- Pharmacology & Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Jialing Liu
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| |
Collapse
|
17
|
Mishra S, Choe Y, Pleasure SJ, Siegenthaler JA. Cerebrovascular defects in Foxc1 mutants correlate with aberrant WNT and VEGF-A pathways downstream of retinoic acid from the meninges. Dev Biol 2016; 420:148-165. [PMID: 27671872 DOI: 10.1016/j.ydbio.2016.09.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 12/28/2022]
Abstract
Growth and maturation of the cerebrovasculature is a vital event in neocortical development however mechanisms that control cerebrovascular development remain poorly understood. Mutations in or deletions that include the FOXC1 gene are associated with congenital cerebrovascular anomalies and increased stroke risk in patients. Foxc1 mutant mice display severe cerebrovascular hemorrhage at late gestational ages. While these data demonstrate Foxc1 is required for cerebrovascular development, its broad expression in the brain vasculature combined with Foxc1 mutant's complex developmental defects have made it difficult to pinpoint its function(s). Using global and conditional Foxc1 mutants, we find 1) significant cerebrovascular growth defects precede cerebral hemorrhage and 2) expression of Foxc1 in neural crest-derived meninges and brain pericytes, though not endothelial cells, is required for normal cerebrovascular development. We provide evidence that reduced levels of meninges-derived retinoic acid (RA), caused by defects in meninges formation in Foxc1 mutants, is a major contributing factor to the cerebrovascular growth defects in Foxc1 mutants. We provide data that suggests that meninges-derived RA ensures adequate growth of the neocortical vasculature via regulating expression of WNT pathway proteins and neural progenitor derived-VEGF-A. Our findings offer the first evidence for a role of the meninges in brain vascular development and provide new insight into potential causes of cerebrovascular defects in patients with FOXC1 mutations.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, School of Medicine Aurora, CO 80045, USA
| | - Youngshik Choe
- Department of Neurology, Programs in Neuroscience and Developmental Biology, Institute for Regenerative Medicine, UC San Francisco, San Francisco, CA 94158, USA
| | - Samuel J Pleasure
- Department of Neurology, Programs in Neuroscience and Developmental Biology, Institute for Regenerative Medicine, UC San Francisco, San Francisco, CA 94158, USA
| | - Julie A Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, School of Medicine Aurora, CO 80045, USA.
| |
Collapse
|
18
|
Dumpich M, Theiss C. VEGF in the nervous system: an important target for research in neurodevelopmental and regenerative medicine. Neural Regen Res 2016; 10:1725-6. [PMID: 26807091 PMCID: PMC4705768 DOI: 10.4103/1673-5374.170287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Matthias Dumpich
- Faculty of Medicine, Institute of Anatomy, Department of Cytology Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Faculty of Medicine, Institute of Anatomy, Department of Cytology Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
19
|
Sun FJ, Wei YJ, Li S, Guo W, Chen X, Liu SY, He JJ, Yin Q, Yang H, Zhang CQ. Elevated Expression of VEGF-C and Its Receptors, VEGFR-2 and VEGFR-3, in Patients with Mesial Temporal Lobe Epilepsy. J Mol Neurosci 2016; 59:241-50. [DOI: 10.1007/s12031-016-0714-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/11/2016] [Indexed: 12/20/2022]
|