1
|
Quesada CLV, Rao SB, Torp R, Niehusmann P, Eide PK. Lack of inflammation or immune response in cyst tissue of patients with symptomatic non-hydrocephalic pineal cysts. J Neurol Sci 2024; 462:123111. [PMID: 38943895 DOI: 10.1016/j.jns.2024.123111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Pineal cysts are frequently encountered as incidental findings in magnetic resonance imaging, usually devoid of symptoms, yet some patients exhibit symptomatic manifestations possibly associated with the cyst, even in the absence of hydrocephalus. The etiology of these symptoms remains contentious. This study aims to investigate the presence of lymphatic endothelial cell (LEC) markers and indications of inflammation or immune response within the pineal cysts of patients experiencing symptomatic non-hydrocephalic presentations. Eight patients who underwent surgical excision of their cysts were included in the study. Immunohistochemistry was utilized to assess the expression of LYVE-1, PDPN, and VEGFR3 as LEC markers, alongside IL-6 and CD3 for indications of inflammation or immune activity. Our analysis revealed an absence of inflammatory markers or immune response. However, a distinct expression of VEGFR3 was observed, likely localized to neurons within the pineal cyst tissue. We propose that these VEGFR3+ neurons within the pineal cyst may contribute to the headache symptoms reported by these patients. Further investigations are warranted to substantiate this hypothesis.
Collapse
Affiliation(s)
- César Luis Vera Quesada
- Department of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Shreyas Balachandra Rao
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Reidun Torp
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pitt Niehusmann
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Brain Fluid Research, University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Barron A, Barrett L, Tuulari J, Karlsson L, Karlsson H, McCarthy C, O'Keeffe G. sFlt-1 impairs neurite growth and neuronal differentiation in SH-SY5Y cells and human neurons. Biosci Rep 2024; 44:BSR20240562. [PMID: 38700092 PMCID: PMC11130541 DOI: 10.1042/bsr20240562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/05/2024] Open
Abstract
Pre-eclampsia (PE) is a hypertensive disorder of pregnancy which is associated with increased risk of neurodevelopmental disorders in exposed offspring. The pathophysiological mechanisms mediating this relationship are currently unknown, and one potential candidate is the anti-angiogenic factor soluble Fms-like tyrosine kinase 1 (sFlt-1), which is highly elevated in PE. While sFlt-1 can impair angiogenesis via inhibition of VEGFA signalling, it is unclear whether it can directly affect neuronal development independently of its effects on the vasculature. To test this hypothesis, the current study differentiated the human neural progenitor cell (NPC) line ReNcell® VM into a mixed culture of mature neurons and glia, and exposed them to sFlt-1 during development. Outcomes measured were neurite growth, cytotoxicity, mRNA expression of nestin, MBP, GFAP, and βIII-tubulin, and neurosphere differentiation. sFlt-1 induced a significant reduction in neurite growth and this effect was timing- and dose-dependent up to 100 ng/ml, with no effect on cytotoxicity. sFlt-1 (100 ng/ml) also reduced βIII-tubulin mRNA and neuronal differentiation of neurospheres. Undifferentiated NPCs and mature neurons/glia expressed VEGFA and VEGFR-2, required for endogenous autocrine and paracrine VEGFA signalling, while sFlt-1 treatment prevented the neurogenic effects of exogenous VEGFA. Overall, these data provide the first experimental evidence for a direct effect of sFlt-1 on neurite growth and neuronal differentiation in human neurons through inhibition of VEGFA signalling, clarifying our understanding of the potential role of sFlt-1 as a mechanism by which PE can affect neuronal development.
Collapse
Affiliation(s)
- Aaron Barron
- Department of Anatomy and Neuroscience, University College, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Lauren Barrett
- Department of Anatomy and Neuroscience, University College, Cork, Ireland
| | - Jetro J. Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry and Turku Brain and Mind Centre, University of Turku and Turku University Hospital, Turku, Finland
- Turku Collegium for Science, Medicine and Technology, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
- Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
- Department of Clinical Medicine, Unit of Public Health, University of Turku, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychiatry and Turku Brain and Mind Centre, University of Turku and Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku, Finland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy and Neuroscience, University College, Cork, Ireland
| |
Collapse
|
3
|
Szepanowski LP, Wruck W, Kapr J, Rossi A, Fritsche E, Krutmann J, Adjaye J. Cockayne Syndrome Patient iPSC-Derived Brain Organoids and Neurospheres Show Early Transcriptional Dysregulation of Biological Processes Associated with Brain Development and Metabolism. Cells 2024; 13:591. [PMID: 38607030 PMCID: PMC11011893 DOI: 10.3390/cells13070591] [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: 02/29/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Cockayne syndrome (CS) is a rare hereditary autosomal recessive disorder primarily caused by mutations in Cockayne syndrome protein A (CSA) or B (CSB). While many of the functions of CSB have been at least partially elucidated, little is known about the actual developmental dysregulation in this devasting disorder. Of particular interest is the regulation of cerebral development as the most debilitating symptoms are of neurological nature. We generated neurospheres and cerebral organoids utilizing Cockayne syndrome B protein (CSB)-deficient induced pluripotent stem cells derived from two patients with distinct severity levels of CS and healthy controls. The transcriptome of both developmental timepoints was explored using RNA-Seq and bioinformatic analysis to identify dysregulated biological processes common to both patients with CS in comparison to the control. CSB-deficient neurospheres displayed upregulation of the VEGFA-VEGFR2 signalling pathway, vesicle-mediated transport and head development. CSB-deficient cerebral organoids exhibited downregulation of brain development, neuron projection development and synaptic signalling. We further identified the upregulation of steroid biosynthesis as common to both timepoints, in particular the upregulation of the cholesterol biosynthesis branch. Our results provide insights into the neurodevelopmental dysregulation in patients with CS and strengthen the theory that CS is not only a neurodegenerative but also a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Leon-Phillip Szepanowski
- Institute for Stem Cell Research and Regenerative Medicine, University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Duesseldorf, Germany; (L.-P.S.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Duesseldorf, Germany; (L.-P.S.)
| | - Julia Kapr
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Andrea Rossi
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Ellen Fritsche
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Jean Krutmann
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Duesseldorf, Germany; (L.-P.S.)
- Zayed Centre for Research into Rare Diseases in Children (ZCR), University College London (UCL)—EGA Institute for Women’s Health, 20 Guilford Street, London WC1N 1DZ, UK
| |
Collapse
|
4
|
Mili B, Choudhary OP. Advancements and mechanisms of stem cell-based therapies for spinal cord injury in animals. Int J Surg 2024; 110:01279778-990000000-01011. [PMID: 38265419 PMCID: PMC11486964 DOI: 10.1097/js9.0000000000001074] [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: 10/23/2023] [Accepted: 12/24/2023] [Indexed: 01/25/2024]
Abstract
Spinal cord injury (SCI) is a neurodegenerative disorder of the central nervous system (CNS) that can lead to permanent loss of sensation and voluntary movement beyond the affected area. Extensive preclinical and clinical trials have been conducted to evaluate the safety and effectiveness of stem cells for the treatment of various CNS diseases or disorders, including SCI. However, several challenges hinder nerve cell regeneration in the injured spinal cord, such as extensive cell loss, limited neural cell regeneration capacity, axonal disruption, and the presence of growth-inhibiting molecules, particularly astroglial scarring or glial scars at the injury site in chronic cases. These obstacles pose significant challenges for physicians in restoring normal motor and sensory nerve function in both humans and animals following SCI. This review focuses on SCI pathogenesis, the mechanisms underlying the therapeutic potential of Mesenchymal Stem Cells (MSCs) in SCI, and the potential of stem cell-based therapies as promising avenues for treatment. This review article also included relevant preclinical and clinical data from animal studies.
Collapse
Affiliation(s)
- Bhabesh Mili
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Jalukie, Peren, Nagaland
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab, India
| |
Collapse
|
5
|
Sun R, Peng Q, Zhang F, Gao H, Li T, Wang L, Zhang L. Effect of vascular endothelial growth factor 165 on dopamine level in the retinas of guinea pigs with form-deprivation myopia. PeerJ 2023; 11:e16255. [PMID: 37849827 PMCID: PMC10578302 DOI: 10.7717/peerj.16255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Background Myopia is the most common refractive error because excessive increase in the axial length of a myopic eye leads to the thinning of the posterior scleral pole and can cause serious complications resulting in blindness. Thus, myopia has become a great concern worldwide. Dopamine (DA) plays a role in the development of myopia. Moreover, in Parkinson's disease, it has been proved that vascular endothelial growth factor 165 (VEGF165) can promote the survival and recovery of DA neurons, resulting in increased DA secretion in the striatum, thereby treating neuropathy. Therefore, we speculate that VEGF165 can also promote the release of DA in the retina to inhibit the occurrence and development of myopia. We aimed to investigate the effect of VEGF165 on DA levels in the retinas of guinea pigs with form-deprivation myopia (FDM) and the effects of DA on myopia prevention and control. Methods Healthy 3-week-old pigmented guinea pigs were randomly divided into blank, FDM, phosphate buffer saline (PBS), 1, 5, and 10 ng groups. The FDM model was established by covering the right eye continuously with a translucent latex balloon pullover for 14 days. The pigs in the PBS, 1, 5, and 10 ng groups were injected with PBS buffer and 1, 5, and 10 ng of VEGF165 recombinant human protein, respectively, in the vitreous of the right eye before masking. The refractive error and axial length were measured before and after modeling. All retinas were used for biomolecular analyses after 14 days. Results We found that the intravitreal injection of VEGF165 elevated DA levels in the retina and was effective in slowing the progression of myopia, and 1 ng of VEGF165 was the most effective. Moreover, the number of vascular endothelial cell nuclei in the 1 ng group was lower than that in the other VEGF165 groups. Conclusions Our data suggest that VEGF165 has a promoting effect on DA in the retinas of guinea pigs with FDM, potentially controlling the development of myopia.
Collapse
Affiliation(s)
- Ruiting Sun
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Qingsheng Peng
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fengyi Zhang
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Honglian Gao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Tong Li
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Lei Wang
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Lei Zhang
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| |
Collapse
|
6
|
Williamson MR, Le SP, Franzen RL, Donlan NA, Rosow JL, Nicot-Cartsonis MS, Cervantes A, Deneen B, Dunn AK, Jones TA, Drew MR. Subventricular zone cytogenesis provides trophic support for neural repair in a mouse model of stroke. Nat Commun 2023; 14:6341. [PMID: 37816732 PMCID: PMC10564905 DOI: 10.1038/s41467-023-42138-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
Stroke enhances proliferation of neural precursor cells within the subventricular zone (SVZ) and induces ectopic migration of newborn cells towards the site of injury. Here, we characterize the identity of cells arising from the SVZ after stroke and uncover a mechanism through which they facilitate neural repair and functional recovery. With genetic lineage tracing, we show that SVZ-derived cells that migrate towards cortical photothrombotic stroke in mice are predominantly undifferentiated precursors. We find that ablation of neural precursor cells or conditional knockout of VEGF impairs neuronal and vascular reparative responses and worsens recovery. Replacement of VEGF is sufficient to induce neural repair and recovery. We also provide evidence that CXCL12 from peri-infarct vasculature signals to CXCR4-expressing cells arising from the SVZ to direct their ectopic migration. These results support a model in which vasculature surrounding the site of injury attracts cells from the SVZ, and these cells subsequently provide trophic support that drives neural repair and recovery.
Collapse
Affiliation(s)
- Michael R Williamson
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
| | - Stephanie P Le
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Ronald L Franzen
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- School of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nicole A Donlan
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jill L Rosow
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | | | - Alexis Cervantes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience and Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience and Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Andrew K Dunn
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Theresa A Jones
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Michael R Drew
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
7
|
Andries L, Kancheva D, Masin L, Scheyltjens I, Van Hove H, De Vlaminck K, Bergmans S, Claes M, De Groef L, Moons L, Movahedi K. Immune stimulation recruits a subset of pro-regenerative macrophages to the retina that promotes axonal regrowth of injured neurons. Acta Neuropathol Commun 2023; 11:85. [PMID: 37226256 DOI: 10.1186/s40478-023-01580-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
The multifaceted nature of neuroinflammation is highlighted by its ability to both aggravate and promote neuronal health. While in mammals retinal ganglion cells (RGCs) are unable to regenerate following injury, acute inflammation can induce axonal regrowth. However, the nature of the cells, cellular states and signalling pathways that drive this inflammation-induced regeneration have remained elusive. Here, we investigated the functional significance of macrophages during RGC de- and regeneration, by characterizing the inflammatory cascade evoked by optic nerve crush (ONC) injury, with or without local inflammatory stimulation in the vitreous. By combining single-cell RNA sequencing and fate mapping approaches, we elucidated the response of retinal microglia and recruited monocyte-derived macrophages (MDMs) to RGC injury. Importantly, inflammatory stimulation recruited large numbers of MDMs to the retina, which exhibited long-term engraftment and promoted axonal regrowth. Ligand-receptor analysis highlighted a subset of recruited macrophages that exhibited expression of pro-regenerative secreted factors, which were able to promote axon regrowth via paracrine signalling. Our work reveals how inflammation may promote CNS regeneration by modulating innate immune responses, providing a rationale for macrophage-centred strategies for driving neuronal repair following injury and disease.
Collapse
Affiliation(s)
- Lien Andries
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Daliya Kancheva
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Isabelle Scheyltjens
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Hannah Van Hove
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Karen De Vlaminck
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Marie Claes
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, 3000, Louvain, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Naamsestraat 61, Box 2464, 3000, Louvain, Belgium.
| | - Kiavash Movahedi
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
8
|
Deyama S, Kaneda K. Role of neurotrophic and growth factors in the rapid and sustained antidepressant actions of ketamine. Neuropharmacology 2023; 224:109335. [PMID: 36403852 DOI: 10.1016/j.neuropharm.2022.109335] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
Abstract
The neurotrophic hypothesis of depression proposes that reduced levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) contribute to neuronal atrophy or loss in the prefrontal cortex (PFC) and hippocampus and impaired hippocampal adult neurogenesis, which are associated with depressive symptoms. Chronic, but acute, treatment with typical monoaminergic antidepressants can at least partially reverse these deficits, in part via induction of BDNF and/or VEGF expression, consistent with their delayed onset of action. Ketamine, an N-methyl-d-aspartate receptor antagonist, exerts rapid and sustained antidepressant effects. Rodent studies have revealed that ketamine rapidly increases BDNF and VEGF release and/or expression in the PFC and hippocampus, which in turn increases the number and function of spine synapses in the PFC and hippocampal neurogenesis. Ketamine also induces the persistent release of insulin-like growth factor 1 (IGF-1) in the PFC of male mice. These neurotrophic effects of ketamine are associated with its rapid and sustained antidepressant effects. In this review, we first provide an overview of the neurotrophic hypothesis of depression and then discuss the role of BDNF, VEGF, IGF-1, and other growth factors (IGF-2 and transforming growth factor-β1) in the antidepressant effects of ketamine and its enantiomers. This article is part of the Special Issue on 'Ketamine and its Metabolites'.
Collapse
Affiliation(s)
- Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| |
Collapse
|
9
|
Mashin VV, Belova LA, Kotova EY, Dolgova DR, Statenina AP, Belyaeva YK, Dergacheva AS, Israfilova RR. [Results of a multicenter observational program to evaluate the effectiveness of complex therapy of patients with chronic cerebrovascular pathology with cognitive impairment with Cortexin and Neuromexol (CORNELia study)]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:34-41. [PMID: 38147380 DOI: 10.17116/jnevro202312312134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
OBJECTIVE To evaluate the effectiveness of complex therapy with Cortexin and Neuromexol in patients with chronic cerebral ischemia (CCI) and cognitive impairment (CI). MATERIAL AND METHODS We examined 801 patients with CCI on the background of arterial hypertension and atherosclerosis with confirmed CI: 329 (41.1%) men and 472 (58.9%) women aged 30 to 80 years (mean age 64±10 years), who were examined. Cortexin and Neuromexol. Examination - Mini-Mental State Examination (MMSE) scale, hour-long drawing test (HDT) and severity of depressive states (Brief Geriatric Depression Scale, Mini Geriatric Depression Scal, MGDS). In 30 patients receiving Cortexin and Neuromexol (main group, MG) and 30 patients in the comparison group (CG), biomarkers of ischemic brain damage (NSE, antibodies to NR2, VEGFA) were determined. The examination was carried out before the start of treatment and after 30 days. RESULTS During therapy with Cortexin and Neuromexol, characteristic signs of a decrease in the severity of CI were noted (p<0.05). A positive correlation was revealed between the performance indicators of the MMSE and TFC tests, both before and after treatment (r=0.5 and r=0.6, respectively; p<0.05). A positive effect of therapy on the emotional background of patients was noted, in particular, a decrease in the severity of depressive symptoms on the MGDS scale. During therapy, a 2-fold decrease in the NSE level (p<0.05) was detected in the MG, which indicates a decrease in the structural and functional parameters of biomembrane neurons in the brain. The concentration of antibodies to NR2 decreased compared to the baseline level in both groups (p<0.05), and VEGFA decreased only in the MG (p<0.05). CONCLUSION The results of the study allow us to recommend the complex prescription of Cortexin 10 mg/day for 10 days and Neuromexol tablets 125 mg (375-750 mg/day) for 30 days for chronic CVD. Complex therapy with Cortexin and Neuromexol is effective and safe in patients with CCI and CI.
Collapse
Affiliation(s)
- V V Mashin
- Ulyanovsk State University, Ulyanovsk, Russia
| | - L A Belova
- Ulyanovsk State University, Ulyanovsk, Russia
| | - E Y Kotova
- Ulyanovsk State University, Ulyanovsk, Russia
| | - D R Dolgova
- Ulyanovsk State University, Ulyanovsk, Russia
| | | | | | | | | |
Collapse
|
10
|
Qin Q, Lee S, Patel N, Walden K, Gomez-Salazar M, Levi B, James AW. Neurovascular coupling in bone regeneration. Exp Mol Med 2022; 54:1844-1849. [PMID: 36446849 PMCID: PMC9722927 DOI: 10.1038/s12276-022-00899-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
The mammalian skeletal system is densely innervated by both neural and vascular networks. Peripheral nerves in the skeleton include sensory and sympathetic nerves. The crosstalk between skeletal and neural tissues is critical for skeletal development and regeneration. The cellular processes of osteogenesis and angiogenesis are coupled in both physiological and pathophysiological contexts. The cellular and molecular regulation of osteogenesis and angiogenesis have yet to be fully defined. This review will provide a detailed characterization of the regulatory role of nerves and blood vessels during bone regeneration. Furthermore, given the importance of the spatial relationship between nerves and blood vessels in bone, we discuss neurovascular coupling during physiological and pathological bone formation. A better understanding of the interactions between nerves and blood vessels will inform future novel therapeutic neural and vascular targeting for clinical bone repair and regeneration.
Collapse
Affiliation(s)
- Qizhi Qin
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Seungyong Lee
- grid.260024.20000 0004 0627 4571Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308 USA ,grid.412977.e0000 0004 0532 7395Department of Physical Education, Incheon National University, Incheon, 22012 South Korea
| | - Nirali Patel
- grid.260024.20000 0004 0627 4571Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| | - Kalah Walden
- grid.260024.20000 0004 0627 4571Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| | - Mario Gomez-Salazar
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Benjamin Levi
- grid.267313.20000 0000 9482 7121Departments of Surgery, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Aaron W. James
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
11
|
Fan XX, Sun WY, Li Y, Tang Q, Li LN, Yu X, Wang SY, Fan AR, Xu XQ, Chang HS. Honokiol improves depression-like behaviors in rats by HIF-1α- VEGF signaling pathway activation. Front Pharmacol 2022; 13:968124. [PMID: 36091747 PMCID: PMC9453876 DOI: 10.3389/fphar.2022.968124] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence indicates that the pathogenesis of depression is closely linked to impairments in neuronal synaptic plasticity. Honokiol, a biologically active substance extracted from Magnolia Officinalis, has been proven to exert significant antidepressant effects. However, the specific mechanism of action remains unclear. In this study, PC12 cells and chronic unpredictable mild stress (CUMS) model rats were used to explore the antidepressant effects and potential mechanisms of honokiol in vitro and in rats. In vitro experiment, a cell viability detection kit was used to screen the concentration and time of honokiol administration. PC12 cells were administered with hypoxia-inducible factor-1α (HIF-1α) blocker, 2-methoxyestradiol (2-ME), and vascular endothelial growth factor receptor 2 (VEGFR-2) blocker, SU5416, to detect the expression of HIF-1α, VEGF, synaptic protein 1 (SYN 1), and postsynaptic density protein 95 (PSD 95) by western blotting. In effect, we investigated whether the synaptic plasticity action of honokiol was dependent on the HIF-1α-VEGF pathway. In vivo, behavioral tests were used to evaluate the reproducibility of the CUMS depression model and depression-like behaviors. Molecular biology techniques were used to examine mRNA and protein expression of the HIF-1α-VEGF signaling pathway and synaptic plasticity-related regulators. Additionally, molecular docking techniques were used to study the interaction between honokiol and target proteins, and predict their binding patterns and affinities. Experimental results showed that honokiol significantly reversed CUMS-induced depression-like behaviors. Mechanically, honokiol exerted a significant antidepressant effect by enhancing synaptic plasticity. At the molecular level, honokiol can activate the HIF-1α-VEGF signaling pathway in vitro and in vivo, as well as promote the protein expression levels of SYN 1 and PSD 95. Taken together, the results do not only provide an experimental basis for honokiol in the clinical treatment of depression but also suggest that the HIF-1α-VEGF pathway may be a potential target for the treatment of depression.
Collapse
Affiliation(s)
- Xiao-Xu Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wen-Yan Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qin Tang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Li-Na Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shu-Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ang-Ran Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiang-Qing Xu
- Experiment Center, Encephalopathy Department, Affiliated Hospital of Shandong University of Chinese Medicine, Jinan, China
- *Correspondence: Hong-Sheng Chang, ; Xiang-Qing Xu,
| | - Hong-Sheng Chang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Hong-Sheng Chang, ; Xiang-Qing Xu,
| |
Collapse
|
12
|
Morphogenesis of vascular and neuronal networks and the relationships between their remodeling processes. Brain Res Bull 2022; 186:62-69. [DOI: 10.1016/j.brainresbull.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/29/2022] [Indexed: 11/21/2022]
|
13
|
Sakai D, Sugawara T, Kurokawa T, Murakami Y, Tomosugi M, Masuta H, Sakata-Haga H, Hatta T, Shoji H. Hif1α-dependent hypoxia signaling contributes to the survival of deep-layer neurons and cortex formation in a mouse model. Mol Brain 2022; 15:28. [PMID: 35361248 PMCID: PMC8973788 DOI: 10.1186/s13041-022-00911-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Hypoxia-inducible factor 1 α (Hif1α) plays a crucial role in brain development. To study the function of Hif1α in early brain development, we generated neuroepithelial cell-specific Hif1α-knockout mice. Hif1α-knockout mice died soon after birth; these mice exhibited an abnormal head shape, indicating the presence of brain defects. Morphological analysis revealed that Hif1α ablation reduced the overall size of the brain, especially affecting the telencephalon. Neuronal apoptosis predominantly occurred in deep-layer neurons, consequently the alignment of cortical layers was severely disorganized in Hif1α knockout mice. Furthermore, we demonstrated that Vegf signaling contributes to the survival of deep-layer neurons as a downstream effector of Hif1α-dependent hypoxia signaling. Taken together, our findings demonstrate that Hif1α plays a critical role in the early stages of telencephalon development.
Collapse
Affiliation(s)
- Daisuke Sakai
- Department of Biology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan.
| | - Takeru Sugawara
- Department of Medical Life Systems, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Tomonori Kurokawa
- Department of Medical Life Systems, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Yuki Murakami
- Department of Hygiene and Public Health, Kansai Medical University, Osaka, Hirakata, 573-1010, Japan
| | - Mitsuhiro Tomosugi
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiroko Masuta
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiroki Shoji
- Department of Biology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
14
|
Rampino A, Annese T, Torretta S, Tamma R, Maria Falcone R, Ribatti D. Involvement of vascular endothelial growth factor in schizophrenia. Neurosci Lett 2021; 760:136093. [PMID: 34216717 DOI: 10.1016/j.neulet.2021.136093] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF), which acts as an angiogenic and neurotrophic factor, is involved the regulation of cerebral blood volume and flow in Schizophrenia (SCZ). Several evidence indicates that modification of brain blood circulation due to alterations in the VEGF system affects cognitive performance and brain function in patients with SCZ. The aim of this study is: 1) To analyze the literature data concerning the role of VEGF in modulating the angiogenic response in SCZ. These data are controversial because some studies found elevated VEGF serum levels of VEGF in patients with SCZ, whereas others demonstrated no significant differences between SCZ patients and controls. 2)To analyze the role of VEGF as a predictive factor on the effects of antipsychotics agents used in the treatment of SCZ. In this context, high VEGF levels, associated to better responses to antipsychotics, might be predictive of the use of first generation antipsycotic drugs, whereas low VEGF levels, expression of resistance to therapy, might be predictive for the use of second generation antipsycotic drugs.
Collapse
Affiliation(s)
- Antonio Rampino
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Silvia Torretta
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Rosa Maria Falcone
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
15
|
Xiao G, Lyu M, Li Z, Cao L, Liu X, Wang Y, He S, Chen Z, Du H, Feng Y, Wang J, Zhu Y. Restoration of early deficiency of axonal guidance signaling by guanxinning injection as a novel therapeutic option for acute ischemic stroke. Pharmacol Res 2021; 165:105460. [PMID: 33513357 DOI: 10.1016/j.phrs.2021.105460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 01/12/2023]
Abstract
Despite of its high morbidity and mortality, there is still a lack of effective treatment for ischemic stroke in part due to our incomplete understanding of molecular mechanisms of its pathogenesis. In this study, we demonstrate that SHH-PTCH1-GLI1-mediated axonal guidance signaling and its related neurogenesis, a central pathway for neuronal development, also plays a critical role in early stage of an acute stroke model. Specifically, in vivo, we evaluated the effect of GXNI on ischemic stroke mice via using the middle cerebral artery embolization model, and found that GXNI significantly alleviated cerebral ischemic reperfusion (I/R) injury by reducing the volume of cerebral infarction, neurological deficit score and cerebral edema, reversing the BBB permeability and histopathological changes. A combined approach of RNA-seq and network pharmacology analysis was used to reveal the underlying mechanisms of GXNI followed by RT-PCR, immunohistochemistry and western blotting validation. It was pointed out that axon guidance signaling pathway played the most prominent role in GXNI action with Shh, Ptch1, and Gli1 genes as the critical contributors in brain protection. In addition, GXNI markedly prevented primary cortical neuron cells from oxygen-glucose deprivation/reoxygenation damage in vitro, and promoted axon growth and synaptogenesis of damaged neurons, which further confirmed the results of in vivo experiments. Moreover, due to the inhibition of the SHH-PTCH1-GLI1 signaling pathway by cyclopropylamine, the effect of GXNI was significantly weakened. Hence, our study provides a novel option for the clinical treatment of acute ischemic stroke by GXNI via SHH-PTCH1-GLI1-mediated axonal guidance signaling, a neuronal development pathway previously considered for after-stroke recovery.
Collapse
Affiliation(s)
- Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Ming Lyu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhixiong Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Linghua Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Zihao Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Hongxia Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Jigang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China.
| |
Collapse
|
16
|
Neuroprotective Effect of Vascular Endothelial Growth Factor on Motoneurons of the Oculomotor System. Int J Mol Sci 2021; 22:ijms22020814. [PMID: 33467517 PMCID: PMC7830098 DOI: 10.3390/ijms22020814] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 01/04/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) was initially characterized as a potent angiogenic factor based on its activity on the vascular system. However, it is now well established that VEGF also plays a crucial role as a neuroprotective factor in the nervous system. A deficit of VEGF has been related to motoneuronal degeneration, such as that occurring in amyotrophic lateral sclerosis (ALS). Strikingly, motoneurons of the oculomotor system show lesser vulnerability to neurodegeneration in ALS compared to other motoneurons. These motoneurons presented higher amounts of VEGF and its receptor Flk-1 than other brainstem pools. That higher VEGF level could be due to an enhanced retrograde input from their target muscles, but it can also be produced by the motoneurons themselves and act in an autocrine way. By contrast, VEGF’s paracrine supply from the vicinity cells, such as glial cells, seems to represent a minor source of VEGF for brainstem motoneurons. In addition, ocular motoneurons experiment an increase in VEGF and Flk-1 level in response to axotomy, not observed in facial or hypoglossal motoneurons. Therefore, in this review, we summarize the differences in VEGF availability that could contribute to the higher resistance of extraocular motoneurons to injury and neurodegenerative diseases.
Collapse
|
17
|
Han H, Yang C, Zhang Y, Han C, Zhang G. Vascular Endothelial Growth Factor Mediates the Sprouted Axonogenesis of Breast Cancer in Rat. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:515-526. [PMID: 33345997 DOI: 10.1016/j.ajpath.2020.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022]
Abstract
Nerve infiltration into the tumor is a common feature of the tumor microenvironment. The mechanisms of axonogenesis in breast cancer remain unclear. We hypothesized that vascular endothelial growth factor (VEGF), as well as nerve growth factor (NGF), is involved in the axonogenesis of breast cancer. A N-methyl-N-nitrosourea (MNU)-induced rat model of breast cancer was used to explore the presence of axonogenesis in breast tumor and the involvement of VEGF, as well as NGF, in the axonogenesis of breast tumor. Nerve infiltration into the tumor was found in MNU-induced rat model of breast cancer including the sensory and sympathetic nerve fibers. Nerve density was increased following the growth of tumor. The sensory neurons innervating the thoracic and abdominal mammary tumors peaked at T5 to T6 and L1 to L2 dorsal root ganglions, respectively. Either VEGF receptor inhibitor or antibody against VEGF receptor 2, as well as NGF receptor inhibitor, apparently decreased both the nerve density and vascular density of breast tumor. The reduced nerve density was correlated with the decreased vascular density induced by these treatments. In cultured dorsal root ganglion neurons, phosphatidylinositol 3 (PI3K)/Akt, extracellular signal-regulated protein kinase (ERK), and p38 inhibitors significantly attenuated VEGF-induced neurite elongation. These findings provide direct evidence that VEGF, as well as NGF, may control the axonogenesis of breast cancer.
Collapse
Affiliation(s)
- Hongxiu Han
- Department of Pathology, Tongji Hospital, Tongji University, Shangha, China; Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chunxue Yang
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Zhang
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changhao Han
- Department of Clinical Medicine, Second Clinical Medical College, Chongqing Medical University, Chongqing, China
| | - Guohua Zhang
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Ureña-Guerrero ME, Castañeda-Cabral JL, Rivera-Cervantes MC, Macias-Velez RJ, Jarero-Basulto JJ, Gudiño-Cabrera G, Beas-Zárate C. Neuroprotective and Neurorestorative Effects of Epo and VEGF: Perspectives for New Therapeutic Approaches to Neurological Diseases. Curr Pharm Des 2020; 26:1263-1276. [PMID: 31942853 DOI: 10.2174/1381612826666200114104342] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Erythropoietin (Epo) and vascular endothelial growth factor (VEGF) are two vasoactive molecules with essential trophic effects for brain development. The expression and secretion of both molecules increase in response to neuronal damage and they exert protective and restorative effects, which may also be accompanied by adverse side effects. OBJECTIVE We review the most relevant evidence on the neuroprotective and neurorestorative effects of Epo and VEGF in three of the most frequent neurological disorders, namely, stroke, epilepsy and Alzheimer's disease, to develop new therapeutic approaches. METHODS Several original scientific manuscripts and reviews that have discussed the evidence in critical way, considering both the beneficial and adverse effects of Epo and VEGF in the selected neurological disorders, were analysed. In addition, throughout this review, we propose several considerations to take into account in the design of therapeutic approaches based on Epo and VEGF signalling. RESULTS Although the three selected disorders are triggered by different mechanisms, they evolve through similar processes: excitotoxicity, oxidative stress, neuroinflammation, neuronal death, glial reactivity and vascular remodelling. Epo and VEGF exert neuroprotective and neurorestorative effects by acting on these processes due to their pleiotropism. In general, the evidence shows that both Epo and VEGF reduce neuronal death but that at the vascular level, their effects are contradictory. CONCLUSION Because the Epo and VEGF signalling pathways are connected in several ways, we conclude that more experimental studies, primarily studies designed to thoroughly assess the functional interactions between Epo and VEGF in the brain under both physiological and pathophysiological conditions, are needed.
Collapse
Affiliation(s)
- Mónica E Ureña-Guerrero
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - José L Castañeda-Cabral
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico.,Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (CINVESTAV sede Sur), IPN, Ciudad de México, México
| | - Martha C Rivera-Cervantes
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Rafael J Macias-Velez
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - José J Jarero-Basulto
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Graciela Gudiño-Cabrera
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Carlos Beas-Zárate
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| |
Collapse
|
19
|
Chen Q, Liu J, Sawada T, Wei C, Wu S, Han F. Possible role of EphA4 and VEGFR2 interactions in neural stem and progenitor cell differentiation. Exp Ther Med 2020; 19:1789-1796. [PMID: 32104234 PMCID: PMC7027147 DOI: 10.3892/etm.2020.8419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Abstract
Neural stem and progenitor cells (NSPCs) are important pluripotent stem cells, which have potential applications for cell replacement therapy. Ephrin receptors (Ephs) and angiogenic growth factor receptors have a major impact on the proliferation and differentiation of NSPCs. Potential interactions between EphA4 and vascular endothelial growth factor (VEGF) receptor (VEGFR) 2, and their roles in NSPC differentiation in vitro remain unknown. In the present study, mouse embryonic NSPCs were treated with ephrin-A1 or VEGF165 alone as well as with combination treatment (ephrin-A1 + VEGF165). Immunoprecipitation and immunoblot assays demonstrated that wild-type EphA4, but not the EphA4 kinase-dead mutant, interacted with VEGFR2 when overexpressed in 293T cells. This interaction was inhibited by dominant-negative EphA4. The percentage of β-tubulin III (Tuj1)+, but not glial fibrillary acid protein (GFAP)+ cells, was increased in the ephrin-A1 + VEGF165 combination group as compared to the VEGF165 alone group in mouse embryonic NSPCs. VEGF165-induced neuronal differentiation was potentiated by ephrin-A1 in NSPCs in vitro and ephrin-A1- or VEGF165-stimulated EphA4 and VEGFR2 interactions may mediate the signaling pathway.
Collapse
Affiliation(s)
- Qingfa Chen
- Centre for Tissue Engineering and Regenerative Medicine, Liaocheng People's Hospital, Liaocheng University, Liaocheng, Shandong 252000, P.R. China
| | - Jia Liu
- Department of Neurology, Liaocheng People's Hospital, Liaocheng University, Liaocheng, Shandong 252000, P.R. China
| | - Takahiro Sawada
- Department of Molecular Cell Biology and Molecular Medicine, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera, Wakayama 641-8509, Japan
| | - Chuanfei Wei
- Centre for Tissue Engineering and Regenerative Medicine, Liaocheng People's Hospital, Liaocheng University, Liaocheng, Shandong 252000, P.R. China
| | - Shichao Wu
- Centre for Tissue Engineering and Regenerative Medicine, Liaocheng People's Hospital, Liaocheng University, Liaocheng, Shandong 252000, P.R. China
| | - Fabin Han
- Centre for Tissue Engineering and Regenerative Medicine, Liaocheng People's Hospital, Liaocheng University, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
20
|
Deyama S, Duman RS. Neurotrophic mechanisms underlying the rapid and sustained antidepressant actions of ketamine. Pharmacol Biochem Behav 2020; 188:172837. [PMID: 31830487 PMCID: PMC6997025 DOI: 10.1016/j.pbb.2019.172837] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/30/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022]
Abstract
Clinical and preclinical studies have demonstrated that depression, one of the most common psychiatric illnesses, is associated with reduced levels of neurotrophic factors, including brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), contributing to neuronal atrophy in the prefrontal cortex (PFC) and hippocampus, and reduced hippocampal adult neurogenesis. Conventional monoaminergic antidepressants can block/reverse, at least partially, these deficits in part via induction of BDNF and/or VEGF, although these drugs have significant limitations, notably a time lag for therapeutic response and low response rates. Recent studies reveal that ketamine, an N-methyl-d-aspartate receptor antagonist produces rapid (within hours) and sustained (up to a week) antidepressant actions in both patients with treatment-resistant depression and rodent models of depression. Rodent studies also demonstrate that ketamine rapidly increases BDNF and VEGF release and/or expression in the medial PFC (mPFC) and hippocampus, leading to increase in the number and function of spine synapses in the mPFC and enhancement of hippocampal neurogenesis. These neurotrophic effects of ketamine are associated with the antidepressant effects of this drug. Together, these findings provide evidence for a neurotrophic mechanism underlying the rapid and sustained antidepressant actions of ketamine and pave the way for the development of rapid and more effective antidepressants with fewer side effects than ketamine.
Collapse
Affiliation(s)
- Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan.
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
21
|
Harde E, Nicholson L, Furones Cuadrado B, Bissen D, Wigge S, Urban S, Segarra M, Ruiz de Almodóvar C, Acker-Palmer A. EphrinB2 regulates VEGFR2 during dendritogenesis and hippocampal circuitry development. eLife 2019; 8:49819. [PMID: 31868584 PMCID: PMC6927743 DOI: 10.7554/elife.49819] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic factor that play important roles in the nervous system, although it is still unclear which receptors transduce those signals in neurons. Here, we show that in the developing hippocampus VEGFR2 (also known as KDR or FLK1) is expressed specifically in the CA3 region and it is required for dendritic arborization and spine morphogenesis in hippocampal neurons. Mice lacking VEGFR2 in neurons (Nes-cre Kdrlox/-) show decreased dendritic arbors and spines as well as a reduction in long-term potentiation (LTP) at the associational-commissural – CA3 synapses. Mechanistically, VEGFR2 internalization is required for VEGF-induced spine maturation. In analogy to endothelial cells, ephrinB2 controls VEGFR2 internalization in neurons. VEGFR2-ephrinB2 compound mice (Nes-cre Kdrlox/+ Efnb2lox/+) show reduced dendritic branching, reduced spine head size and impaired LTP. Our results demonstrate the functional crosstalk of VEGFR2 and ephrinB2 in vivo to control dendritic arborization, spine morphogenesis and hippocampal circuitry development.
Collapse
Affiliation(s)
- Eva Harde
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - LaShae Nicholson
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Beatriz Furones Cuadrado
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Diane Bissen
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Sylvia Wigge
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Severino Urban
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Marta Segarra
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Carmen Ruiz de Almodóvar
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany.,European Center for Angioscience, Medicine Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Institute for Transfusion Medicine and Immunology, Medicine Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
22
|
Duman RS, Deyama S, Fogaça MV. Role of BDNF in the pathophysiology and treatment of depression: Activity-dependent effects distinguish rapid-acting antidepressants. Eur J Neurosci 2019; 53:126-139. [PMID: 31811669 DOI: 10.1111/ejn.14630] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/14/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
The pathophysiology and treatment of depression have been the focus of intense research and while there is much that remains unknown, modern neurobiological approaches are making progress. This work demonstrates that stress and depression are associated with atrophy of neurons and reduced synaptic connectivity in brain regions such as the hippocampus and prefrontal cortex that contribute to depressive behaviors, and conversely that antidepressant treatment can reverse these deficits. The role of neurotrophic factors, particularly brain-derived neurotrophic factor (BDNF), has been of particular interest as these factors play a key role in activity-dependent regulation of synaptic plasticity. Here, we review the literature demonstrating that exposure to stress and depression decreases BDNF expression in the hippocampus and PFC and conversely that antidepressant treatment can up-regulate BDNF in the adult brain and reverse the effects of stress. We then focus on rapid-acting antidepressants, particularly the NMDA receptor antagonist ketamine, which produces rapid synaptic and antidepressant behavioral actions that are dependent on activity-dependent release of BDNF. This rapid release of BDNF differs from typical monoaminergic agents that require chronic administration to produce a slow induction of BDNF expression, consistent with the time lag for the therapeutic action of these agents. We review evidence that other classes of rapid-acting agents also require BDNF release, demonstrating that this is a common, convergent downstream mechanism. Finally, we discuss evidence that the actions of ketamine are also dependent on another growth factor, vascular endothelial growth factor (VEGF) and its complex interplay with BDNF.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Manoela Viar Fogaça
- Department of Psychiatry and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
23
|
Abstract
Impaired neurocognitive function is an increasingly recognized morbidity in patients who have cancer. Cancer treatments, psychosocial stressors, and the malignancy itself can alter brain function. The mechanisms by which this occurs are under active investigation. Although there is a growing appreciation of its prevalence and causes, there remain limited therapeutic options for the treatment of neurocognitive dysfunction in this population. A persistent scientific and clinical effort to understand its mechanisms and impact is critical to the care of oncology patients.
Collapse
Affiliation(s)
- Rebecca A Harrison
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 0431, Houston, TX 77030, USA.
| | - Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 0431, Houston, TX 77030, USA
| |
Collapse
|
24
|
Lee YJ, Ch'ng TH. RIP at the Synapse and the Role of Intracellular Domains in Neurons. Neuromolecular Med 2019; 22:1-24. [PMID: 31346933 DOI: 10.1007/s12017-019-08556-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
Abstract
Regulated intramembrane proteolysis (RIP) occurs in a cell when transmembrane proteins are cleaved by intramembrane proteases such as secretases to generate soluble protein fragments in the extracellular environment and the cytosol. In the cytosol, these soluble intracellular domains (ICDs) have local functions near the site of cleavage or in many cases, translocate to the nucleus to modulate gene expression. While the mechanism of RIP is relatively well studied, the fate and function of ICDs for most substrate proteins remain poorly characterized. In neurons, RIP occurs in various subcellular compartments including at the synapse. In this review, we summarize current research on RIP in neurons, focusing specifically on synaptic proteins where the presence and function of the ICDs have been reported. We also briefly discuss activity-driven processing of RIP substrates at the synapse and the cellular machinery that support long-distance transport of ICDs from the synapse to the nucleus. Finally, we describe future challenges in this field of research in the context of understanding the contribution of ICDs in neuronal function.
Collapse
Affiliation(s)
- Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore.,Interdisciplinary Graduate School (IGS), Nanyang Technological University, Singapore, Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
25
|
Deyama S, Bang E, Kato T, Li XY, Duman RS. Neurotrophic and Antidepressant Actions of Brain-Derived Neurotrophic Factor Require Vascular Endothelial Growth Factor. Biol Psychiatry 2019; 86:143-152. [PMID: 30712809 PMCID: PMC6597338 DOI: 10.1016/j.biopsych.2018.12.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Activity-dependent release of brain-derived neurotrophic factor (BDNF) in the medial prefrontal cortex (mPFC) is essential for the rapid and sustained antidepressant actions of ketamine, and a recent study shows a similar requirement for vascular endothelial growth factor (VEGF). Since BDNF is reported to stimulate VEGF expression and/or release in neuroblastoma cells, the present study tested the hypothesis that the actions of BDNF are mediated by VEGF. METHODS The role of VEGF in the antidepressant behavioral actions of BDNF was tested by intra-mPFC coinfusion of a VEGF neutralizing antibody and by neuron-specific deletion of VEGF. The influence of BDNF on the release of VEGF and the role of VEGF in the neurotrophic actions of BDNF were determined in rat primary cortical neurons. The role of BDNF in the behavioral and neurotrophic actions of VEGF was also determined. RESULTS The results show that the rapid and sustained antidepressant-like actions of intra-mPFC BDNF are blocked by coinfusion of a VEGF neutralizing antibody, and that neuron-specific mPFC deletion of VEGF blocks the antidepressant-like actions of BDNF. Studies in primary cortical neurons demonstrate that BDNF stimulates the release of VEGF and that BDNF induction of dendrite complexity is blocked by a selective VEGF-fetal liver kinase 1 receptor antagonist. Surprisingly, the results also show reciprocal interactions, indicating that the behavioral and neurotrophic actions of VEGF are dependent on BDNF. CONCLUSIONS These findings indicate that the antidepressant-like and neurotrophic actions of BDNF require VEGF signaling, but they also demonstrate reciprocal interdependence for BDNF in the actions of VEGF.
Collapse
Affiliation(s)
- Satoshi Deyama
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Eunyoung Bang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Taro Kato
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Drug Development Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Suita 564-0053, Japan
| | - Xiao-Yuan Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ronald S. Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Correspondence: Ronald S. Duman, Ph.D., Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA. Tel: 203-974-7726, Fax: 203-974-7724,
| |
Collapse
|
26
|
Deyama S, Bang E, Wohleb ES, Li XY, Kato T, Gerhard DM, Dutheil S, Dwyer JM, Taylor SR, Picciotto MR, Duman RS. Role of Neuronal VEGF Signaling in the Prefrontal Cortex in the Rapid Antidepressant Effects of Ketamine. Am J Psychiatry 2019; 176:388-400. [PMID: 30606046 PMCID: PMC6494682 DOI: 10.1176/appi.ajp.2018.17121368] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The N-methyl-d-aspartate receptor antagonist ketamine produces rapid and sustained antidepressant actions even in patients with treatment-resistant depression. Vascular endothelial growth factor (VEGF) has been implicated in the effects of conventional monoamine-based antidepressants, but the role of VEGF in the rapid antidepressant actions of ketamine remains unclear. The authors examined whether neuronal VEGF signaling in the medial prefrontal cortex (mPFC) mediates the rapid antidepressant actions of ketamine. METHODS The authors used a combination of approaches, including conditional, neuron-specific knockout of VEGF or its receptor, Flk-1; antibody neutralization; viral-mediated knockdown of Flk-1; and pharmacological inhibitors. Further in vitro and in vivo experiments were performed to examine whether neuronal VEGF signaling was required for the neurotrophic and synaptogenic actions of ketamine that underlie its behavioral actions. RESULTS The behavioral actions of systemic ketamine are blocked by forebrain excitatory neuron-specific deletion of either VEGF or Flk-1 or by intra-mPFC infusion of a VEGF neutralizing antibody. Moreover, intra-mPFC infusions of VEGF are sufficient to produce rapid ketamine-like behavioral actions, and these effects are blocked by neuron-specific Flk-1 deletion. The results also show that local knockdown of Flk-1 in mPFC excitatory neurons in adulthood blocks the behavioral effects of systemic ketamine. Moreover, inhibition of neuronal VEGF signaling blocks the neurotrophic and synaptogenic effects of ketamine. CONCLUSIONS Together, these findings indicate that neuronal VEGF-Flk-1 signaling in the mPFC plays an essential role in the antidepressant actions of ketamine.
Collapse
Affiliation(s)
- Satoshi Deyama
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Eunyoung Bang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Eric S. Wohleb
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - Xiao-Yuan Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Taro Kato
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Drug Development Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Suita 564-0053, Japan
| | - Danielle M. Gerhard
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Sophie Dutheil
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Jason M. Dwyer
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Seth R. Taylor
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ronald S. Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA.,Correspondence: Ronald S. Duman, Ph.D., Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA. Tel: 203-974-7726 Fax: 203-974-7724
| |
Collapse
|
27
|
Latzer P, Shchyglo O, Hartl T, Matschke V, Schlegel U, Manahan-Vaughan D, Theiss C. Blocking VEGF by Bevacizumab Compromises Electrophysiological and Morphological Properties of Hippocampal Neurons. Front Cell Neurosci 2019; 13:113. [PMID: 30971896 PMCID: PMC6445260 DOI: 10.3389/fncel.2019.00113] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/07/2019] [Indexed: 12/20/2022] Open
Abstract
A hallmark of glioblastoma multiforme (GBM) is neoangiogenesis, mediated by the overexpression of vascular endothelial growth factor (VEGF). Anti-VEGF antibodies, like bevacizumab, prolong progression-free survival in GBM, however, this treatment has been reported to be associated with a decline in neurocognitive function. Therefore, this study focused on the effects of bevacizumab on neuronal function and plasticity. We analyzed neuronal membrane properties and synaptic plasticity in rat hippocampal slices, as well as spine dynamics in dissociated hippocampal neurons, to examine the impact of bevacizumab on hippocampal function and viability. VEGF inhibition resulted in profound impairments in hippocampal synaptic plasticity as well as reductions in dendritic spine number and length. Physiological properties of hippocampal neurons were also affected. These effects of VEGF blockade on hippocampal function may play a role in compromising memory and information processing and thus, may contribute to neurocognitive dysfunction in GBM patients treated with bevacizumab.
Collapse
Affiliation(s)
- Pauline Latzer
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Olena Shchyglo
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Tim Hartl
- International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany.,Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Uwe Schlegel
- Department of Neurology, Knappschaftskrankenhaus, Ruhr University Bochum, Bochum, Germany
| | | | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
28
|
Treadmill exercise ameliorates focal cerebral ischemia/reperfusion-induced neurological deficit by promoting dendritic modification and synaptic plasticity via upregulating caveolin-1/VEGF signaling pathways. Exp Neurol 2019; 313:60-78. [DOI: 10.1016/j.expneurol.2018.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/02/2018] [Accepted: 12/10/2018] [Indexed: 11/18/2022]
|
29
|
Marques BL, Carvalho GA, Freitas EMM, Chiareli RA, Barbosa TG, Di Araújo AGP, Nogueira YL, Ribeiro RI, Parreira RC, Vieira MS, Resende RR, Gomez RS, Oliveira-Lima OC, Pinto MCX. The role of neurogenesis in neurorepair after ischemic stroke. Semin Cell Dev Biol 2019; 95:98-110. [PMID: 30550812 DOI: 10.1016/j.semcdb.2018.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
Stroke consists of an abrupt reduction of cerebral blood flow resulting in hypoxia that triggers an excitotoxicity, oxidative stress, and neuroinflammation. After the ischemic process, neural precursor cells present in the subventricular zone of the lateral ventricle and subgranular zone of the dentate gyrus proliferate and migrate towards the lesion, contributing to the brain repair. The neurogenesis is induced by signal transduction pathways, growth factors, attractive factors for neuroblasts, transcription factors, pro and anti-inflammatory mediators and specific neurotransmissions. However, this endogenous neurogenesis occurs slowly and does not allow a complete restoration of brain function. Despite that, understanding the mechanisms of neurogenesis could improve the therapeutic strategies for brain repair. This review presents the current knowledge about brain repair process after stroke and the perspectives regarding the development of promising therapies that aim to improve neurogenesis and its potential to form new neural networks.
Collapse
Affiliation(s)
- Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Gustavo A Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Elis M M Freitas
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Raphaela A Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Thiago G Barbosa
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Armani G P Di Araújo
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Yanley L Nogueira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Raul I Ribeiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Ricardo C Parreira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renato S Gomez
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Onésia C Oliveira-Lima
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Mauro C X Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil.
| |
Collapse
|
30
|
Tang F, LeBlanc ME, Wang W, Liang D, Chen P, Chou TH, Tian H, Li W. Anti-secretogranin III therapy of oxygen-induced retinopathy with optimal safety. Angiogenesis 2019; 22:369-382. [PMID: 30644010 DOI: 10.1007/s10456-019-09662-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Retinopathy of prematurity (ROP) with pathological retinal neovascularization is the most common cause of blindness in children. ROP is currently treated with laser therapy or cryotherapy, both of which may adversely affect the peripheral vision with limited efficacy. Owing to the susceptibility of the developing retina and vasculatures to pharmacological intervention, there is currently no approved drug therapy for ROP in preterm infants. Secretogranin III (Scg3) was recently discovered as a highly disease-restricted angiogenic factor, and a Scg3-neutralizing monoclonal antibody (mAb) was reported with high efficacy to alleviate oxygen-induced retinopathy (OIR) in mice, a surrogate model of ROP. Herein we independently investigated the efficacy of anti-Scg3 mAb in OIR mice and characterized its safety in neonatal mice. We developed a new Scg3-neutralizing mAb recognizing a distinct epitope and independently established the therapeutic activity of anti-Scg3 therapy to alleviate OIR-induced pathological retinal neovascularization in mice. Importantly, anti-Scg3 mAb showed no detectable adverse effects on electroretinography and developing retinal vasculature. Furthermore, systemic anti-Scg3 mAb induced no renal tubular injury or abnormality in kidney vessel development and body weight gain of neonatal mice. In contrast, anti-vascular endothelial growth factor drug aflibercept showed significant side effects in neonatal mice. These results suggest that anti-Scg3 mAb may have the safety and efficacy profiles required for ROP therapy.
Collapse
Affiliation(s)
- Fen Tang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Michelle E LeBlanc
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Weiwen Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Dan Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Ping Chen
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
- Department of Ophthalmology, Renji Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Tsung-Han Chou
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Hong Tian
- Everglades Biopharma, LLC, Miami, FL, USA
| | - Wei Li
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.
| |
Collapse
|
31
|
Secretogranin III as a novel target for the therapy of choroidal neovascularization. Exp Eye Res 2019; 181:120-126. [PMID: 30633921 DOI: 10.1016/j.exer.2019.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 12/20/2018] [Accepted: 01/07/2019] [Indexed: 01/08/2023]
Abstract
Wet age-related macular degeneration (AMD) with choroidal neovascularization (CNV) is a leading cause of vision loss in the elderly. The advent of anti-vascular endothelial growth factor (VEGF) drugs represents a major breakthrough in wet AMD therapy but with limited efficacy to improve visual acuity. Secretogranin III (Scg3, SgIII) was recently discovered as a novel angiogenic factor with VEGF-independent mechanisms. Scg3-neutralizing monoclonal antibody (mAb) was reported to alleviate pathological retinal neovascularization in oxygen-induced retinopathy mice and retinal vascular leakage in diabetic mice with high efficacy and disease selectivity. Herein we investigated whether Scg3 is a novel angiogenic target for CNV therapy in mouse models. We found that anti-Scg3 ML49.3 mAb inhibited Scg3-induced proliferation and Src phosphorylation in human retinal microvascular endothelial cells. Intravitreal injection of Scg3-neutralizing polyclonal antibodies (pAb) or mAb significantly attenuated laser-induced CNV leakage, CNV 3D volume, lesion area and vessel density. Furthermore, subcutaneous administration of Scg3-neutralizing pAb or mAb significantly prevented Matrigel-induced CNV. The efficacy of anti-Scg3 pAb or mAb was comparable to VEGF inhibitor aflibercept. These findings suggest that Scg3 plays an important role in CNV pathogenesis and that anti-Scg3 mAb efficiently ameliorates laser- or Matrigel-induced CNV.
Collapse
|
32
|
Zhang H, Wang Y, He Z. Glycine-Histidine-Lysine (GHK) Alleviates Neuronal Apoptosis Due to Intracerebral Hemorrhage via the miR-339-5p/VEGFA Pathway. Front Neurosci 2018; 12:644. [PMID: 30294253 PMCID: PMC6158323 DOI: 10.3389/fnins.2018.00644] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/29/2018] [Indexed: 01/17/2023] Open
Abstract
Glycine-histidine-lysine (GHK) is a human tripeptide that enhances wound healing, exerts neuroprotective effects against neurodegenerative disease, and improves tissue regeneration. This study examined whether GHK can alleviate injury due to intracerebral hemorrhage (ICH). Briefly, adult Wistar rats in GHK pretreatment groups were injected with GHK (1 or 10 mg/kg, i.p.) every 24 h for 3 days. Water content and intact neurons were detected in the rats 3 days after ICH, and the neurological deficit scores were examined in the rats at 4, 24, 72, and 168 h after ICH. Apoptosis was evaluated via caspase-3 immunohistochemistry, Nissl staining, and TUNEL assay. We also examined the effect of GHK on the expression of related proteins in SH-SY5Y cells via Western blotting. The expression of miR-339-5p was examined via real-time polymerase chain reaction analyses. GHK improved neurological deficits, reduced water content in the brain and inhibited neuronal apoptosis in ICH rats. It also prevented the apoptosis of SH-SY5Y cells with hemin treatment. Furthermore, GHK downregulated miR-339-5p expression, and overexpression of miR-339-5p partially reversed the anti-apoptotic effects of GHK in SH-SY5Y cells. Our findings suggest that the p38 MAPK pathway is involved in the GHK-induced downregulation of miR-339-5p, and that the miR-339-5p/VEGFA axis plays a role in preventing neuronal apoptosis following ICH injury. These findings indicate that GHK may represent a novel therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Heyu Zhang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Yanzhe Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| |
Collapse
|
33
|
Levy MJF, Boulle F, Steinbusch HW, van den Hove DLA, Kenis G, Lanfumey L. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology (Berl) 2018; 235:2195-2220. [PMID: 29961124 PMCID: PMC6061771 DOI: 10.1007/s00213-018-4950-4] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023]
Abstract
Depression is a major health problem with a high prevalence and a heavy socioeconomic burden in western societies. It is associated with atrophy and impaired functioning of cortico-limbic regions involved in mood and emotion regulation. It has been suggested that alterations in neurotrophins underlie impaired neuroplasticity, which may be causally related to the development and course of depression. Accordingly, mounting evidence suggests that antidepressant treatment may exert its beneficial effects by enhancing trophic signaling on neuronal and synaptic plasticity. However, current antidepressants still show a delayed onset of action, as well as lack of efficacy. Hence, a deeper understanding of the molecular and cellular mechanisms involved in the pathophysiology of depression, as well as in the action of antidepressants, might provide further insight to drive the development of novel fast-acting and more effective therapies. Here, we summarize the current literature on the involvement of neurotrophic factors in the pathophysiology and treatment of depression. Further, we advocate that future development of antidepressants should be based on the neurotrophin theory.
Collapse
Affiliation(s)
- Marion J F Levy
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Fabien Boulle
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Harry W Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Daniël L A van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Laurence Lanfumey
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France.
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands.
| |
Collapse
|
34
|
S M, T P, Goli D. Effect of wedelolactone and gallic acid on quinolinic acid-induced neurotoxicity and impaired motor function: significance to sporadic amyotrophic lateral sclerosis. Neurotoxicology 2018; 68:1-12. [PMID: 29981346 DOI: 10.1016/j.neuro.2018.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022]
Abstract
Quinolinic acid (QUIN) is a well-known neuroactive metabolite of tryptophan degradation pathway or kynurenine pathway. The QUIN is involved in the development of several toxic cascades which leads to the neuronal degeneration processes. The QUIN-induced toxicity is also responsible for the impairment of the motor function and motor learning ability. This study seeks to investigate the several mechanisms which are involved in the intrastriatal administration of QUIN-induced neurodegeneration and the neuroprotective effects of wedelolactone (WL) and gallic acid (GA) over QUIN-induced toxicity. The Wistar rats were used for the study and conducted behavioral model to evaluate the effects of WL (100 & 200 mg/kg) and GA (100 & 200 mg/kg) on impaired motor function and motor learning ability. We also assessed the effects of WL and GA on the antioxidant profile, cytotoxicity, apoptosis, excitotoxicity, inflammatory cascades, and on growth factors which helps in neurogenesis. The compounds effectively improved the motor function, motor learning memory in the rats. Similarly, enhanced the activity of Glutathione peroxidase, SOD, catalase, and declined the lipid peroxidation and nitrite production in the brain. The treatment with WL and GA lowered the activities of LDH, m-calpain, and caspase-3. The reports strongly support that both compounds are useful in the prevention of glutamate excitotoxicity induced by QUIN. The NAA, IGF-1, and VEGF levels in the brain were improved after treatment with WL and GA. The neuroprotective effects of WL and GA further proved through the anti-inflammatory effects. The compounds significantly down-regulated the expression of TNF-α, IL-6, and IL-β in the brain. Immunohistochemical analysis shows that the WL and GA reduced the expression of NF-κB. The histopathological studies for cerebellum, hippocampus, striatum, and spinal cord confirms the toxic effects of QUIN and neuroprotective effects of WL and GA. The results suggest that WL and GA could ameliorate the toxic events triggered by QUIN and might be effective in the prevention and progression of several cascades which lead to the development of sALS.
Collapse
Affiliation(s)
- Maya S
- Department of Pharmacology, Acharya & B.M. Reddy College of Pharmacy, Bangalore 560107, Karnataka, India.
| | - Prakash T
- Department of Pharmacology, Acharya & B.M. Reddy College of Pharmacy, Bangalore 560107, Karnataka, India.
| | - Divakar Goli
- Department of Pharmacology, Acharya & B.M. Reddy College of Pharmacy, Bangalore 560107, Karnataka, India
| |
Collapse
|
35
|
Geiseler SJ, Morland C. The Janus Face of VEGF in Stroke. Int J Mol Sci 2018; 19:ijms19051362. [PMID: 29734653 PMCID: PMC5983623 DOI: 10.3390/ijms19051362] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 12/12/2022] Open
Abstract
The family of vascular endothelial growth factors (VEGFs) are known for their regulation of vascularization. In the brain, VEGFs are important regulators of angiogenesis, neuroprotection and neurogenesis. Dysregulation of VEGFs is involved in a large number of neurodegenerative diseases and acute neurological insults, including stroke. Stroke is the main cause of acquired disabilities, and normally results from an occlusion of a cerebral artery or a hemorrhage, both leading to focal ischemia. Neurons in the ischemic core rapidly undergo necrosis. Cells in the penumbra are exposed to ischemia, but may be rescued if adequate perfusion is restored in time. The neuroprotective and angiogenic effects of VEGFs would theoretically make VEGFs ideal candidates for drug therapy in stroke. However, contradictory to what one might expect, endogenously upregulated levels of VEGF as well as the administration of exogenous VEGF is detrimental in acute stroke. This is probably due to VEGF-mediated blood–brain-barrier breakdown and vascular leakage, leading to edema and increased intracranial pressure as well as neuroinflammation. The key to understanding this Janus face of VEGF function in stroke may lie in the timing; the harmful effect of VEGFs on vessel integrity is transient, as both VEGF preconditioning and increased VEGF after the acute phase has a neuroprotective effect. The present review discusses the multifaceted action of VEGFs in stroke prevention and therapy.
Collapse
Affiliation(s)
- Samuel J Geiseler
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
| | - Cecilie Morland
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
- Institute for Behavioral Sciences, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, 0166 Oslo, Norway.
| |
Collapse
|
36
|
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: 0.5] [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
|
37
|
Ligandomics: a paradigm shift in biological drug discovery. Drug Discov Today 2018; 23:636-643. [PMID: 29326083 DOI: 10.1016/j.drudis.2018.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/10/2017] [Accepted: 01/04/2018] [Indexed: 02/06/2023]
Abstract
As productivity of pharmaceutical research and development (R&D) for small-molecule drugs declines, the trend in drug discovery strategies is shifting towards biologics, which predominantly target secreted or cell surface proteins. Receptors and ligands are the most-valuable drug targets. In contrast to conventional approaches of discovering one ligand at a time, the emerging technology of ligandomics can systematically map disease-selective cellular ligands in the absence of molecular probes. Biologics targeting these ligands with disease selectivity have the advantages of high efficacy, minimal adverse effects, wide therapeutic indices, and low safety-related attrition rates. Therefore, ligandomics represents a paradigm shift to address the bottleneck of target discovery for biologics development.
Collapse
|
38
|
Abstract
Vascular endothelial growth factor (VEGF) is a potent angiogenic factor. Despite upregulation of VEGF in the brain in Alzheimer's disease (AD), probably in response to amyloid-β, vasoconstriction, and tissue hypoxia, there is no consequent increase in microvessel density. VEGF binds to and activates VEGF receptor 2 (VEGFR2), but also binds to VEGF receptor 1 (VEGFR1), which exists in less-active membrane-bound and inactive soluble (sVEGFR1) forms and inhibits pro-angiogenic signaling. We have investigated whether altered expression of VEGF receptors might account for the lack of angiogenic response to VEGF in AD. We assessed the cellular distribution and protein level of VEGFR1 and VEGFR2 in parietal cortex from 50 AD and 36 age-matched control brains, and related the findings to measurements of VEGF and von Willebrand factor level (a marker of microvessel density) in the same tissue samples. VEGFR2 was expressed by neurons, astrocytes and endothelial cells. VEGFR1 was expressed predominantly neuronally and was significantly reduced in AD (p = 0.02). Western blot analysis on a subset of brains showed reduction in VEGFR1:sVEGFR1 in AD (p = 0.046). The lack of angiogenesis despite cerebral hypoperfusion in AD is not explained by altered expression of VEGFR2 or total VEGFR1; indeed, the downregulation of VEGFR1 may represent a pro-angiogenic response to the hypoperfusion. However, the relative increase in sVEGFR1 would be expected to have an anti-angiogenic effect which may be a factor in AD.
Collapse
Affiliation(s)
- Rachel Harris
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - James Scott Miners
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - Shelley Allen
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - Seth Love
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| |
Collapse
|
39
|
Wu KW, Mo JL, Kou ZW, Liu Q, Lv LL, Lei Y, Sun FY. Neurovascular Interaction Promotes the Morphological and Functional Maturation of Cortical Neurons. Front Cell Neurosci 2017; 11:290. [PMID: 28966577 PMCID: PMC5605567 DOI: 10.3389/fncel.2017.00290] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/04/2017] [Indexed: 01/19/2023] Open
Abstract
Brain microvascular endothelial cells (BMEC) have been found to guide the migration, promote the survival and regulate the differentiation of neural cells. However, whether BMEC promote development and maturation of immature neurons is still unknown. Therefore, in this study, we used a direct endothelium-neuron co-culture system combined with patch clamp recordings and confocal imaging analysis, to investigate the effects of endothelial cells on neuronal morphology and function during development. We found that endothelial cells co-culture or BMEC-conditioned medium (B-CM) promoted neurite outgrowth and spine formation, accelerated electrophysiological development and enhanced synapse function. Moreover, B-CM treatment induced vascular endothelial growth factor (VEGF) expression and p38 phosphorylation in the cortical neurons. Through pharmacological analysis, we found that incubation with SU1498, an inhibitor of VEGF receptor, abolished B-CM-induced p-p38 upregulation and suppressed the enhancement of synapse formation and transmission. SB203580, an inhibitor of p38 MAPK also blocked B-CM-mediated synaptic regulation. Together these results clearly reveal that the endothelium-neuron interactions promote morphological and functional maturation of neurons. In addition, neurovascular interaction-mediated promotion of neural network maturation relies on activation of VEGF/Flk-1/p38 MAPK signaling. This study provides novel aspects of endothelium-neuron interactions and novel mechanism of neurovascular crosstalk.
Collapse
Affiliation(s)
- Kun-Wei Wu
- Institute of Biomedical Sciences and Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Jia-Lin Mo
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Zeng-Wei Kou
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Qi Liu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Ling-Ling Lv
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Yu Lei
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Feng-Yan Sun
- Institute of Biomedical Sciences and Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| |
Collapse
|
40
|
Silva-Hucha S, Hernández RG, Benítez-Temiño B, Pastor ÁM, de la Cruz RR, Morcuende S. Extraocular motoneurons of the adult rat show higher levels of vascular endothelial growth factor and its receptor Flk-1 than other cranial motoneurons. PLoS One 2017; 12:e0178616. [PMID: 28570669 PMCID: PMC5453543 DOI: 10.1371/journal.pone.0178616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/16/2017] [Indexed: 12/23/2022] Open
Abstract
Recent studies show a relationship between the deficit of vascular endothelial growth factor (VEGF) and motoneuronal degeneration, such as that occurring in amyotrophic lateral sclerosis (ALS). VEGF delivery protects motoneurons from cell death and delayed neurodegeneration in animal models of ALS. Strikingly, extraocular motoneurons show lesser vulnerability to neurodegeneration in ALS compared to other cranial or spinal motoneurons. Therefore, the present study investigates possible differences in VEGF and its main receptor VEGFR-2 or Flk-1 between extraocular and non-extraocular brainstem motoneurons. We performed immunohistochemistry and Western blot to determine the presence of VEGF and Flk-1 in rat motoneurons located in the three extraocular motor nuclei (abducens, trochlear and oculomotor) and to compare it to that observed in two other brainstem nuclei (hypoglossal and facial) that are vulnerable to degeneration. Extraocular motoneurons presented higher amounts of VEGF and its receptor Flk-1 than other brainstem motoneurons, and thus these molecules could be participating in their higher resistance to neurodegeneration. In conclusion, we hypothesize that differences in VEGF availability and signaling could be a contributing factor to the different susceptibility of extraocular motoneurons, when compared with other motoneurons, in neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Silva-Hucha
- Laboratorio de Fisiología y Plasticidad Neuronal, Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Rosendo G. Hernández
- Laboratorio de Fisiología y Plasticidad Neuronal, Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Beatriz Benítez-Temiño
- Laboratorio de Fisiología y Plasticidad Neuronal, Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Ángel M. Pastor
- Laboratorio de Fisiología y Plasticidad Neuronal, Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Rosa R. de la Cruz
- Laboratorio de Fisiología y Plasticidad Neuronal, Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Sara Morcuende
- Laboratorio de Fisiología y Plasticidad Neuronal, Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
- * E-mail:
| |
Collapse
|
41
|
Wang W, LeBlanc ME, Chen X, Chen P, Ji Y, Brewer M, Tian H, Spring SR, Webster KA, Li W. Pathogenic role and therapeutic potential of pleiotrophin in mouse models of ocular vascular disease. Angiogenesis 2017; 20:479-492. [PMID: 28447229 DOI: 10.1007/s10456-017-9557-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/18/2017] [Indexed: 01/06/2023]
Abstract
Angiogenic factors play an important role in the pathogenesis of diabetic retinopathy (DR), neovascular age-related macular degeneration (nAMD) and retinopathy of prematurity (ROP). Pleiotrophin, a well-known angiogenic factor, was recently reported to be upregulated in the vitreous fluid of patients with proliferative DR (PDR). However, its pathogenic role and therapeutic potential in ocular vascular diseases have not been defined in vivo. Here using corneal pocket assays, we demonstrated that pleiotrophin induced angiogenesis in vivo. To investigate the pathological role of pleiotrophin we used neutralizing antibody to block its function in multiple in vivo models of ocular vascular diseases. In a mouse model of DR, intravitreal injection of pleiotrophin-neutralizing antibody alleviated diabetic retinal vascular leakage. In a mouse model of oxygen-induced retinopathy (OIR), which is a surrogate model of ROP and PDR, we demonstrated that intravitreal injection of anti-pleiotrophin antibody prevented OIR-induced pathological retinal neovascularization and aberrant vessel tufts. Finally, pleiotrophin-neutralizing antibody ameliorated laser-induced choroidal neovascularization, a mouse model of nAMD, suggesting that pleiotrophin is involved in choroidal vascular disease. These findings suggest that pleiotrophin plays an important role in the pathogenesis of DR with retinal vascular leakage, ROP with retinal neovascularization and nAMD with choroidal neovascularization. The results also support pleiotrophin as a promising target for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Weiwen Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Michelle E LeBlanc
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Xiuping Chen
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,Department of Ophthalmology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Ping Chen
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,Department of Ophthalmology, Renji Hospital of Jiaotong University, Shanghai, China
| | - Yanli Ji
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,Department of Ophthalmology, Zhengzhou Eye Hospital, Zhengzhou, Henan, China
| | - Megan Brewer
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Hong Tian
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,School of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Samantha R Spring
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Keith A Webster
- Vascular Biology Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Wei Li
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA. .,Vascular Biology Institute, University of Miami School of Medicine, Miami, FL, USA.
| |
Collapse
|
42
|
Valentin-Kahan A, García-Tejedor GB, Robello C, Trujillo-Cenóz O, Russo RE, Alvarez-Valin F. Gene Expression Profiling in the Injured Spinal Cord of Trachemys scripta elegans: An Amniote with Self-Repair Capabilities. Front Mol Neurosci 2017; 10:17. [PMID: 28223917 PMCID: PMC5293771 DOI: 10.3389/fnmol.2017.00017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
Slider turtles are the only known amniotes with self-repair mechanisms of the spinal cord that lead to substantial functional recovery. Their strategic phylogenetic position makes them a relevant model to investigate the peculiar genetic programs that allow anatomical reconnection in some vertebrate groups but are absent in others. Here, we analyze the gene expression profile of the response to spinal cord injury (SCI) in the turtle Trachemys scripta elegans. We found that this response comprises more than 1000 genes affecting diverse functions: reaction to ischemic insult, extracellular matrix re-organization, cell proliferation and death, immune response, and inflammation. Genes related to synapses and cholesterol biosynthesis are down-regulated. The analysis of the evolutionary distribution of these genes shows that almost all are present in most vertebrates. Additionally, we failed to find genes that were exclusive of regenerating taxa. The comparison of expression patterns among species shows that the response to SCI in the turtle is more similar to that of mice and non-regenerative Xenopus than to Xenopus during its regenerative stage. This observation, along with the lack of conserved “regeneration genes” and the current accepted phylogenetic placement of turtles (sister group of crocodilians and birds), indicates that the ability of spinal cord self-repair of turtles does not represent the retention of an ancestral vertebrate character. Instead, our results suggest that turtles developed this capability from a non-regenerative ancestor (i.e., a lineage specific innovation) that was achieved by re-organizing gene expression patterns on an essentially non-regenerative genetic background. Among the genes activated by SCI exclusively in turtles, those related to anoxia tolerance, extracellular matrix remodeling, and axonal regrowth are good candidates to underlie functional recovery.
Collapse
Affiliation(s)
- Adrián Valentin-Kahan
- Department of Cellular and Molecular Neurophysiology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Gabriela B García-Tejedor
- Department of Cellular and Molecular Neurophysiology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Carlos Robello
- Molecular Biology Unit, Institut Pasteur de MontevideoMontevideo, Uruguay; Departamento de Bioquímica, Facultad de Medicina, Universidad de la RepublicaMontevideo, Uruguay
| | - Omar Trujillo-Cenóz
- Department of Cellular and Molecular Neurophysiology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Raúl E Russo
- Department of Cellular and Molecular Neurophysiology, Instituto de Investigaciones Biológicas Clemente Estable Montevideo, Uruguay
| | - Fernando Alvarez-Valin
- Sección Biomatemática, Unidad de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República Montevideo, Uruguay
| |
Collapse
|
43
|
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: 10] [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
|
44
|
Dexamethasone prevents motor deficits and neurovascular damage produced by shiga toxin 2 and lipopolysaccharide in the mouse striatum. Neuroscience 2016; 344:25-38. [PMID: 28042026 DOI: 10.1016/j.neuroscience.2016.12.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 12/31/2022]
Abstract
Shiga toxin 2 (Stx2) from enterohemorrhagic Escherichia coli (EHEC) causes bloody diarrhea and Hemolytic Uremic Syndrome (HUS) that may derive to fatal neurological outcomes. Neurological abnormalities in the striatum are frequently observed in affected patients and in studies with animal models while motor disorders are usually associated with pyramidal and extra pyramidal systems. A translational murine model of encephalopathy was employed to demonstrate that systemic administration of a sublethal dose of Stx2 damaged the striatal microvasculature and astrocytes, increase the blood brain barrier permeability and caused neuronal degeneration. All these events were aggravated by lipopolysaccharide (LPS). The injury observed in the striatum coincided with locomotor behavioral alterations. The anti-inflammatory Dexamethasone resulted to prevent the observed neurologic and clinical signs, proving to be an effective drug. Therefore, the present work demonstrates that: (i) systemic sub-lethal Stx2 damages the striatal neurovascular unit as it succeeds to pass through the blood brain barrier. (ii) This damage is aggravated by the contribution of LPS which is also produced and secreted by EHEC, and (iii) the observed neurological alterations may be prevented by an anti-inflammatory treatment.
Collapse
|
45
|
Park HW, Jeon HJ, Chang MS. Vascular endothelial growth factor enhances axonal outgrowth in organotypic spinal cord slices via vascular endothelial growth factor receptor 1 and 2. Tissue Eng Regen Med 2016; 13:601-609. [PMID: 30603441 DOI: 10.1007/s13770-016-0051-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/13/2016] [Accepted: 06/01/2016] [Indexed: 01/26/2023] Open
Abstract
Enhancing adult nerve regeneration is a potential therapeutic strategy for treating spinal cord injury. Vascular endothelial growth factor (VEGF) is a major contributor to angiogenesis, which can reduce the spinal cord injury by inhibiting the inflammation and improve recovery after spinal cord injury. We have previously demonstrated that exogenous VEGF has neurotrophic effects on injured spinal nerves in organotypic spinal cord slice cultures. However, the mechanisms underlying the neurite growth by exogenous VEGF remain to be explored in spinal cord. In this study, we found out that exogenous VEGF mediated axonal outgrowth through VEGF receptor 1 (VEGFR1) and VEGFR2, both of which were expressed on organotypic spinal cord slices. Although VEGFR1 and VEGFR2 were constitutively expressed in some cells of control spinal cord slices, VEGF treatment upregulated expression of VEGFR1 and VEGFR2. Both VEGFR1 and VEGFR2 were expressed in neuronal cells as well as glial cells of organotypic spinal cord slices. We also observed that VEGF-induced axonal outgrowth was attenuated by a specific mitogen-activated protein kinase (MAPK) inhibitor PD98059 and a specific phosphoinositide 3-kinase (PI3K) inhibitor wortmannin. Thus, these findings suggest that these MAPK and PI3K pathways have important roles in regulating VEGF-induced axonal outgrowth in the postnatal spinal cord.
Collapse
Affiliation(s)
- Hwan-Woo Park
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 2Department of Cell Biology, College of Medicine, Konyang University, Daejeon, Korea
| | - Hyo-Jin Jeon
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Mi-Sook Chang
- 1Laboratory of Stem Cell & Neurobiology, Department of Oral Anatomy, Dental Research Institute & School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 3Neuroscience Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
46
|
Gruber HE, Hoelscher GL, Bullock L, Ingram JA, Norton HJ, Hanley EN. Human annulus signaling cues for nerve outgrowth: In vitro studies. J Orthop Res 2016; 34:1456-65. [PMID: 27155444 DOI: 10.1002/jor.23286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/06/2016] [Indexed: 02/04/2023]
Abstract
The relationship between neurotrophins produced by human annulus cells, such as neurotrophin-4 (NT4) and brain-derived neurotrophic factor (BDNF) which function in neurite survival and outgrowth, and nerve ingrowth into the disc remains poorly understood. In this work, we tested F11 neurite growth during exposure to control media, media with added nerve growth factor (NGF), conditioned media (CM) harvested from previous human annulus culture, or co-culture with annulus cells. Co-culture of F11 cells with annulus cells significantly increased media levels of amphiregulin, BDNF, glial-derived neurotrophic factor, and vascular endothelial growth factor compared to levels from in culture of F11 cells alone (p ≤ 0.04). Cell-based assays of neurite growth revealed that BDNF levels present in CM bore a significant (p = 0.01) positive relationship to neurite length and accounted for 38.5% of the change in neurite length. NT4 levels produced during co-culture with annulus cells bore a significant (p = 0.04) positive relationship to neurite length and accounted for 40.9% of the change in length. Statement of clinical significance: In vitro findings point to a potential role of annulus cells related to nerve ingrowth in vivo, and may have relevance in the outer annulus (where cell numbers are high) or in regions where nerves penetrate into annular tears or fissures. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1456-1465, 2016.
Collapse
Affiliation(s)
- Helen E Gruber
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, North Carolina, 28232
| | - Gretchen L Hoelscher
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, North Carolina, 28232
| | - Letitia Bullock
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, North Carolina, 28232
| | - Jane A Ingram
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, North Carolina, 28232
| | - H James Norton
- Dickson Advanced Analytics, Carolinas Medical Center, PO Box 32861, Charlotte, North Carolina
| | - Edward N Hanley
- Department of Orthopaedic Surgery, Carolinas Medical Center, PO Box 32861, Charlotte, North Carolina, 28232
| |
Collapse
|
47
|
Mateos L, Perez-Alvarez MJ, Wandosell F. Angiotensin II type-2 receptor stimulation induces neuronal VEGF synthesis after cerebral ischemia. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1297-308. [DOI: 10.1016/j.bbadis.2016.03.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/04/2016] [Accepted: 03/30/2016] [Indexed: 10/22/2022]
|
48
|
Association of serum VEGF levels with prefrontal cortex volume in schizophrenia. Mol Psychiatry 2016; 21:686-92. [PMID: 26169975 DOI: 10.1038/mp.2015.96] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/05/2015] [Accepted: 06/01/2015] [Indexed: 12/30/2022]
Abstract
A large body of evidence indicates alterations in brain regional cellular energy metabolism and blood flow in schizophrenia. Among the different molecules regulating blood flow, vascular endothelial growth factor (VEGF) is generally accepted as the major factor involved in the process of angiogenesis. In the present study, we examined whether peripheral VEGF levels correlate with changes in the prefrontal cortex (PFC) volume in patients with schizophrenia and in healthy controls. Whole-blood samples were obtained from 96 people with schizophrenia or schizoaffective disorder and 83 healthy controls. Serum VEGF protein levels were analyzed by enzyme-linked immunosorbent assay, whereas quantitative PCR was performed to measure interleukin-6 (IL-6, a pro-inflammatory marker implicated in schizophrenia) mRNA levels in the blood samples. Structural magnetic resonance imaging scans were obtained using a 3T Achieva scanner on a subset of 59 people with schizophrenia or schizoaffective disorder and 65 healthy controls, and prefrontal volumes were obtained using FreeSurfer software. As compared with healthy controls, individuals with schizophrenia had a significant increase in log-transformed mean serum VEGF levels (t(177)=2.9, P=0.005). A significant inverse correlation (r=-0.40, P=0.002) between serum VEGF and total frontal pole volume was found in patients with schizophrenia/schizoaffective disorder. Moreover, we observed a significant positive association (r=0.24, P=0.03) between serum VEGF and IL-6 mRNA levels in patients with schizophrenia. These findings suggest an association between serum VEGF and inflammation, and that serum VEGF levels are related to structural abnormalities in the PFC of people with schizophrenia.
Collapse
|
49
|
Ke X, Liu C, Wang Y, Ma J, Mao X, Li Q. Netrin-1 promotes mesenchymal stem cell revascularization of limb ischaemia. Diab Vasc Dis Res 2016; 13:145-56. [PMID: 26818229 DOI: 10.1177/1479164115611594] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study examines the effect and mechanism of action of Netrin-1 on bone marrow mesenchymal stem cells in angiogenesis. Tube formation and migration of bone marrow mesenchymal stem cells were observed in cell culture. Bone marrow mesenchymal stem cells or Netrin-1-bone marrow mesenchymal stem cells were injected into the ischaemic area of the rat hind limb on the first day after surgery. Laser Doppler perfusion imaging was performed to analyse the levels of vascular endothelial growth factor in plasma and muscles, and immunohistochemistry and immunofluorescence were used to analyse angiogenesis. Bone marrow mesenchymal stem cells in medium containing Netrin-1 markedly increased the number of tubes formed and the migration of bone marrow mesenchymal stem cells compared with the untreated control group. The function of Netrin-1 in tube formation and migration is similar to vascular endothelial growth factor, and combined with vascular endothelial growth factor, Netrin-1 has more enhanced effect than in the other three groups. The Netrin-1-bone marrow mesenchymal stem cell group had better augmented blood-perfusion scores and vessel densities, as well as improved function of the ischaemic limb than that of the group injected with bone marrow mesenchymal stem cells (treated with bone marrow mesenchymal stem cells individually) or the control group (treated with medium). These results suggest that Netrin-1 has the ability to augment the angiogenesis of bone marrow mesenchymal stem cells and improve the function of the ischaemic hind limb by increasing the level of vascular endothelial growth factor.
Collapse
Affiliation(s)
- Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chenxiao Liu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoming Mao
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
50
|
Choi SH, Ruggiero D, Sorice R, Song C, Nutile T, Vernon Smith A, Concas MP, Traglia M, Barbieri C, Ndiaye NC, Stathopoulou MG, Lagou V, Maestrale GB, Sala C, Debette S, Kovacs P, Lind L, Lamont J, Fitzgerald P, Tönjes A, Gudnason V, Toniolo D, Pirastu M, Bellenguez C, Vasan RS, Ingelsson E, Leutenegger AL, Johnson AD, DeStefano AL, Visvikis-Siest S, Seshadri S, Ciullo M. Six Novel Loci Associated with Circulating VEGF Levels Identified by a Meta-analysis of Genome-Wide Association Studies. PLoS Genet 2016; 12:e1005874. [PMID: 26910538 PMCID: PMC4766012 DOI: 10.1371/journal.pgen.1005874] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/26/2016] [Indexed: 12/31/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic and neurotrophic factor, secreted by endothelial cells, known to impact various physiological and disease processes from cancer to cardiovascular disease and to be pharmacologically modifiable. We sought to identify novel loci associated with circulating VEGF levels through a genome-wide association meta-analysis combining data from European-ancestry individuals and using a dense variant map from 1000 genomes imputation panel. Six discovery cohorts including 13,312 samples were analyzed, followed by in-silico and de-novo replication studies including an additional 2,800 individuals. A total of 10 genome-wide significant variants were identified at 7 loci. Four were novel loci (5q14.3, 10q21.3, 16q24.2 and 18q22.3) and the leading variants at these loci were rs114694170 (MEF2C, P = 6.79x10-13), rs74506613 (JMJD1C, P = 1.17x10-19), rs4782371 (ZFPM1, P = 1.59x10-9) and rs2639990 (ZADH2, P = 1.72x10-8), respectively. We also identified two new independent variants (rs34528081, VEGFA, P = 1.52x10-18; rs7043199, VLDLR-AS1, P = 5.12x10-14) at the 3 previously identified loci and strengthened the evidence for the four previously identified SNPs (rs6921438, LOC100132354, P = 7.39x10-1467; rs1740073, C6orf223, P = 2.34x10-17; rs6993770, ZFPM2, P = 2.44x10-60; rs2375981, KCNV2, P = 1.48x10-100). These variants collectively explained up to 52% of the VEGF phenotypic variance. We explored biological links between genes in the associated loci using Ingenuity Pathway Analysis that emphasized their roles in embryonic development and function. Gene set enrichment analysis identified the ERK5 pathway as enriched in genes containing VEGF associated variants. eQTL analysis showed, in three of the identified regions, variants acting as both cis and trans eQTLs for multiple genes. Most of these genes, as well as some of those in the associated loci, were involved in platelet biogenesis and functionality, suggesting the importance of this process in regulation of VEGF levels. This work also provided new insights into the involvement of genes implicated in various angiogenesis related pathologies in determining circulating VEGF levels. The understanding of the molecular mechanisms by which the identified genes affect circulating VEGF levels could be important in the development of novel VEGF-related therapies for such diseases. Vascular Endothelial Growth Factor (VEGF) is a protein with a fundamental role in development of vascular system. The protein, produced by many types of cells, is released in the blood. High levels of VEGF have been observed in different pathological conditions especially in cancer, cardiovascular, and inflammatory diseases. Therefore, identifying the genetic factors influencing VEGF levels is important for predicting and treating such pathologies. The number of genetic variants associated with VEGF levels has been limited. To identify new loci, we have performed a Genome Wide Association Study meta-analysis on a sample of more than 16,000 individuals from 10 cohorts, using a high-density genetic map. This analysis revealed 10 variants associated with VEGF circulating levels, 6 of these being novel associations. The 10 variants cumulatively explain more than 50% of the variability of VEGF serum levels. Our analyses have identified genes known to be involved in angiogenesis related diseases and genes implicated in platelet metabolism, suggesting the importance of links between this process and VEGF regulation. Overall, these data have improved our understanding of the genetic variation underlying circulating VEGF levels. This in turn could guide our response to the challenge posed by various VEGF-related pathologies.
Collapse
Affiliation(s)
- Seung Hoan Choi
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Rossella Sorice
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Ci Song
- Population Sciences Branch, National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Teresa Nutile
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Maria Pina Concas
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Caterina Barbieri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Ndeye Coumba Ndiaye
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Maria G. Stathopoulou
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Stephanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
- INSERM U897, Bordeaux, France
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - John Lamont
- Randox Laboratories, Crumlin, United Kingdom
| | | | - Anke Tönjes
- University of Leipzig, Department of Medicine, Leipzig, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Celine Bellenguez
- Institut Pasteur de Lille, Lille, France
- INSEM U744, Lille, France
- Université Lille-Nord de France, Lille, France
| | - Ramachandran S. Vasan
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, United States of America
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anne-Louise Leutenegger
- INSERM U946, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, IUH, UMR-S 946, Paris, France
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Anita L. DeStefano
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Sophie Visvikis-Siest
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- * E-mail: (SS); (MC)
| | - Marina Ciullo
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
- * E-mail: (SS); (MC)
| |
Collapse
|