1
|
Bukatova S, Bacova Z, Osacka J, Bakos J. Mini review of molecules involved in altered postnatal neurogenesis in autism. Int J Neurosci 2024; 134:1429-1443. [PMID: 37815399 DOI: 10.1080/00207454.2023.2269304] [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: 12/15/2022] [Revised: 04/06/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023]
Abstract
The neurobiology of autism is complex, but emerging research points to potential abnormalities and alterations in neurogenesis. The aim of the present review is to describe the advances in the understanding of the role of selected neurotrophins, neuropeptides, and other compounds secreted by neuronal cells in the processes of postnatal neurogenesis in conjunction with autism. We characterize the fundamental mechanisms of neuronal cell proliferation, generation of major neuronal cell types with special emphasis on neurogenic niches - the subventricular zone and hippocampal areas. We also discuss changes in intracellular calcium levels and calcium-dependent transcription factors in the context of the regulation of neurogenesis and cell fate determination. To sum up, this review provides specific insight into the known association between alterations in the function of the entire spectrum of molecules involved in neurogenesis and the etiology of autism pathogenesis.
Collapse
Affiliation(s)
- Stanislava Bukatova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jana Osacka
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
2
|
Goksu AY, Kocanci FG, Akinci E, Demir-Dora D, Erendor F, Sanlioglu S, Uysal H. Microglia cells treated with synthetic vasoactive intestinal peptide or transduced with LentiVIP protect neuronal cells against degeneration. Eur J Neurosci 2024; 59:1993-2015. [PMID: 38382910 DOI: 10.1111/ejn.16273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 02/23/2024]
Abstract
A common pathological hallmark of neurodegenerative disorders is neuronal cell death, accompanied by neuroinflammation and oxidative stress. The vasoactive intestinal peptide (VIP) is a pleiotropic peptide that combines neuroprotective and immunomodulatory actions. The gene therapy field shows long-term promise for treating a wide range of neurodegenerative diseases (ND). In this study, we aimed to investigate the in vitro efficacy of transduction of microglia using lentiviral gene therapy vectors encoding VIP (LentiVIP). Additionally, we tested the protective effects of the secretome derived from LentiVIP-infected "immortalized human" microglia HMC3 cells, and cells treated with Synthetic VIP (SynVIP), against toxin-induced neurodegeneration. First, LentiVIP, which stably expresses VIP, was generated and purified. VIP secretion in microglial conditioned media (MG CM) for LentiVIP-infected HMC3 microglia cells was confirmed. Microglia cells were activated with lipopolysaccharide, and groups were formed as follows: 1) Control, 2) SynVIP-treated, or 3) LentiVIP-transduced. These MG CM were applied on an in vitro neurodegenerative model formed by differentiated (d)-SH-SY5Y cells. Then, cell survival analysis and apoptotic nuclear staining, besides measurement of oxidative/inflammatory parameters in CM of cells were performed. Activated MG CM reduced survival rates of both control and toxin-applied (d)-SH-SY5Y cells, whereas LentiVIP-infected MG CM and SynVIP-treated ones exhibited better survival rates. These findings were supported by apoptotic nuclear evaluations of (d)-SH-SY5Y cells, alongside oxidative/inflammatory parameters in their CM. LentiVIP seems worthy of further studies for the treatment of ND because of the potential of gene therapy to treat diseases effectively with a single injection.
Collapse
Affiliation(s)
- Azize Yasemin Goksu
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Fatma Gonca Kocanci
- Department of Medical Laboratory Techniques, Vocational High School of Health Services, Alanya Alaaddin Keykubat University, Alanya/Antalya, Turkey
| | - Ersin Akinci
- Brigham and Women's Hospital, Division of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Biotechnology, Faculty of Agriculture, Akdeniz University, Antalya, Turkey
| | - Devrim Demir-Dora
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Fulya Erendor
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Salih Sanlioglu
- Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Hilmi Uysal
- Department of Neurology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
3
|
Korkmaz OT, Ay H, Aytan N, Carreras I, Kowall NW, Dedeoglu A, Tuncel N. Vasoactive Intestinal Peptide Decreases β-Amyloid Accumulation and Prevents Brain Atrophy in the 5xFAD Mouse Model of Alzheimer's Disease. J Mol Neurosci 2018; 68:389-396. [PMID: 30498985 DOI: 10.1007/s12031-018-1226-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by extracellular deposits of fibrillary β-amyloid (Aβ) plaques in the brain that initiate an inflammatory process resulting in neurodegeneration. The neuronal loss associated with AD results in gross atrophy of affected regions causing a progressive loss of cognitive ability and memory function, ultimately leading to dementia. Growing evidence suggests that vasoactive intestinal peptide (VIP) could be beneficial for various neurodegenerative diseases, including AD. The study investigated the effects of VIP on 5xFAD, a transgenic mouse model of AD. Toward this aim, we used 20 5xFAD mice in two groups (n = 10 each), VIP-treated (25 ng/kg i.p. injection, three times per week) and saline-treated (the drug's vehicle) following the same administration regimen. Treatment started at 1 month of age and ended 2 months later. After 2 months of treatment, the mice were euthanized, their brains dissected out, and immunohistochemically stained for Aβ40 and Aβ42 on serial sections. Then, plaque analysis and stereological morphometric analysis were performed in different brain regions. Chronic VIP administration in 5xFAD mice significantly decreased the levels of Aβ40 and Aβ42 plaques in the subiculum compared to the saline treated 5xFAD mice. VIP treatment also significantly decreased Aβ40 and Aβ42 plaques in cortical areas and significantly increased the hippocampus/cerebrum and corpus callosum/cerebrum ratio but not the cerebral cortex/cerebrum ratio. In summary, we found that chronic administration of VIP significantly decreased Aβ plaques and preserved against atrophy for related brain regions in 5xFAD AD mice.
Collapse
Affiliation(s)
- Orhan Tansel Korkmaz
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA. .,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA. .,Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26480, Eskisehir, Turkey. .,Faculty of Medicine, Department of Physiology, Eskisehir Osmangazi University, Odunpazari, 26040, Eskisehir, Turkey.
| | - Hakan Ay
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Nurgul Aytan
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Isabel Carreras
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Neil W Kowall
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Alpaslan Dedeoglu
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Radiology, MGH and Harvard Medical School, Boston, MA, 02114, USA
| | - Nese Tuncel
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26480, Eskisehir, Turkey
| |
Collapse
|
4
|
The Impact of T-2 Toxin on Vasoactive Intestinal Polypeptide-Like Immunoreactive (VIP-LI) Nerve Structures in the Wall of the Porcine Stomach and Duodenum. Toxins (Basel) 2018; 10:toxins10040138. [PMID: 29587461 PMCID: PMC5923304 DOI: 10.3390/toxins10040138] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/21/2018] [Accepted: 03/25/2018] [Indexed: 01/29/2023] Open
Abstract
T-2 toxin is a secondary metabolite of some Fusarium species. It is well-known that this substance can harmfully impact living organisms. Among others, thanks to the ability of crossing the blood–brain barrier, T-2 toxin can affect the central nervous system. Mycotoxins mostly get into the organism through the digestive tract; therefore, first of all they have to break the intestinal barrier, wherein the important component is the enteric nervous system (ENS). However, knowledge about the impact of T-2 toxin on the ENS is rather scant. As a result of the influence of various physiological and pathological agents, ENS can undergo adaptive and reparative processes which manifest as changes in the immunoreactivity of perikaryons for neuronal active substances. So, the aim of the present investigation was to study how low doses of T-2 toxin affect vasoactive intestinal polypeptide-like immunoreactive (VIP-LI) nervous structures in the ENS of the porcine stomach and duodenum. Obtained results have shown that T-2 toxin causes an percentage increase of VIP-LI nerve cells and nerve fibers in every enteric plexus in both fragments of gastrointestinal tract studied. This shows that even low doses of T-2 toxin can have an influence on living organisms.
Collapse
|
5
|
Maduna T, Lelievre V. Neuropeptides shaping the central nervous system development: Spatiotemporal actions of VIP and PACAP through complementary signaling pathways. J Neurosci Res 2016; 94:1472-1487. [PMID: 27717098 DOI: 10.1002/jnr.23915] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 01/18/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are neuropeptides with wide, complementary, and overlapping distributions in the central and peripheral nervous systems, where they exert important regulatory roles in many physiological processes. VIP and PACAP display a large range of biological cellular targets and functions in the adult nervous system including regulation of neurotransmission and neuroendocrine secretion and neuroprotective and neuroimmune responses. As the main focus of the present review, VIP and PACAP also have been long implicated in nervous system development and maturation through their interaction with the seven transmembrane domain G protein-coupled receptors, PAC1, VPAC1, and VPAC2, initiating multiple signaling pathways. Compared with PAC1, which solely binds PACAP with very high affinity, VPACs exhibit high affinities for both VIP and PACAP but differ from each other because of their pharmacological profile for both natural accessory peptides and synthetic or chimeric molecules, with agonistic and antagonistic properties. Complementary to initial pharmacological studies, transgenic animals lacking these neuropeptides or their receptors have been used to further characterize the neuroanatomical, electrophysiological, and behavioral roles of PACAP and VIP in the developing central nervous system. In this review, we recapitulate the critical steps and processes guiding/driving neurodevelopment in vertebrates and superimposing the potential contribution of PACAP and VIP receptors on the given timeline. We also describe how alterations in VIP/PACAP signaling may contribute to both (neuro)developmental and adult pathologies and suggest that tuning of VIP/PACAP signaling in a spatiotemporal manner may represent a novel avenue for preventive therapies of neurological and psychiatric disorders. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tando Maduna
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique UPR3212, Université de Strasbourg, Strasbourg, France
| | - Vincent Lelievre
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique UPR3212, Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
6
|
Hauk V, Azzam S, Calo G, Gallino L, Paparini D, Franchi A, Ramhorst R, Leirós CP. Vasoactive Intestinal Peptide Induces an Immunosuppressant Microenvironment in the Maternal-Fetal Interface ofNon-Obese DiabeticMice and Improves Early Pregnancy Outcome. Am J Reprod Immunol 2013; 71:120-30. [DOI: 10.1111/aji.12167] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/18/2013] [Indexed: 12/12/2022] Open
Affiliation(s)
- Vanesa Hauk
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| | - Sofía Azzam
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| | - Guillermina Calo
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| | - Lucila Gallino
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| | - Daniel Paparini
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| | - Ana Franchi
- Facultad de Medicina; Universidad de Buenos Aires; CEFYBO-CONICET; Buenos Aires Argentina
| | - Rosanna Ramhorst
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| | - Claudia Pérez Leirós
- Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; IQUIBICEN-CONICET; Buenos Aires Argentina
| |
Collapse
|
7
|
Cobo M, Anderson P, Benabdellah K, Toscano MG, Muñoz P, García-Pérez A, Gutierrez I, Delgado M, Martin F. Mesenchymal Stem Cells Expressing Vasoactive Intestinal Peptide Ameliorate Symptoms in a Model of Chronic Multiple Sclerosis. Cell Transplant 2013; 22:839-54. [DOI: 10.3727/096368912x657404] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Multiple sclerosis (MS) is a severe debilitating disorder characterized by progressive demyelination and axonal damage of the central nervous system (CNS). Current therapies for MS inhibit the immune response and demonstrate reasonable benefits if applied during the early phase of relapsing–remitting MS (RRMS) while there are no treatments for patients that progress neither to the chronic phase nor for the primary progressive form of the disease. In this manuscript, we have studied the therapeutic efficacy of a cell and gene therapy strategy for the treatment of a mouse model of chronic MS [myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE)]. We used allogenic mesenchymal stem cells (MSCs) as a therapeutic tool and also as vehicle to deliver fully processed 3.3-kDa vasoactive intestinal peptide (VIP) to the peripheral immune organs and to the inflamed CNS. Intraperitoneal administrations of MSCs expressing VIP stopped progression and reduced symptoms when administered at peak of disease. The improvement in clinical score correlated with diminished peripheral T-cell responses against MOG as well as lower inflammation, lower demyelination, and higher neuronal integrity in the CNS. Interestingly, neither lentiviral vectors expressing VIP nor unmodified MSCs were therapeutic when administer at the peak of disease. The increased therapeutic effect of MSCs expressing VIP over unmodified MSCs requires the immunoregulatory and neuroprotective roles of both VIP and MSCs and the ability of the MSCs to migrate to peripheral lymph organs and the inflamed CNS.
Collapse
Affiliation(s)
- Marién Cobo
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| | - Per Anderson
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| | - Karim Benabdellah
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| | - Miguel G. Toscano
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| | - Pilar Muñoz
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| | - Angélica García-Pérez
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| | - Iván Gutierrez
- Biobanco, Parque Tecnológico Salud (PTS), Armilla, Universidad de Granada, Granada, Spain
| | - Mario Delgado
- IPB Lopez Neyra, CSIC, Parque Tecnológico Salud (PTS), Armilla, Granada, Spain
| | - Francisco Martin
- GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Parque Tecnológico Salud (PTS), Granada, Spain
| |
Collapse
|
8
|
Korkmaz O, Ay H, Ulupinar E, Tunçel N. Vasoactive intestinal peptide enhances striatal plasticity and prevents dopaminergic cell loss in Parkinsonian rats. J Mol Neurosci 2012; 48:565-73. [PMID: 22544516 DOI: 10.1007/s12031-012-9781-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/12/2012] [Indexed: 01/18/2023]
Abstract
Destruction of the nigrostriatal dopaminergic pathway by the administration of 6-OHDA generates an animal model of Parkinson's disease. The main characteristic of this progressive neurological disorder is the loss of the dopaminergic neurons located in the substantia nigra pars compacta (SNc). Dopaminergic inputs from the SNc innervate the medium spiny neurons of the striatum and modulate the spontaneous activity of the primary output nuclei of the basal ganglia, globus pallidus interna, and substantia nigra pars reticulata. In our previous studies, we showed that systematically administered vasoactive intestinal peptide (VIP) is effective at reversing motor deficits, decreasing neuronal cell death, and repairing the myelin sheet in parkinsonian rats. In the current study, the effects of VIP on the dendritic morphology of the striatal neurons and the number of dopaminergic neurons in the SNc were examined in 6-OHDA-lesioned rats using Golgi-Cox staining and design-based stereological methods, respectively. Adult Sprague-Dawley rats were separated into sham-operated, bilaterally 6-OHDA lesioned and lesioned + i.p. VIP-injected (25 ng/kg) groups. VIP was first injected 1 h after the intrastriatal 6-OHDA microinjection (every 2 days for 15 days). The 6-OHDA significantly decreased the total number of dopaminergic neurons, branching, and spine density of the medium spiny neurons in the striatum. VIP significantly increased the number of neurons immunostained with tyrosine hydroxylase and the density of spines without altering the branching and the total length of dendrites. In conclusion, VIP might display synaptogenetic activity by enhancing the spine density in the striatum of the parkinsonian rats.
Collapse
Affiliation(s)
- OrhanTansel Korkmaz
- Department of Physiology and Neurophysiology, Eskişehir Osmangazi University, 26040 Eskisehir, Turkey
| | | | | | | |
Collapse
|
9
|
Roca V, Calafat M, Larocca L, Ramhorst R, Farina M, Franchi AM, Pérez Leirós C. Potential immunomodulatory role of VIP in the implantation sites of prediabetic nonobese diabetic mice. Reproduction 2009; 138:733-42. [DOI: 10.1530/rep-09-0171] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Among several factors known to modulate embryo implantation and survival, uterine quiescence and neovascularization, maternal immunotolerance through the Th1/Th2 cytokine balance towards a Th2 profile, local regulatory T-cell (Treg) activation, and high levels of progesterone were assigned a prominent role. Vasoactive intestinal peptide (VIP) is a neuroimmunopeptide that has anti-inflammatory effects, promotes Th2 cytokines and CD4+CD25+FOXP3+Treg activation, and stimulates exocrine secretion, smooth muscle relaxation, and vasodilatation favoring uterus quiescence. The goal of the present work was to explore the participation of VIP in the implantation sites of normal and pregnant prediabetic nonobese diabetic (NOD) females, a mouse strain that spontaneously develops an autoimmune exocrinopathy similar to Sjögren's syndrome. Our results indicate a reduction in litter size from the third parturition onwards in the NOD female lifespan with increased resorption rates. Progesterone systemic levels were significantly decreased in pregnant NOD mice compared with BALB/c mice, although the allogeneic response to progesterone by spleen cells was not impaired. VIP receptors,Vipr1andVipr2(Vpac1andVpac2), were expressed at the implantation sites and VIP induced leukemia inhibitory factor (LIF) and Treg marker expression in both strains; however, a reducedVipexpression was found in NOD implantation sites. We conclude that the reduced birth rate at 16-week-old NOD mice with a Th1 systemic cytokine profile involves resorption processes with a lower expression of VIP at the sites of implantation, which acts as a local inducer of pro-implantatory LIF and Treg activation.
Collapse
|
10
|
Griesmaier E, Keller M. Neuroprotective strategies in excitotoxic brain injury: potential applications to the preterm brain. FUTURE NEUROLOGY 2009. [DOI: 10.2217/fnl.09.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuronal and oligodendroglial cell death owing to increased glutamate levels plays an important role in the pathophysiology of hypoxic-, ischemic- and inflammation-mediated brain injury as well as in disorders such as epilepsy, Alzheimer’s, Parkinson’s or Huntington’s disease. In addition, excitotoxic brain injury is known to be a major contributing factor to brain injury in preterm infants. Excitotoxicity is characterized as excessive glutamatergic activation of postsynaptic receptors that consequently leads to cell injury and cell death. The major excitatory amino acid neurotransmitter is glutamate. Glutamate plays a key role in brain development, affecting progenitor cell differentiation, proliferation, migration and survival. In physiological conditions the presence of glutamate in the synapse is regulated by ATP-dependent glutamate transporters in neurons and glial cells, with astrocytes being responsible for a major part of glutamate uptake in the brain. In pathologic circumstances the function of the transporters is impaired, leading to glutamate accumulation in the synaptic cleft and in turn excessive activation of postsynaptic glutamate receptors with subsequent massive Ca2+ influx, activation of neuronal nitric oxide synthase, translocation of proapoptotic genes to the mitochondria, mitochondrial dysfunction, release of cytochrome C into the cytosol, activation of caspases and subsequent cell death. Based on the pathogenic concept of an overactivation of the excitatory pathways, glutamate receptors have been a longstanding therapeutic target for rational drug design. This article reviews the pathophysiology of excitotoxic brain injury in the example of preterm brain injury, as well as current research on therapeutic antiexcitotoxic strategies.
Collapse
Affiliation(s)
- Elke Griesmaier
- Department of Pediatrics IV, Medical University Innsbruck, Austria, Anichstr. 35, 6020 Innsbruck, Austria
| | - Matthias Keller
- Department of Pediatrics I University Hospital Essen, Hufelandstraße 55, 45147 Essen Germany
| |
Collapse
|
11
|
Dickson L, Finlayson K. VPAC and PAC receptors: From ligands to function. Pharmacol Ther 2008; 121:294-316. [PMID: 19109992 DOI: 10.1016/j.pharmthera.2008.11.006] [Citation(s) in RCA: 287] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Accepted: 11/18/2008] [Indexed: 02/03/2023]
Abstract
Vasoactive intestinal peptide (VIP) and the pituitary adenylate cyclase activating polypeptides (PACAPs) share 68% identity at the amino acid level and belong to the secretin peptide family. Following the initial discovery of VIP almost four decades ago a substantial amount of knowledge has been presented describing the mechanisms of action, distribution and pleiotropic functions of these related peptides. It is now known that the physiological actions of these widely distributed peptides are produced through activation of three common G-protein coupled receptors (VPAC(1), VPAC(2) and PAC(1)R) which preferentially stimulate adenylate cyclase and increase intracellular cAMP, although stimulation of other intracellular messengers, including calcium and phospholipase D, has been reported. Using a range of in vitro and in vivo approaches, including cell-based functional assays, transgenic animals and rodent models of disease, VPAC/PAC receptor activation has been associated with numerous physiological processes (e.g. control of circadian rhythms) and clinical conditions (e.g. pulmonary hypertension), which underlies on-going research efforts and makes these peptides and their cognate receptors attractive targets for the pharmaceutical industry. However, despite the considerable interest in VPAC/PAC receptors and the processes which they mediate, there is still a paucity of selective and available, non-peptide ligands, which has hindered further advances in this field both at the basic research and clinical level. This review summarises the current knowledge of VIP/PACAP and the VPAC/PAC receptors with regard to their distribution, pharmacology, signalling pathways, splice variants and finally, the utility of animal models in exploring their physiological roles.
Collapse
Affiliation(s)
- Louise Dickson
- Centre for Integrative Physiology, University of Edinburgh, EH8 9XD, UK
| | | |
Collapse
|
12
|
Arciszewski MB, Sand E, Ekblad E. Vasoactive intestinal peptide rescues cultured rat myenteric neurons from lipopolysaccharide induced cell death. ACTA ACUST UNITED AC 2007; 146:218-23. [PMID: 17919746 DOI: 10.1016/j.regpep.2007.09.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 08/22/2007] [Accepted: 09/06/2007] [Indexed: 12/28/2022]
Abstract
UNLABELLED The role of the enteric nervous system in intestinal inflammation is not fully understood and the plethora of cellular activities concurrently ongoing in vivo renders intelligible studies difficult. In order to explore possible effects of bacterial lipopolysaccharide (LPS) on enteric neurons we utilised cultured myenteric neurons from rat small intestine. Exposure to LPS caused markedly reduced neuronal survival and increased neuronal expression of vasoactive intestinal peptide (VIP), while the expression of Toll-like receptor 4 (TLR4) was unchanged. TLR4 was expressed in approximately 35% of all myenteric neurons irrespective of if they were cultured in the presence or absence of LPS. In neurons cultured in medium, without LPS, 50% of all TLR4-immunoreactive neurons contained also VIP. Addition of LPS to the neuronal cultures markedly increased the proportion of TLR4-immunoreactive neurons also expressing VIP, while the proportion of TLR4 neurons devoid of VIP decreased. Simultaneous addition of LPS and VIP to the neuronal cultures resulted in a neuronal survival comparable to controls. CONCLUSIONS LPS recognition by myenteric neurons is mediated via TLR4 and causes neuronal cell death. Presence of VIP rescues the neurons from LPS-induced neurodegeneration.
Collapse
Affiliation(s)
- Marcin B Arciszewski
- Department of Animal Anatomy and Histology, Agricultural University, Lublin, Poland
| | | | | |
Collapse
|
13
|
Favrais G, Couvineau A, Laburthe M, Gressens P, Lelievre V. Involvement of VIP and PACAP in neonatal brain lesions generated by a combined excitotoxic/inflammatory challenge. Peptides 2007; 28:1727-1737. [PMID: 17683829 DOI: 10.1016/j.peptides.2007.06.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 06/18/2007] [Accepted: 06/21/2007] [Indexed: 02/08/2023]
Abstract
Several reports have highlighted the potential roles for the VIP-related neuropeptides in regeneration/neuroprotection after brain or nerve injuries. We previously reported that peripheral inflammation worsened ibotenate-induced cystic white matter lesions. Because VIP is also known as an immunomodulator, we wonder if VIP could also limit the deleterious effects of local inflammation. Therefore, we first tested the effects of peripheral IL-1beta on VIP and PACAP central production. Second, we observed that cox-2 activation by IL-1beta was essential to generate changes in ligand/receptor gene expression. We further tested whether the intraperitoneal injection of IL-1beta, known to aggravate the ibotenate-induced lesions, could modify the expression pattern of VIP-related genes. Finally, we concluded using histological analysis that VIP[ala(11,22,28)], a synthetic VPAC(1) agonist completely reversed the aggravating effects of IL-1beta on ibotenate-induced lesions of the periventricular white matter. Conversely, VIP-neurotensin hybrid, a nonselective VIP receptor antagonist, worsened the lesions. All together, our results suggest that an activation of VIP/VPAC(1) signaling cascade in the vicinity of the injury site could circumvent the synergizing degenerative effects of ibotenate and pro-inflammatory cytokines. Therefore, development of therapeutic tools inducing/sustaining the activation of VIP/VPAC(1) signaling cascade may lead to future preventive treatments for inflammatory conditions during pregnancy.
Collapse
Affiliation(s)
- Geraldine Favrais
- Inserm U676, hôpital Robert-Debré, 48, boulevard Sérurier, 75019 Paris, France
| | | | | | | | | |
Collapse
|
14
|
Muller JM, Debaigt C, Goursaud S, Montoni A, Pineau N, Meunier AC, Janet T. Unconventional binding sites and receptors for VIP and related peptides PACAP and PHI/PHM: an update. Peptides 2007; 28:1655-66. [PMID: 17555844 DOI: 10.1016/j.peptides.2007.04.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2007] [Revised: 03/23/2007] [Accepted: 04/10/2007] [Indexed: 11/25/2022]
Abstract
The 28-amino-acid neuropeptide VIP and related peptides PACAP and PHI/PHM modulate virtually all of the vital functions in the body. These peptides are also commonly recognized as major regulators of cell growth and differentiation. Through their trophic and cytoprotective functions, they appear to play major roles in embryonic development, neurogenesis and the progression of a number of cancer types. These peptides bind to three well-characterized subtypes of G-protein coupled receptors: VPAC1 and VPAC2 share a common high affinity in the nanomolar range for VIP and PACAP; a third receptor type, PAC1, has been characterized for its high affinity for PACAP but its low affinity for VIP. Complex effects and pharmacological behaviors of these peptides suggest that multiple subtypes of binding sites may cooperate to mediate their function in target cells and tissues. In this complex response, some of these binding sites correspond to the definition of the conventional receptors cited above, while others display unexpected pharmacological and functional properties. Here we present potential clues that may lead investigators to further characterize the molecular nature and functions of these atypical binding species.
Collapse
Affiliation(s)
- Jean-Marc Muller
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, Pôle Biologie-Santé, 40 Avenue du Recteur Pineau, 86022 Poitiers Cedex, France.
| | | | | | | | | | | | | |
Collapse
|