1
|
SH3- and actin-binding domains connect ADNP and SHANK3, revealing a fundamental shared mechanism underlying autism. Mol Psychiatry 2022; 27:3316-3327. [PMID: 35538192 DOI: 10.1038/s41380-022-01603-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022]
Abstract
De novo heterozygous mutations in activity-dependent neuroprotective protein (ADNP) cause autistic ADNP syndrome. ADNP mutations impair microtubule (MT) function, essential for synaptic activity. The ADNP MT-associating fragment NAPVSIPQ (called NAP) contains an MT end-binding protein interacting domain, SxIP (mimicking the active-peptide, SKIP). We hypothesized that not all ADNP mutations are similarly deleterious and that the NAPV portion of NAPVSIPQ is biologically active. Using the eukaryotic linear motif (ELM) resource, we identified a Src homology 3 (SH3) domain-ligand association site in NAP responsible for controlling signaling pathways regulating the cytoskeleton, namely NAPVSIP. Altogether, we mapped multiple SH3-binding sites in ADNP. Comparisons of the effects of ADNP mutations p.Glu830synfs*83, p.Lys408Valfs*31, p.Ser404* on MT dynamics and Tau interactions (live-cell fluorescence-microscopy) suggested spared toxic function in p.Lys408Valfs*31, with a regained SH3-binding motif due to the frameshift insertion. Site-directed-mutagenesis, abolishing the p.Lys408Valfs*31 SH3-binding motif, produced MT toxicity. NAP normalized MT activities in the face of all ADNP mutations, although, SKIP, missing the SH3-binding motif, showed reduced efficacy in terms of MT-Tau interactions, as compared with NAP. Lastly, SH3 and multiple ankyrin repeat domains protein 3 (SHANK3), a major autism gene product, interact with the cytoskeleton through an actin-binding motif to modify behavior. Similarly, ELM analysis identified an actin-binding site on ADNP, suggesting direct SH3 and indirect SHANK3/ADNP associations. Actin co-immunoprecipitations from mouse brain extracts showed NAP-mediated normalization of Shank3-Adnp-actin interactions. Furthermore, NAP treatment ameliorated aberrant behavior in mice homozygous for the Shank3 ASD-linked InsG3680 mutation, revealing a fundamental shared mechanism between ADNP and SHANK3.
Collapse
|
2
|
Karmon G, Sragovich S, Hacohen-Kleiman G, Ben-Horin-Hazak I, Kasparek P, Schuster B, Sedlacek R, Pasmanik-Chor M, Theotokis P, Touloumi O, Zoidou S, Huang L, Wu PY, Shi R, Kapitansky O, Lobyntseva A, Giladi E, Shapira G, Shomron N, Bereswill S, Heimesaat MM, Grigoriadis N, McKinney RA, Rubinstein M, Gozes I. Novel ADNP Syndrome Mice Reveal Dramatic Sex-Specific Peripheral Gene Expression With Brain Synaptic and Tau Pathologies. Biol Psychiatry 2022; 92:81-95. [PMID: 34865853 DOI: 10.1016/j.biopsych.2021.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/19/2021] [Accepted: 09/17/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND ADNP is essential for embryonic development. As such, de novo ADNP mutations lead to an intractable autism/intellectual disability syndrome requiring investigation. METHODS Mimicking humans, CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 editing produced mice carrying heterozygous Adnp p.Tyr718∗ (Tyr), a paralog of the most common ADNP syndrome mutation. Phenotypic rescue was validated by treatment with the microtubule/autophagy-protective ADNP fragment NAPVSIPQ (NAP). RESULTS RNA sequencing of spleens, representing a peripheral biomarker source, revealed Tyr-specific sex differences (e.g., cell cycle), accentuated in females (with significant effects on antigen processing and cellular senescence) and corrected by NAP. Differentially expressed, NAP-correctable transcripts, including the autophagy and microbiome resilience-linked FOXO3, were also deregulated in human patient-derived ADNP-mutated lymphoblastoid cells. There were also Tyr sex-specific microbiota signatures. Phenotypically, Tyr mice, similar to patients with ADNP syndrome, exhibited delayed development coupled with sex-dependent gait defects. Speech acquisition delays paralleled sex-specific mouse syntax abnormalities. Anatomically, dendritic spine densities/morphologies were decreased with NAP amelioration. These findings were replicated in the Adnp+/- mouse, including Foxo3 deregulation, required for dendritic spine formation. Grooming duration and nociception threshold (autistic traits) were significantly affected only in males. Early-onset tauopathy was accentuated in males (hippocampus and visual cortex), mimicking humans, and was paralleled by impaired visual evoked potentials and correction by acute NAP treatment. CONCLUSIONS Tyr mice model ADNP syndrome pathology. The newly discovered ADNP/NAP target FOXO3 controls the autophagy initiator LC3 (microtubule-associated protein 1 light chain 3), with known ADNP binding to LC3 augmented by NAP, protecting against tauopathy. NAP amelioration attests to specificity, with potential for drug development targeting accessible biomarkers.
Collapse
Affiliation(s)
- Gidon Karmon
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Shlomo Sragovich
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Gal Hacohen-Kleiman
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Ben-Horin-Hazak
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Petr Kasparek
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Björn Schuster
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Radislav Sedlacek
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Olga Touloumi
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sofia Zoidou
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Linxuan Huang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Pei You Wu
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Roy Shi
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Oxana Kapitansky
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Alexandra Lobyntseva
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Eliezer Giladi
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - Guy Shapira
- Department of Cell and Developmental Biology and Edmond J. Safra Center for Bioinformatics, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology and Edmond J. Safra Center for Bioinformatics, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute for Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Markus M Heimesaat
- Gastrointestinal Microbiology Research Group, Institute for Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Nikolaos Grigoriadis
- Department of Neurology, Laboratory of Experimental Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - R Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Moran Rubinstein
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel; Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
3
|
Ernenwein D, St. John SE, Stewart AJ, Morimoto BH, Chmielewski J, Lipton MA. Structural studies and cyclization of the neuroprotective octapeptide
NAPVSIPQ
to improve cell permeability. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Dawn Ernenwein
- Department of ChemistryPurdue University West Lafayette Indiana USA
| | | | | | | | - Jean Chmielewski
- Department of ChemistryPurdue University West Lafayette Indiana USA
| | - Mark A. Lipton
- Department of ChemistryPurdue University West Lafayette Indiana USA
| |
Collapse
|
4
|
Arya A, Meena R, Sethy NK, Das M, Sharma M, Bhargava K. NAP (davunetide) protects primary hippocampus culture by modulating expression profile of antioxidant genes during limiting oxygen conditions. Free Radic Res 2015; 49:440-52. [DOI: 10.3109/10715762.2015.1011153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
5
|
Goursaud S, Schäfer S, Dumont AO, Vergouts M, Gallo A, Desmet N, Deumens R, Hermans E. The anti-inflammatory peptide stearyl-norleucine-VIP delays disease onset and extends survival in a rat model of inherited amyotrophic lateral sclerosis. Exp Neurol 2014; 263:91-101. [PMID: 25311268 DOI: 10.1016/j.expneurol.2014.09.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 12/13/2022]
Abstract
Vasoactive intestinal peptide (VIP) has potent immune modulatory actions that may influence the course of neurodegenerative disorders associated with chronic inflammation. Here, we show the therapeutic benefits of a modified peptide agonist stearyl-norleucine-VIP (SNV) in a transgenic rat model of amyotrophic lateral sclerosis (mutated superoxide dismutase 1, hSOD1(G93A)). When administered by systemic every-other-day intraperitoneal injections during a period of 80 days before disease, SNV delayed the onset of motor dysfunction by no less than three weeks, while survival was extended by nearly two months. SNV-treated rats showed reduced astro- and microgliosis in the lumbar ventral spinal cord and a significant degree of motor neuron preservation. Throughout the treatment, SNV promoted the expression of the anti-inflammatory cytokine interleukin-10 as well as neurotrophic factors commonly considered as beneficial in amyotrophic lateral sclerosis management (glial derived neuroptrophic factor, insulin like growth factor, brain derived neurotrophic factor). The peptide nearly totally suppressed the expression of tumor necrosis factor-α and repressed the production of the pro-inflammatory mediators interleukin-1β, nitric oxide and of the transcription factor nuclear factor kappa B. Inhibition of tumor necrosis factor-α likely accounted for the observed down-regulation of nuclear factor kappa B that modulates the transcription of genes specifically involved in amyotrophic lateral sclerosis (sod1 and the glutamate transporter slc1a2). In line with this, levels of human superoxide dismutase 1 mRNA and protein were decreased by SNV treatment, while the expression and activity of the glutamate transporter-1 was promoted. Considering the large diversity of influences of this peptide on both clinical features of the disease and associated biochemical markers, we propose that SNV or related peptides may constitute promising candidates for amyotrophic lateral sclerosis treatment.
Collapse
Affiliation(s)
- Stéphanie Goursaud
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Sabrina Schäfer
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Amélie O Dumont
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Maxime Vergouts
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Alessandro Gallo
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Nathalie Desmet
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Ronald Deumens
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Emmanuel Hermans
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, B-1200 Brussels, Belgium.
| |
Collapse
|
6
|
Greggio S, de Paula S, de Oliveira IM, Trindade C, Rosa RM, Henriques JAP, DaCosta JC. NAP prevents acute cerebral oxidative stress and protects against long-term brain injury and cognitive impairment in a model of neonatal hypoxia-ischemia. Neurobiol Dis 2011; 44:152-9. [PMID: 21757007 DOI: 10.1016/j.nbd.2011.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 06/17/2011] [Accepted: 06/26/2011] [Indexed: 10/18/2022] Open
Abstract
Hypoxia-ischemia (HI) is a common cause of neonatal brain damage with lifelong morbidities in which current therapies are limited. In this study, we investigated the effect of neuropeptide NAP (NAPVSIPQ) on early cerebral oxidative stress, long-term neurological function and brain injury after neonatal HI. Seven-day-old rat pups were subjected to an HI model by applying a unilateral carotid artery occlusion and systemic hypoxia. The animals were randomly assigned to groups receiving an intraperitoneal injection of NAP (3 μg/g) or vehicle immediately (0 h) and 24 h after HI. Brain DNA damage, lipid peroxidation and reduced glutathione (GSH) content were determined 24 h after the last NAP injection. Cognitive impairment was assessed on postnatal day 60 using the spatial version of the Morris water maze learning task. Next, the animals were euthanized to assess the cerebral hemispheric volume using the Cavalieri principle associated with the counting point method. We observed that NAP prevented the acute HI-induced DNA and lipid membrane damage and also recovered the GSH levels in the injured hemisphere of the HI rat pups. Further, NAP was able to prevent impairments in learning and long-term spatial memory and to significantly reduce brain damage up to 7 weeks following the neonatal HI injury. Our findings demonstrate that NAP confers potent neuroprotection from acute brain oxidative stress, long-term cognitive impairment and brain lesions induced by neonatal HI through, at least in part, the modulation of the glutathione-mediated antioxidant system.
Collapse
Affiliation(s)
- Samuel Greggio
- Laboratório de Neurociências, Instituto do Cérebro e Instituto de Pesquisas Biomédicas, Programa de Pós-Graduação em Pediatria e Saúde da Criança, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
7
|
JUNG EUNYOUNG, KIM SEONGYEONG, BAE SONGHWAN, CHANG UNJAE, CHOI JANGWON, SUH HYUNGJOO. WEIGHT REDUCTION EFFECTS OF YEAST HYDROLYSATE BELOW 10 kDa ON OBESE YOUNG WOMEN. J Food Biochem 2011. [DOI: 10.1111/j.1745-4514.2010.00385.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
8
|
Greggio S, Rosa RM, Dolganov A, de Oliveira IM, Menegat FD, Henriques JA, DaCosta JC. NAP prevents hippocampal oxidative damage in neonatal rats subjected to hypoxia-induced seizures. Neurobiol Dis 2009; 36:435-44. [DOI: 10.1016/j.nbd.2009.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 08/08/2009] [Accepted: 08/17/2009] [Indexed: 11/29/2022] Open
|
9
|
Jung EY, Suh HJ, Kim SY, Hong YS, Kim MJ, Chang UJ. Appetite suppressive effects of yeast hydrolysate on nitric oxide synthase (NOS) expression and vasoactive intestinal peptide (VIP) immunoreactivity in hypothalamus. Phytother Res 2008; 22:1417-22. [DOI: 10.1002/ptr.2264] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
10
|
VIP, from gene to behavior and back: summarizing my 25 years of research. J Mol Neurosci 2008; 36:115-24. [PMID: 18607776 DOI: 10.1007/s12031-008-9105-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Accepted: 05/15/2008] [Indexed: 11/25/2022]
Abstract
Vasoactive intestinal peptide (VIP) is an interesting example of a 28-amino acid neuropeptide that is abundantly expressed in discrete brain regions/neurons and hence may contribute to brain function. This short review summarizes my own point of view and encompasses 25 years of work and over 100 publications targeting the understanding of VIP production and biological activity. The review starts with our original cloning of the VIP gene, it then continues to discoveries of regulation of VIP synthesis and the establishment of the first VIP transgenic mice. The review ends with the identification of novel VIP analogs that helped decipher VIP's important role during development, in regulation of the biological clock(s) and diurnal rhythms, sexual activity, learning and memory as well as social behavior, and cancer. This review cites only articles that I have coauthored and gives my own perspective of this exciting ever-growing field.
Collapse
|
11
|
The neuropeptide NAP provides neuroprotection against retinal ganglion cell damage after retinal ischemia and optic nerve crush. Graefes Arch Clin Exp Ophthalmol 2008; 246:1255-63. [PMID: 18414890 DOI: 10.1007/s00417-007-0746-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 11/21/2007] [Accepted: 11/26/2007] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND NAP, an 8-amino acid peptide (NAPVSIPQ=Asn-Ala-Pro-Val-Ser-Ile-Pro-Gln) derived from activity-dependent neuroprotective protein (ADNP), plays an important role in neuronal differentiation and the survival of neurons in different pathological situations. We already discovered that NAP increases the survival of retinal ganglion cells (RGC) in vitro, and supports neurite outgrowth in retinal explants at femtomolar concentrations. The aim of this study was to investigate the effects of NAP on RGC survival after transient retinal ischemia and optic nerve crush. METHODS RGC of male Wistar rats were labelled retrogradely with 6 l FluoroGold injected stereotactically into both superior colliculi. Seven days later, retinal ischemia was induced by elevating the intraocular pressure to 120 mm Hg for 60 minutes or by crushing one optic nerve for 10 s after a partial orbitotomy. NAP was either injected intraperitoneally in the concentration of 100 microg/kg [corrected] 1 day before, directly after, and on the first and the second days after damage, or intravitreally (0.05 or 0.5 microg/eye) [corrected] directly after the optic nerve crush. Controls received the same concentrations of a control peptide. Densities of surviving RGC and activated microglial cells (AMC) were quantified in a masked fashion 10 days after damage by counting FluoroGold-labelled cells. RESULTS After retinal ischemia, intraperitoneal injections of NAP increased the number of surviving RGC by 40% (p < 0.005) compared to the control group. After optic nerve crush, NAP raised the number of surviving RGC by 31% (p = 0.07) when injected intraperitoneally and by 54% (p < 0.05) when administered intravitreally. CONCLUSIONS NAP acts neuroprotectively in vivo after retinal ischemia and optic nerve crush, and may have potential in treating optic nerve diseases.
Collapse
|
12
|
Cosgrave AS, McKay JS, Bubb V, Morris R, Quinn JP, Thippeswamy T. Regulation of activity-dependent neuroprotective protein (ADNP) by the NO-cGMP pathway in the hippocampus during kainic acid-induced seizure. Neurobiol Dis 2008; 30:281-292. [PMID: 18375135 DOI: 10.1016/j.nbd.2008.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 01/30/2008] [Accepted: 02/01/2008] [Indexed: 12/11/2022] Open
Abstract
Activity-dependent neuroprotective protein (ADNP) is widely distributed in the cytoplasm of neurons and astrocytes of the hippocampus. Kainic acid (KA)-induced seizures increases neuronal nitric oxide synthase (nNOS) in neurons and inducible NOS (iNOS) in glia cells which coincides with a reduction in ADNP in the hippocampus. Inhibitors of NOS or soluble guanylyl cyclase (sGC) activity reduce ADNP under basal conditions in the absence of seizures. Treating animals with these inhibitors prior to KA-induced seizure, in particular, L-NAME (N(G)-nitro-l-arginine methyl ester), advances the onset of the first seizure but reverses the loss of ADNP by 3 days after the first seizure. This suggests that the NO-cGMP pathway has a role in regulating ADNP under both basal physiological conditions and in the pathophysiological changes produced during epileptogenesis.
Collapse
Affiliation(s)
- Anna S Cosgrave
- Department of Veterinary Preclinical Science, Veterinary Faculty, University of Liverpool, Brownlow Hill, Liverpool, L69 7ZJ, UK
| | - Jennifer S McKay
- AstraZeneca, Department of Pathology Safety Assessment, Alderley Park, Macclesfield, SK10 4TG, UK
| | - Vivien Bubb
- School of Biomedical Sciences, Medical School, University of Liverpool, Liverpool L69 3BX, UK
| | - Richard Morris
- Department of Veterinary Preclinical Science, Veterinary Faculty, University of Liverpool, Brownlow Hill, Liverpool, L69 7ZJ, UK
| | - John P Quinn
- School of Biomedical Sciences, Medical School, University of Liverpool, Liverpool L69 3BX, UK
| | - Thimmasettappa Thippeswamy
- Department of Veterinary Preclinical Science, Veterinary Faculty, University of Liverpool, Brownlow Hill, Liverpool, L69 7ZJ, UK.
| |
Collapse
|
13
|
Blat D, Weiner L, Youdim MBH, Fridkin M. A Novel Iron-Chelating Derivative of the Neuroprotective Peptide NAPVSIPQ Shows Superior Antioxidant and Antineurodegenerative Capabilities. J Med Chem 2007; 51:126-34. [DOI: 10.1021/jm070800l] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Dan Blat
- Department of Organic Chemistry and Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel, and Eve Topf and U.S.A. National Parkinson Foundation Centers for Neurodegenerative Diseases and Department of Pharmacology, Faculty of Medicine, Technion, Haifa, Israel
| | - Lev Weiner
- Department of Organic Chemistry and Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel, and Eve Topf and U.S.A. National Parkinson Foundation Centers for Neurodegenerative Diseases and Department of Pharmacology, Faculty of Medicine, Technion, Haifa, Israel
| | - Moussa B. H. Youdim
- Department of Organic Chemistry and Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel, and Eve Topf and U.S.A. National Parkinson Foundation Centers for Neurodegenerative Diseases and Department of Pharmacology, Faculty of Medicine, Technion, Haifa, Israel
| | - Mati Fridkin
- Department of Organic Chemistry and Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel, and Eve Topf and U.S.A. National Parkinson Foundation Centers for Neurodegenerative Diseases and Department of Pharmacology, Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
14
|
Thippeswamy T, Howard MR, Cosgrave AS, Arora DK, McKay JS, Quinn JP. Nitric oxide-NGF mediated PPTA/SP, ADNP, and VIP expression in the peripheral nervous system. J Mol Neurosci 2007; 33:268-77. [PMID: 17952636 DOI: 10.1007/s12031-007-0066-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Accepted: 11/30/2006] [Indexed: 12/14/2022]
Abstract
Nerve growth factor (NGF)-deprivation or axotomy of dorsal root ganglion (DRG) neurons causes stress, which they cope by triggering various mechanisms. Among several molecular changes, in the present study, we demonstrate preprotachykinin-A-substance P (PPTA-SP) and activity-dependent neuroprotective protein-vasoactive intestinal peptide (ADNP-VIP) expression pattern using DRG neurons-Schwann cells coculture and axotomy model. In the presence of NGF, DRG cultures showed high levels of PPTA and ADNP mRNA expression, which were significantly suppressed in the absence of NGF and/or nitric oxide synthase (NOS) inhibition by NG-nitro-L-arginine methyl ester (L-NAME), suggesting that both NGF and nitric oxide (NO) can regulate PPTA and ADNP expression. However, treating coculture with NO donor, diethylenetriamine nitric oxide (DETA-NO) did not increase PPTA and ADNP expression in the presence or absence of NGF, although there was a marginal increase in ADNP expression in the absence of NGF. NGF-deprivation increases endogenous NO; thus, DETA-NO had no further effect on PPTA and ADNP expression. Alternatively, NGF produced from NO-stimulated Schwann cells influence gene expression. In addition, interestingly, DETA-NO treatment of Schwann cells alone suppresses both PPTA and ADNP, suggesting differential response of DRG neurons-Schwann cells coculture to DETA-NO. SP and ADNP immunostaining of axotomized DRGs revealed significant reduction in SP and ADNP compared to intact DRG, which was partially recovered in neuronal NOS blocker, 7-nitroindazole (7-NI)-treated DRGs, particularly intense ADNP staining in satellite glia. As ADNP is VIP-responsive gene, we further explored VIP expression in DRGs. Axotomy increased VIP in DRG neurons, but 7-NI treatment caused intense VIP staining in satellite glia. These observations suggest a complex interaction of NO-NGF with PPTA/SP and ADNP-VIP in neuron-glial communication when neurons are stressed.
Collapse
Affiliation(s)
- Thimmasettappa Thippeswamy
- Department of Veterinary Preclinical Sciences, University of Liverpool, Brownlowhill Street, Liverpool, L69 7ZJ, UK.
| | | | | | | | | | | |
Collapse
|
15
|
Gozes I, Morimoto BH, Tiong J, Fox A, Sutherland K, Dangoor D, Holser-Cochav M, Vered K, Newton P, Aisen PS, Matsuoka Y, van Dyck CH, Thal L. NAP: research and development of a peptide derived from activity-dependent neuroprotective protein (ADNP). CNS DRUG REVIEWS 2006; 11:353-68. [PMID: 16614735 PMCID: PMC6741706 DOI: 10.1111/j.1527-3458.2005.tb00053.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Activity-dependent neuroprotective protein (ADNP) is essential for brain formation. Peptide activity scanning identified NAP (NAPVSIPQ) as a small active fragment of ADNP that provides neuroprotection at very low concentrations. In cell culture, NAP has demonstrated protection against toxicity associated with the beta-amyloid peptide, N-methyl-D-aspartate, electrical blockade, the envelope protein of the AIDS virus, dopamine, H2O2, nutrient starvation and zinc overload. NAP has also provided neuroprotection in animal models of apolipoprotein E deficiency, cholinergic toxicity, closed head injury, stroke, middle aged anxiety and cognitive dysfunction. NAP binds to tubulin and facilitates microtubule assembly leading to enhanced cellular survival that is associated with fundamental cytoskeletal elements. A liquid-chromatography, mass spectrometry assay demonstrated that NAP reaches the brain after either intravenous or intranasal administration. In a battery of toxicological tests including repeated dose toxicity in rats and dogs, cardiopulmonary tests in dogs, and functional behavioral assays in rats, no adverse side effects were observed with NAP concentrations that were approximately 500-fold higher than the biologically active dose. A Phase Ia clinical trial in the US assessed the tolerability and pharmacokinetics of intranasal administration of NAP in sequential ascending doses. The results supported the safety and tolerability of a single dose of NAP administered at up to 15 mg intranasally. Furthermore, dosing was recently completed for a second Phase I clinical trial in healthy adults and elderly volunteers with an intravenous formulation of NAP. NAP is poised for further clinical development targeting several indications, including Alzheimer's disease.
Collapse
Affiliation(s)
- Illana Gozes
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Smith-Swintosky VL, Gozes I, Brenneman DE, D'Andrea MR, Plata-Salaman CR. Activity-dependent neurotrophic factor-9 and NAP promote neurite outgrowth in rat hippocampal and cortical cultures. J Mol Neurosci 2005; 25:225-38. [PMID: 15800376 DOI: 10.1385/jmn:25:3:225] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2004] [Accepted: 10/19/2004] [Indexed: 11/11/2022]
Abstract
Activity-dependent neurotrophic factor (ADNF) is a novel, femtomolar-acting, glial-derived polypeptide (14 kDa) known to protect neurons from a variety of toxic insults. The active site for ADNF function is localized to a 9-amino-acid stretch (SALLRSIPA; ADNF-9). A few years later, a novel ADNF-9-like active peptide (NAPVSIPQ or NAP) was identified and shown to be expressed in the CNS and exhibit an activity profile similar to ADNF-9. Such studies suggest that ADNF-9 and NAP might function like other known neurotrophins and play a role in neural development and maintenance. The purpose of the present studies was to determine if ADNF-9 or NAP affects neurite outgrowth and synaptogenesis in rat hippocampal and cortical cultures. Using MAP2-FITC immunofluorescent labeling, we found that ADNF-9 and NAP promoted neurite outgrowth in a concentration-dependent manner, with maximal activity observed at femtomolar concentrations. Both peptides stimulated robust outgrowth in hippocampal cells (approximately 150% of control; p < 0.01) with a modest effect on cortical cells (approximately 20% of control; p < 0.05) similar to other known growth factors. However, the outgrowth-promoting effect was abolished in the absence of serum, suggesting that soluble factors might be necessary for the neurotrophic activity. Finally, we found that ADNF-9 and NAP increased synaptophysin expression in both rat hippocampal and cortical cultures. These results suggest that ADNF-9 and NAP might contribute to neuronal plasticity associated with development and repair after injury.
Collapse
Affiliation(s)
- Virginia L Smith-Swintosky
- CNS Research, Johnson & Johnson Pharmaceutical Research and Development, LLC, Spring House, PA 19447-0776, USA.
| | | | | | | | | |
Collapse
|
17
|
Abstract
An 8-amino-acid peptide, NAPVSIPQ (NAP), was identified as the smallest active element of activity-dependent neuroprotective protein that exhibits potent neuroprotective action. Potential signal transduction pathways include cGMP production and interference with inflammatory mechanisms, tumor necrosis factor-alpha, and MAC1-related changes. Because of its intrinsic structure, NAP might interact with extracellular proteins and also transverse membranes. NAP-associated protection against oxidative stress, glucose deprivation, and apoptotic mechanisms suggests interference with fundamental processes. This paper identifies p53, a key regulator of cellular apoptosis, as an intracellular target for NAP's activity.
Collapse
Affiliation(s)
- Illana Gozes
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | |
Collapse
|
18
|
Zusev M, Gozes I. Differential regulation of activity-dependent neuroprotective protein in rat astrocytes by VIP and PACAP. ACTA ACUST UNITED AC 2004; 123:33-41. [PMID: 15518891 DOI: 10.1016/j.regpep.2004.05.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Activity-dependent neuroprotective protein (ADNP) was shown to be a vasoactive intestinal peptide (VIP) responsive gene in astrocytes derived from the cerebral cortex of newborn rats. The present study was set out to identify VIP receptors that are associated with increases in ADNP expression in developing astrocytes. Using VIP analogues specific for the VPAC1 and the VPAC2 receptors, it was discovered that VIP induced changes in ADNP expression in astrocytes via the VPAC2 receptor. The constitutive synthesis of ADNP and VPAC2 was shown to be age-dependent and increased as the astrocyte culture developed. Pituitary adenylate cyclase-activating polypeptide (PACAP) also induced changes in ADNP expression. The apparent changes induced by VIP and PACAP on ADNP expression were developmentally dependent, and while stimulating expression in young astrocytes, an inhibition was demonstrated in older cultures. In conclusion, VIP, PACAP and the VPAC2 receptor may all contribute to the regulation of ADNP gene expression in the developing astrocyte.
Collapse
MESH Headings
- Animals
- Astrocytes/cytology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Base Sequence
- Cell Differentiation
- Cells, Cultured
- Cellular Senescence
- Gene Expression Regulation, Developmental/drug effects
- Homeodomain Proteins/biosynthesis
- Homeodomain Proteins/genetics
- Models, Biological
- Nerve Growth Factors/pharmacology
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/genetics
- Neuropeptides/pharmacology
- Neurotransmitter Agents/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Receptors, Cell Surface/genetics
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Vasoactive Intestinal Peptide/genetics
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Vasoactive Intestinal Peptide/pharmacology
Collapse
Affiliation(s)
- Margalit Zusev
- Department of Clinical Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | |
Collapse
|
19
|
Chen L, Taishi P, Duricka D, Krueger JM. Brainstem prolactin mRNA is enhanced in mice with suppressed neuronal nitric oxide synthase activity. ACTA ACUST UNITED AC 2004; 129:179-84. [PMID: 15469894 DOI: 10.1016/j.molbrainres.2004.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2004] [Indexed: 11/17/2022]
Abstract
Prolactin (PRL) and vasoactive intestinal polypeptide (VIP) mRNA levels were elevated in the brainstem of neuronal nitric oxide synthase (nNOS) gene knockout (KO) mice compared to the levels in nNOS control mice. In addition, PRL mRNA levels increased in the hypothalamus and the brainstem of nNOS control mice after administration of 7-nitro-indazole (7-NI), a relatively selective nNOS inhibitor. The results suggest that NO inhibits PRL. No differences in the genes measured were observed in inducible NOS KO mice.
Collapse
Affiliation(s)
- Lichao Chen
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, 205 Wegner Hall, P.O. Box 646520, Pullman, WA 99164-6520, USA
| | | | | | | |
Collapse
|
20
|
Divinski I, Mittelman L, Gozes I. A Femtomolar Acting Octapeptide Interacts with Tubulin and Protects Astrocytes against Zinc Intoxication. J Biol Chem 2004; 279:28531-8. [PMID: 15123709 DOI: 10.1074/jbc.m403197200] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An octapeptide was previously described that protects neurons against a wide variety of insults directly and indirectly as a result of interactions (at femtomolar concentrations) with supporting glial cells. The current study set out to identify the octapeptide binding molecules so as to understand the high affinity mechanisms of cellular protection. Studies utilizing affinity chromatography of brain extracts identified tubulin, the brain major protein, as the octapeptide-binding ligand. Dot blot analysis with pure tubulin and the biotinylated octapeptide verified this finding. When added to cerebral cortical astrocytes, the octapeptide (10(-15)-10(-10) m) induced a rapid microtubule reorganization into distinct microtubular structures that were stained by monoclonal tubulin antibodies and visualized by confocal microscopy. Fluorescein-labeled octapeptide induced a similar change and was detected in the intracellular milieu, even when cells were incubated at 4 degrees C or at low pH. In a cell-free system, the octapeptide stimulated tubulin assembly into microtubules. Furthermore, treatment of astrocytes with zinc chloride resulted in microtubule disassembly and cell death that was protected by the octapeptide. In conclusion, the results suggest that the octapeptide crosses the plasma membrane and interacts directly with tubulin, the microtubule subunit, to induce microtubule reorganization and improved survival. Because microtubules are the key component of the neuronal and glial cytoskeleton that regulates cell division, differentiation, and protection, this finding may explain the breadth and efficiency of the cellular protective capacities of the octapeptide.
Collapse
Affiliation(s)
- Inna Divinski
- Department of Clinical Biochemistry and Interdepartmental Core Facility, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | |
Collapse
|
21
|
Ekblad E, Bauer AJ. Role of vasoactive intestinal peptide and inflammatory mediators in enteric neuronal plasticity. Neurogastroenterol Motil 2004; 16 Suppl 1:123-8. [PMID: 15066017 DOI: 10.1111/j.1743-3150.2004.00487.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Complex circuits involving both local intrinsic neurones (i.e. enteric nervous system; ENS) and extrinsic neurones achieve nervous control of digestive functions. The ENS is comprised of many functionally different types of neurons: sensory neurons, interneurons and secreto-motor neurons. Each neuronal population is required to manifest local reflex behavior and is central to the regulation of both motor and secretory activities. It must be emphasized, however, that not only muscle and secretory cells but also other intestinal cells are targeted by enteric neurones, i.e. endocrine cells, interstitial cells of Cajal, immune cells, blood vessels and enteric glia. In addition to the ENS the gastrointestinal tract receives an extrinsic innervation by sympathetic, parasympathetic and sensory fibres. Neuronal projections from the intestine to prevertebral ganglia also exist. Taken together, the picture of a complex nervous regulation of digestive functions highly integrated with the central nervous system and the rest of the autonomic nervous system has emerged. The ENS is adaptive and plastic, but also vulnerable, system and ENS disturbances may be of pathogenic importance in functional bowel disease. In particular the interplay between the enteric neurones and the immune cells is suggested to be of crucial importance. The review discusses possible roles of the mediators vasoactive intestinal peptide (VIP) and prostanoids in ENS plasticity in response to injury and inflammation.
Collapse
Affiliation(s)
- E Ekblad
- Department of Physiological Sciences, Neuroendocrine Cell Biology, Lund University, Lund, Sweden
| | | |
Collapse
|
22
|
Brenneman DE, Spong CY, Hauser JM, Abebe D, Pinhasov A, Golian T, Gozes I. Protective peptides that are orally active and mechanistically nonchiral. J Pharmacol Exp Ther 2004; 309:1190-7. [PMID: 15007105 DOI: 10.1124/jpet.103.063891] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous reports identified two peptides that mimic the action of neuroprotective proteins derived from astrocytes. These peptides, NAPVSIPQ and SALLRSIPA, prevent neuronal cell death produced by electrical blockade, N-methyl-d-aspartate, and beta-amyloid peptide (25-35). In the present study, all d-amino acid peptides of NAPVSIPQ and SALLRSIPA were synthesized and compared respectively to the corresponding all l-amino acid peptides. In rat cerebral cortical test cultures cotreated with 1 microM tetrodotoxin, the d-amino acid peptides produced similar potency and efficacy for neuroprotection as that observed for their respective l-amino acid peptides. Since all these peptides tested individually exhibited attenuation of efficacy at concentrations of >10 pM, combinations of these peptides were tested for possible synergies. Equimolar d-NAPVSIPQ and d-SALLRSIPA combination treatment produced potent neuroprotection (EC(50), 0.03 fM) that did not attenuate with increasing concentrations. Similarly, the combination of l-NAPVSIPQ and d-SALLRSIPA also had high potency (EC(50), 0.07 fM) without attenuation of efficacy. Combined administration of peptides was tested in a model of fetal alcohol syndrome and in a model of learning impairment: apolipoprotein E knockout mice. Intraperitoneal administration of d-NAPVSIPQ plus d-SALLRSIPA to pregnant mice (embryonic day 8) attenuated fetal demise after treatment with an acute high dose of alcohol. Furthermore, oral administration of d-NAPVSIPQ plus d-SALLRSIPA significantly increased fetal survival after maternal alcohol treatment. Apolipoprotein E knockout mice injected with d-NAPVSIPQ plus d-SALLRSIPA showed improved performance in the Morris water maze. These studies suggest therapeutic potential for the combined administration of neuroprotective peptides that can act through a mechanism independent of chiral recognition.
Collapse
Affiliation(s)
- Douglas E Brenneman
- Section of Developmental and Molecular Pharmacology, Laboratory of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Gozes I, Divinsky I, Pilzer I, Fridkin M, Brenneman DE, Spier AD. From vasoactive intestinal peptide (VIP) through activity-dependent neuroprotective protein (ADNP) to NAP: a view of neuroprotection and cell division. J Mol Neurosci 2003; 20:315-22. [PMID: 14501014 DOI: 10.1385/jmn:20:3:315] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2002] [Accepted: 03/24/2003] [Indexed: 01/01/2023]
Abstract
Accelerated neuronal death brings about cognitive as well as motor and other dysfunctions. A major neuropeptide, vasoactive intestinal peptide (VIP), has been shown to be neuroprotective. However, VIP-based drug design is hampered by the instability of the peptide and its limited bioavailability. Two independent approaches were thus taken to exploit VIP as a lead drug candidate: (1) Potent neuroprotective lipophilic analogs of VIP were synthesized, e.g. [stearyl-norleucine-17] VIP (SNV); and (2) potent neuroprotective peptide derivatives were identified that mimic the activity of VIP-responsive neuroprotective glial proteins. VIP provides neuronal defense by inducing the synthesis and secretion of neuroprotective proteins from astrocytes; activity-dependent neuroprotective protein (ADNP) was discovered as such glial cell mediator of VIP- and SNV-induced neuroprotection. In subsequent studies, an eight-amino-acid peptide, NAP, was identified as the smallest active element of ADNP exhibiting potent neuroprotective activities. This paper summarizes the biological effects of SNV and NAP and further reports advances in NAP studies toward clinical development. An original finding described here shows that NAP, while protecting neurons, demonstrated no apparent effect on cell division in a multiplicity of cell lines, strengthening the notion that NAP is a specific neuroprotective drug candidate.
Collapse
Affiliation(s)
- Illana Gozes
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | | | | | | | |
Collapse
|
24
|
Kim MJ, Joo KM, Chung YH, Lee YJ, Kim J, Lee BH, Shin DH, Lee KH, Cha CI. Vasoactive intestinal peptide (VIP) and VIP mRNA decrease in the cerebral cortex of nNOS knock-out(-/-) mice. Brain Res 2003; 978:233-40. [PMID: 12834919 DOI: 10.1016/s0006-8993(03)02950-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although there is much evidence showing that NO regulates the release of VIP in several areas, there is no report about the influence of NO on VIP in the cerebral cortex. We therefore examined changes in VIP expression in the cerebral cortex of nNOS knock-out(-/-) mice using immunohistochemistry and in situ hybridization. The nNOS((-/-)) mice had significantly fewer VIP-immunoreactive neurons than the control mice and the VIP mRNA as well as the VIP-immunoreactivity of the individual neuron was decreased in the nNOS((-/-)) mice. The first demonstration of decrease in VIP expression in the cerebral cortex of nNOS((-/-)) mice may provide useful data for investigating the relation between NO and VIP in the cerebral cortex and the mechanisms of many functions of these two neurotransmitters.
Collapse
Affiliation(s)
- Myeung Ju Kim
- Department of Anatomy, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul 110-799, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wilkemeyer MF, Chen SY, Menkari CE, Brenneman DE, Sulik KK, Charness ME. Differential effects of ethanol antagonism and neuroprotection in peptide fragment NAPVSIPQ prevention of ethanol-induced developmental toxicity. Proc Natl Acad Sci U S A 2003; 100:8543-8. [PMID: 12808140 PMCID: PMC166265 DOI: 10.1073/pnas.1331636100] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
NAPVSIPQ (NAP), an active fragment of the glial-derived activity-dependent neuroprotective protein, is protective at femtomolar concentrations against a wide array of neural insults and prevents ethanol-induced fetal wastage and growth retardation in mice. NAP also antagonizes ethanol inhibition of L1-mediated cell adhesion (ethanol antagonism). We performed an Ala scanning substitution of NAP to determine the role of ethanol antagonism and neuroprotection in NAP prevention of ethanol embryotoxicity. The Ser-Ile-Pro region of NAP was crucial for both ethanol antagonism and protection of cortical neurons from tetrodotoxin toxicity (neuroprotection). Ala replacement of either Ser-5 or Pro-7 (P7A-NAP) abolished NAP neuroprotection but minimally changed the efficacy of NAP ethanol antagonism. In contrast, Ala replacement of Ile-6 (I6A-NAP) caused a decrease in potency (>2 logarithmic orders) with only a small reduction (<10%) in the efficacy of NAP neuroprotection but markedly reduced the efficacy (50%) and the potency (5 logarithmic orders) of NAP ethanol antagonism. Ethanol significantly reduced the number of paired somites in mouse whole-embryo culture; this effect was prevented significantly by 100 pM NAP or by 100 pM P7A-NAP, but not by 100 pM I6A-NAP. The structure-activity relation for NAP prevention of ethanol embryotoxicity was similar to that for NAP ethanol antagonism and different from that for NAP neuroprotection. These findings support the hypothesis that NAP antagonism of ethanol inhibition of L1 adhesion plays a central role in NAP prevention of ethanol embryotoxicity and highlight the potential importance of ethanol effects on L1 in the pathophysiology of fetal alcohol syndrome.
Collapse
Affiliation(s)
- Michael F. Wilkemeyer
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Shao-yu Chen
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Carrie E. Menkari
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Douglas E. Brenneman
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Kathleen K. Sulik
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Michael E. Charness
- Neurology Service, Veterans Affairs Boston
Healthcare System, West Roxbury, MA 02132;
Department of Neurology, Harvard Medical
School, Boston, MA 02115; Department of
Neurology, Brigham and Women's Hospital, Boston, MA 02115;
Bowles Center for Alcohol Studies, University of
North Carolina, Chapel Hill, NC 27599; Department
of Cell and Developmental Biology, University of North Carolina School of
Medicine, Chapel Hill, NC 27599; and Section on
Developmental and Molecular Pharmacology, National Institute of Child Health
and Human Development, National Institutes of Health, Bethesda, MD 20892
- To whom correspondence should be addressed at: Department of Neurology (127),
Harvard Medical School, Veterans Affairs Boston Healthcare System, 1400 VFW
Parkway, West Roxbury, MA 02132. E-mail:
| |
Collapse
|
26
|
Sandgren K, Lin Z, Fex Svenningsen A, Ekblad E. Vasoactive intestinal peptide and nitric oxide promote survival of adult rat myenteric neurons in culture. J Neurosci Res 2003; 72:595-602. [PMID: 12749024 DOI: 10.1002/jnr.10612] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Several motility disorders originate in the enteric nervous system (ENS). Our knowledge of factors governing survival of the ENS is poor. Changes in the expression of vasoactive intestinal peptide (VIP) and nitric oxide synthase (NOS) in enteric neurons occur after neuronal injury and in intestinal adaptation. The aim of this study was to evaluate whether VIP and nitric oxide (NO) influence survival of cultured, dissociated myenteric neurons. Neuronal survival was evaluated after 0, 4, and 8 days in culture. Influence of VIP and NO on neuronal survival was examined after culturing in the presence of VIP, NO donor, VIP antiserum, or NOS inhibitor. A marked loss of neurons was noted during culturing. VIP and NO significantly promoted neuronal survival. Corroborating this was the finding of an enhanced neuronal cell loss when cultures were grown in the presence of VIP antiserum or NOS inhibitor.
Collapse
Affiliation(s)
- Katarina Sandgren
- Department of Physiological Sciences, Neuroendocrine Cell Biology, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
27
|
Wilkemeyer MF, Menkari CE, Spong CY, Charness ME. Peptide antagonists of ethanol inhibition of l1-mediated cell-cell adhesion. J Pharmacol Exp Ther 2002; 303:110-6. [PMID: 12235240 DOI: 10.1124/jpet.102.036277] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ethanol inhibits cell-cell adhesion mediated by the L1 cell adhesion molecule. 1-Octanol potently antagonizes this cellular action of ethanol and also prevents ethanol-induced dysmorphology and cell death in mouse whole embryo culture. NAPVSIPQ (NAP) and SALLRSIPA (SAL) are active peptide fragments of two neuroprotective proteins: activity-dependent neuroprotective protein and activity-dependent neurotrophic factor. NAP and SAL are neuroprotective at femtomolar concentrations against a variety of neurotoxins and also prevent ethanol teratogenesis in mice. To explore the cellular basis for this action, we asked whether NAP and SAL antagonize ethanol inhibition of L1 adhesion. Aggregation assays were carried out in ethanol-sensitive, human L1-transfected NIH/3T3 cells in the absence and presence of NAP and SAL. Neither NAP nor SAL altered L1 adhesion or L1 expression; however, both peptides potently and completely antagonized the inhibition of L1 adhesion by 100 mM ethanol (EC(50): NAP, 6 x 10(-14) M; SAL, 4 x 10(-11) M). NAP also antagonized ethanol inhibition of cell-cell adhesion in bone morphogenetic protein-7-treated NG108-15 cells. In L1-expressing NIH/3T3 cells, SAL antagonism was reversible and could be overcome by increasing concentrations of ethanol. In contrast, NAP antagonism was irreversible and could not be overcome by increasing agonist concentration. Two scrambled NAP peptides (ASPNQPIV and PNIQVASP) were not antagonists at concentrations as high as 10(-7) M. Thus, two structurally unrelated classes of compounds, alcohols and small polypeptides, share two common actions: antagonism of ethanol inhibition of L1-mediated cell adhesion and prevention of ethanol teratogenesis. These findings support the hypothesis that ethanol inhibition of L1 adhesion contributes to ethanol teratogenesis.
Collapse
Affiliation(s)
- Michael F Wilkemeyer
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
28
|
|
29
|
Gozes I, Alcalay R, Giladi E, Pinhasov A, Furman S, Brenneman DE. NAP accelerates the performance of normal rats in the water maze. J Mol Neurosci 2002; 19:167-70. [PMID: 12212775 DOI: 10.1007/s12031-002-0028-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
NAP (Asn-Ala-Pro-Val-Ser-Ile-Pro-Gln) has neuroprotective, memory enhancing, and neurotrophic properties. NAP is a short peptide sequence derived from the recently cloned, activity-dependent neuroprotective protein. The current study was designed to evaluate NAP activity in normal middle-aged animals to further assess NAP's breadth of neuroprotection. NAP was administered by inhalation. Results showed that in the paradigm of the Morris water maze, assessing short-term memory, only the NAP-treated middle-aged rats and not placebo-treated rats showed significant improvements by the end of the testing period. These results suggest efficacy for NAP in normal aging that is associated with accumulating environmental and genetic toxic factors.
Collapse
Affiliation(s)
- Illana Gozes
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Israel.
| | | | | | | | | | | |
Collapse
|
30
|
Leker RR, Teichner A, Grigoriadis N, Ovadia H, Brenneman DE, Fridkin M, Giladi E, Romano J, Gozes I. NAP, a femtomolar-acting peptide, protects the brain against ischemic injury by reducing apoptotic death. Stroke 2002; 33:1085-92. [PMID: 11935065 DOI: 10.1161/01.str.0000014207.05597.d7] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE We sought to determine the cerebroprotective potential of NAP, a synthetic octapeptide related to vasoactive intestinal peptide. Activity-dependent neuroprotective protein mediates some of the protective effects of vasoactive intestinal peptide. The neuroprotective NAP sequence is derived from activity-dependent neuroprotective protein. METHODS Spontaneously hypertensive rats underwent permanent middle cerebral artery occlusion by craniotomy and electrocoagulation. After dose-response and time-course experiments, the animals were injected with NAP (3 microg/kg) or vehicle intravenously 1 hour after stroke onset. Another group of rats was injected with the D-amino acid isomer of NAP (D-NAP) and served as a negative control. Rats were examined for motor and behavioral deficits 24 hours to 30 days later, and infarct volumes were determined. The effect of NAP administration on apoptotic death was determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and caspase-3 stainings. RESULTS NAP significantly reduced motor disability and infarct volumes compared with vehicle or D-NAP when tested at 24 hours after stroke onset (9.67+/-1.4% versus 17.04+/-1.18% and 19.19+/-1.9% of hemispheric volume, respectively; P<0.05). NAP given 4 but not 6 hours after permanent middle cerebral artery occlusion still conferred significant neuroprotection (infarct volume 10.9+/-3.9% of hemispheric volume; P<0.05 versus vehicle). Long-term studies demonstrated that infarct volumes and disability scores remained significantly lower after 30 days in NAP-treated animals. NAP significantly reduced the number of apoptotic cells. CONCLUSIONS Our results indicate that the durable cerebroprotection by NAP involves antiapoptotic mechanisms.
Collapse
Affiliation(s)
- Ronen R Leker
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hebrew University-Hadassah Medical School, University Hospital, Jerusalem, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gozes I, Brenneman DE, Geppetti P, Kastin AJ, Mains RE, Moody TW, Seroogy K, Spier AD, Zimmermann M. Neuropeptides: brain messengers of many faces. Trends Neurosci 2001; 24:687-90. [PMID: 11718856 DOI: 10.1016/s0166-2236(00)02001-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Neuropeptides 2001, 2nd Joint Meeting of the European Neuropeptide Club and the American Summer Neuropeptide Conference (11th Annual Meeting). 6-11 May 2001 with Satellite Symposium, Israeli-French Symposium, Israel Ministry of Science, Culture and Sport, 6 May 2001, held at Maale Hachmicha and Tel Aviv University, Israel.
Collapse
Affiliation(s)
- I Gozes
- Dept of Clinical Biochemnistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|