Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Physiol Rep ; 10(13): e15368, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35822260

RESUMO

Infants with neonatal cholestasis are prone to neurodevelopmental deficits, however, the underlying pathogenesis is unclear. Lipid malabsorption and accumulation of potentially neurotoxic molecules in the blood such as bile acids are important yet relatively unexplored pathways. Here, we developed a translational piglet model to understand how the molecular bile acid and lipid composition of the brain is affected by this disease and relates to motor function. Piglets (8-days old) had bile duct ligation or sham surgery and were fed a formula diet for 3 weeks. Alongside sensory-motor deficits observed in bile duct-ligated animals, we found a shift toward a more hydrophilic and conjugated bile acid profile in the brain. Additionally, comprehensive lipidomics of the cerebellum revealed a decrease in total lipids including phosphatidylinositols and phosphatidylserines and increases in lysophospholipid species. This was paralleled by elevated cerebellar expression of genes related to inflammation and tissue damage albeit without significant impact on the brain transcriptome. This study offers new insights into the developing brain's molecular response to neonatal cholestasis indicating that bile acids and lipids may contribute in mediating motor deficits.


Assuntos
Ácidos e Sais Biliares , Colestase , Animais , Ductos Biliares/metabolismo , Encéfalo/metabolismo , Colestase/metabolismo , Humanos , Lipídeos , Suínos
2.
Am J Physiol Cell Physiol ; 321(5): C770-C778, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34495765

RESUMO

Skeletal muscle is an endocrine organ secreting exercise-induced factors (exerkines), which play a pivotal role in interorgan cross talk. Using mass spectrometry (MS)-based proteomics, we characterized the secretome and identified thymosin ß4 (TMSB4X) as the most upregulated secreted protein in the media of contracting C2C12 myotubes. TMSB4X was also acutely increased in the plasma of exercising humans irrespective of the insulin resistance condition or exercise mode. Treatment of mice with TMSB4X did not ameliorate the metabolic disruptions associated with diet induced-obesity, nor did it enhance muscle regeneration in vivo. However, TMSB4X increased osteoblast proliferation and neurite outgrowth, consistent with its WADA classification as a prohibited growth factor. Therefore, we report TMSB4X as a human exerkine with a potential role in cellular cross talk.


Assuntos
Proliferação de Células/efeitos dos fármacos , Contração Muscular , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Crescimento Neuronal/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Timosina/metabolismo , Timosina/farmacologia , Animais , Estudos de Casos e Controles , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Humanos , Resistência à Insulina , Masculino , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Doenças Musculares/fisiopatologia , Osteoblastos/patologia , Resistência Física , Proteômica , Transdução de Sinais , Espectrometria de Massas em Tandem
3.
Sci Rep ; 10(1): 20675, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33244084

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating incurable neurological disorder characterized by motor neuron (MN) death and muscle dysfunction leading to mean survival time after diagnosis of only 2-5 years. A potential ALS treatment is to delay the loss of MNs and disease progression by the delivery of trophic factors. Previously, we demonstrated that implanted mesoporous silica nanoparticles (MSPs) loaded with trophic factor peptide mimetics support survival and induce differentiation of co-implanted embryonic stem cell (ESC)-derived MNs. Here, we investigate whether MSP loaded with peptide mimetics of ciliary neurotrophic factor (Cintrofin), glial-derived neurotrophic factor (Gliafin), and vascular endothelial growth factor (Vefin1) injected into the cervical spinal cord of mutant SOD1 mice affect disease progression and extend survival. We also transplanted boundary cap neural crest stem cells (bNCSCs) which have been shown previously to have a positive effect on MN survival in vitro and in vivo. We show that mimetic-loaded MSPs and bNCSCs significantly delay disease progression and increase survival of mutant SOD1 mice, and also that empty particles significantly improve the condition of ALS mice. Our results suggest that intraspinal delivery of MSPs is a potential therapeutic approach for the treatment of ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/patologia , Sobrevivência Celular/efeitos dos fármacos , Dióxido de Silício/farmacologia , Esclerose Lateral Amiotrófica/metabolismo , Animais , Células Cultivadas , Medula Cervical/efeitos dos fármacos , Medula Cervical/metabolismo , Medula Cervical/patologia , Modelos Animais de Doenças , Progressão da Doença , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Camundongos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Crista Neural/efeitos dos fármacos , Crista Neural/metabolismo , Crista Neural/patologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
ACS Chem Neurosci ; 11(9): 1270-1282, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32283014

RESUMO

Vascular endothelial growth factor B (VEGFB) is a pleiotropic trophic factor, which in contrast to the closely related VEGFA is known to have a limited effect on angiogenesis. VEGFB improves survival in various tissues including the nervous system, where the effect was observed mainly for peripheral neurons. The neurotrophic effect of VEGFB on central nervous system neurons has been less investigated. Here we demonstrated that VEGFB promotes neurite outgrowth from primary cerebellar granule, hippocampal, and retinal neurons in vitro. VEGFB protected hippocampal and retinal neurons from both oxidative stress and glutamate-induced neuronal death. The VEGF receptor 1 (VEGFR1) is required for VEGFB-induced neurotrophic and neuroprotective effects. Using a structure-based approach, we designed short peptides, termed Vefin1-7, mimicking the binding interface of VEGFB to VEGFR1. Vefins were analyzed for their secondary structure and binding to VEGF receptors and compared with previously described peptides derived from VEGFA, another ligand of VEGFR1. We show that Vefins have neurotrophic and neuroprotective effects on primary hippocampal, cerebellar granule, and retinal neurons in vitro with potencies comparable to VEGFB. Similar to VEGFB, Vefins were not mitogenic for MCF-7 cancer cells. Furthermore, one of the peptides, Vefin7, even dose-dependently inhibited the proliferation of MCF-7 cells in vitro. Unraveling the neurotrophic and neuroprotective potentials of VEGFB, the only nonangiogenic factor of the VEGF family, is promising for the development of neuroprotective peptide-based therapies.


Assuntos
Fator B de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Sistema Nervoso Central , Neurônios , Peptídeos/farmacologia
5.
Neurobiol Aging ; 81: 88-101, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31255922

RESUMO

Neurodegenerative disorders such as Alzheimer's disease (AD) are characterized by the irreversible neuronal loss and memory impairment, and current treatments are merely symptomatic. Erythropoietin (EPO) has been shown to possess neurotrophic, neuroprotective, anti-inflammatory, and memory-enhancing effects, which could be therapeutically beneficial in the different aspects of AD. However, the hematopoietic effect of EPO has hampered its potential as a neuroprotective and procognitive agent. In this study, we characterized a novel small peptide, NL100, derived from a conserved C-helix region of EPO. NL100 was shown to bind to the EPO receptor, induce neuritogenesis, and protect hippocampal neurons from oxidative- and Aß25-35-induced neurodegeneration in vitro. Importantly, long-term NL100 treatment did not induce hematopoiesis, overcoming this challenge associated with EPO. Memory-enhancing effects were demonstrated after NL100 treatment in social recognition test for short-term memory, in both healthy rats and rats challenged centrally with Aß25-35 peptide, and in the Morris water maze test for spatial memory. Moreover, NL100 was shown to reverse Aß25-35-induced hippocampal degeneration and gliosis as well as pilocarpine-induced suppression of long-term potentiation in rats. In conclusion, NL100 is a novel EPO-derived nonhematopoietic peptide with neuroprotective and memory-enhancing effects and could therefore be a potential candidate for the development of new treatments for neurodegenerative disorders and dementia.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Demência/tratamento farmacológico , Demência/etiologia , Eritropoetina , Potenciação de Longa Duração/efeitos dos fármacos , Memória/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/etiologia , Fármacos Neuroprotetores , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Animais , Eritropoetina/química , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos Endogâmicos BALB C , Crescimento Neuronal/efeitos dos fármacos , Ratos Sprague-Dawley , Ratos Wistar
6.
Am J Physiol Gastrointest Liver Physiol ; 317(1): G67-G77, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31091150

RESUMO

Prenatal inflammation may predispose to preterm birth and postnatal inflammatory disorders such as necrotizing enterocolitis (NEC). Bioactive milk ingredients may help to support gut maturation in such neonates, but mother's milk is often insufficient after preterm birth. We hypothesized that supplementation with bioactive ingredients from bovine milk [osteopontin (OPN), caseinoglycomacropeptide (CGMP), colostrum (COL)] supports gut, immunity, and NEC resistance in neonates born preterm after gram-negative infection before birth. Using preterm pigs as a model for preterm infants, fetal pigs were given intraamniotic injections of lipopolysaccharide (LPS; 1 mg/fetus) and delivered 3 days later (90% gestation). For 5 days, groups of LPS-exposed pigs were fed formula (FOR), bovine colostrum (COL), or formula enriched with OPN or CGMP. LPS induced intraamniotic inflammation and postnatal systemic inflammation but limited effects on postnatal gut parameters and NEC. Relative to FOR, COL feeding to LPS-exposed pigs showed less diarrhea, NEC severity, reduced gut IL-1ß and IL-8 levels, greater gut goblet cell density and digestive enzyme activities, and blood helper T-cell fraction. CGMP improved neonatal arousal and gut lactase activities and reduced LPS-induced IL-8 secretion in intestinal epithelial cells (IECs) in vitro. Finally, OPN tended to reduce diarrhea and stimulated IEC proliferation in vitro. No effects on villus morphology, circulating cytokines, or colonic microbiota were observed among groups. In conclusion, bioactive milk ingredients exerted only modest effects on gut and systemic immune parameters in preterm pigs exposed to prenatal inflammation. Short-term, prenatal exposure to inflammation may render the gut less sensitive to immune-modulatory milk effects. NEW & NOTEWORTHY Prenatal inflammation is a risk factor for preterm birth and postnatal complications including infections. However, from clinical studies, it is difficult to separate the effects of only prenatal inflammation from preterm birth. Using cesarean-delivered preterm pigs with prenatal inflammation, we documented some beneficial gut effects of bioactive milk diets relative to formula, but prenatal inflammation appeared to decrease the sensitivity of enteral feeding. Special treatments and diets may be required for this neonatal population.


Assuntos
Caseínas/administração & dosagem , Corioamnionite/dietoterapia , Enterocolite Necrosante/prevenção & controle , Alimentos Fortificados , Imunidade nas Mucosas , Fórmulas Infantis , Intestinos/imunologia , Osteopontina/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Nascimento Prematuro , Animais , Animais Recém-Nascidos , Caseínas/imunologia , Linhagem Celular , Corioamnionite/induzido quimicamente , Corioamnionite/imunologia , Corioamnionite/metabolismo , Colostro/imunologia , Modelos Animais de Doenças , Enterocolite Necrosante/etiologia , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Microbioma Gastrointestinal , Idade Gestacional , Humanos , Recém-Nascido , Absorção Intestinal , Intestinos/microbiologia , Intestinos/patologia , Lipopolissacarídeos , Valor Nutritivo , Osteopontina/imunologia , Fragmentos de Peptídeos/imunologia , Permeabilidade , Gravidez , Sus scrofa
7.
Theranostics ; 8(14): 3977-3990, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30083275

RESUMO

Understanding the mechanisms of neurodegeneration is crucial for development of therapies to treat neurological disorders. S100 proteins are extensively expressed in the injured brain but S100's role and signalling in neural cells remain elusive. We recently demonstrated that the S100A4 protein protects neurons in brain injury and designed S100A4-derived peptides mimicking its beneficial effects. Here we show that neuroprotection by S100A4 involves the growth factor family receptor ErbB4 and its ligand Neuregulin 1 (NRG), key regulators of neuronal plasticity and implicated in multiple brain pathologies. The neuroprotective effect of S100A4 depends on ErbB4 expression and the ErbB4 signalling partners ErbB2/Akt, and is reduced by functional blockade of NRG/ErbB4 in cell models of neurodegeneration. We also detect binding of S100A4 with ErbB1 (EGFR) and ErbB3. S100A4-derived peptides interact with, and signal through ErbB, are neuroprotective in primary and immortalized dopaminergic neurons, and do not affect cell proliferation/motility - features which make them promising as potential neuroprotectants. Our data suggest that the S100-ErbB axis may be an important mechanism regulating neuronal survival and plasticity.


Assuntos
Neurônios/fisiologia , Receptor ErbB-4/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Transdução de Sinais , Animais , Sobrevivência Celular , Neuregulina-1/metabolismo , Plasticidade Neuronal , Ratos
8.
J Neuroinflammation ; 15(1): 180, 2018 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-29885660

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) is an acute gut inflammatory disorder that occurs in preterm infants in the first weeks after birth. Infants surviving NEC often show impaired neurodevelopment. The mechanisms linking NEC lesions with later neurodevelopment are poorly understood but may include proinflammatory signaling in the immature brain. Using preterm pigs as a model for preterm infants, we hypothesized that severe intestinal NEC lesions are associated with acute effects on the developing hippocampus. METHODS: Cesarean-delivered preterm pigs (n = 117) were reared for 8 days and spontaneously developed variable severity of NEC lesions. Neonatal arousal, physical activity, and in vitro neuritogenic effects of cerebrospinal fluid (CSF) were investigated in pigs showing NEC lesions in the colon (Co-NEC) or in the small intestine (Si-NEC). Hippocampal transcriptome analysis and qPCR were used to assess gene expressions and their relation to biological processes, including neuroinflammation, and neural plasticity. Microglia activation was quantified by stereology. The neuritogenic response to selected proteins was investigated in primary cultures of hippocampal neurons. RESULTS: NEC development rapidly reduced the physical activity of pigs, especially when lesions occurred in the small intestine. Si-NEC and Co-NEC were associated with 27 and 12 hippocampal differentially expressed genes (DEGs), respectively. These included genes related to neuroinflammation (i.e., S100A8, S100A9, IL8, IL6, MMP8, SAA, TAGLN2) and hypoxia (i.e., PDK4, IER3, TXNIP, AGER), and they were all upregulated in Si-NEC pigs. Genes related to protection against oxidative stress (HBB, ALAS2) and oligodendrocytes (OPALIN) were downregulated in Si-NEC pigs. CSF collected from NEC pigs promoted neurite outgrowth in vitro, and the S100A9 and S100A8/S100A9 proteins may mediate the neuritogenic effects of NEC-related CSF on hippocampal neurons. NEC lesions did not affect total microglial cell number but markedly increased the proportion of Iba1-positive amoeboid microglial cells. CONCLUSIONS: NEC lesions, especially when present in the small intestine, are associated with changes to hippocampal gene expression that potentially mediate neuroinflammation and disturbed neural circuit formation via enhanced neuronal differentiation. Early brain-protective interventions may be critical for preterm infants affected by intestinal NEC lesions to reduce their later neurological dysfunctions.


Assuntos
Encéfalo/fisiopatologia , Citocinas/metabolismo , Enterocolite Necrosante/etiologia , Nascimento Prematuro/patologia , Nascimento Prematuro/fisiopatologia , Animais , Encéfalo/patologia , Líquido Cefalorraquidiano/metabolismo , Proteínas de Ligação a DNA/metabolismo , Trato Gastrointestinal/metabolismo , Hipóxia/metabolismo , Inflamação/etiologia , Microglia/metabolismo , Microglia/patologia , Proteínas do Tecido Nervoso/metabolismo , Crescimento Neuronal , Condicionamento Físico Animal , Proteínas S100/metabolismo , Suínos , Fatores de Tempo , Transcriptoma/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Stem Cells Dev ; 26(14): 1065-1077, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28562227

RESUMO

Spinal root injuries result in newly formed glial scar formation, which prevents regeneration of sensory axons causing permanent sensory loss. Previous studies showed that delivery of trophic factors or implantation of human neural progenitor cells supports sensory axon regeneration and partly restores sensory functions. In this study, we elucidate mechanisms underlying stem cell-mediated ingrowth of sensory axons after dorsal root avulsion (DRA). We show that human spinal cord neural stem/progenitor cells (hscNSPC), and also, mesoporous silica particles loaded with growth factor mimetics (MesoMIM), supported sensory axon regeneration. However, when hscNSPC and MesoMIM were combined, sensory axon regeneration failed. Morphological and tracing analysis showed that sensory axons grow through the newly established glial scar along "bridges" formed by migrating stem cells. Coimplantation of MesoMIM prevented stem cell migration, "bridges" were not formed, and sensory axons failed to enter the spinal cord. MesoMIM applied alone supported sensory axons ingrowth, but without affecting glial scar formation. In vitro, the presence of MesoMIM significantly impaired migration of hscNSPC without affecting their level of differentiation. Our data show that (1) the ability of stem cells to migrate into the spinal cord and organize cellular "bridges" in the newly formed interface is crucial for successful sensory axon regeneration, (2) trophic factor mimetics delivered by mesoporous silica may be a convenient alternative way to induce sensory axon regeneration, and (3) a combinatorial approach of individually beneficial components is not necessarily additive, but can be counterproductive for axonal growth.


Assuntos
Axônios/patologia , Regeneração Nervosa , Traumatismos da Medula Espinal/fisiopatologia , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Raízes Nervosas Espinhais/patologia , Raízes Nervosas Espinhais/fisiopatologia , Animais , Diferenciação Celular , Movimento Celular , Cistos Glanglionares/patologia , Humanos , Camundongos , Células-Tronco Neurais/transplante , Neuroglia/patologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco
10.
Mediators Inflamm ; 2016: 1346390, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27990061

RESUMO

The cytokine erythropoietin (EPO) stimulates proliferation and differentiation of erythroid progenitor cells. Moreover, EPO has neuroprotective, anti-inflammatory, and antioxidative effects, but the use of EPO as a neuroprotective agent is hampered by its erythropoietic activity. We have recently designed the synthetic, dendrimeric peptide, Epobis, derived from the sequence of human EPO. This peptide binds the EPO receptor and promotes neuritogenesis and neuronal cell survival. Here we demonstrate that Epobis in vitro promotes neuritogenesis in primary motoneurons and has anti-inflammatory effects as demonstrated by its ability to decrease TNF release from activated AMJ2-C8 macrophages and rat primary microglia. When administered systemically Epobis is detectable in both plasma and cerebrospinal fluid, demonstrating that the peptide crosses the blood-brain barrier. Importantly, Epobis is not erythropoietic, but systemic administration of Epobis in rats delays the clinical signs of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, and the peptide has long-term, but not short-term, effects on working memory, detected as an improved social memory 3 days after administration. These data reveal Epobis to be a nonerythropoietic and neuroprotective EPO receptor agonist with anti-inflammatory and memory enhancing properties.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células Cultivadas , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/tratamento farmacológico , Eritropoetina/química , Feminino , Hematopoese/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Memória de Curto Prazo/efeitos dos fármacos , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Neuritos/efeitos dos fármacos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Peptídeos/uso terapêutico , Ratos , Ratos Wistar
11.
J Neuroinflammation ; 11: 27, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24490798

RESUMO

BACKGROUND: Interleukin 1 (IL-1) is implicated in neuroinflammation, an essential component of neurodegeneration. We evaluated the potential anti-inflammatory effect of a novel peptide antagonist of IL-1 signaling, Ilantide. METHODS: We investigated the binding of Ilantide to IL-1 receptor type I (IL-1RI) using surface plasmon resonance, the inhibition of Il-1ß-induced activation of nuclear factor κB (NF-κB) in HEK-Blue cells that contained an IL-1ß-sensitive reporter, the secretion of TNF-α in macrophages, protection against IL-1-induced apoptosis in neonatal pancreatic islets, and the penetration of Ilantide through the blood-brain barrier using competitive enzyme-linked immunosorbent assay (ELISA). We studied the effects of the peptide on social behavior and memory in rat models of lipopolysaccharide (LPS)- and amyloid-induced neuroinflammation, respectively, and its effect in a rat model of experimental autoimmune enchephalomyelitis. RESULTS: Ilantide bound IL-1RI, inhibited the IL-1ß-induced activation of NF-κB, and inhibited the secretion of TNF-α in vitro. Ilantide protected pancreatic islets from apoptosis in vitro and reduced inflammation in an animal model of arthritis. The peptide penetrated the blood-brain barrier. It reduced the deficits in social activity and memory in LPS- and amyloid-treated animals and delayed the development of experimental autoimmune enchephalomyelitis. CONCLUSIONS: These findings indicate that Ilantide is a novel and potent IL-1RI antagonist that is able to reduce inflammatory damage in the central nervous system and pancreatic islets.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Artrite/induzido quimicamente , Células Cultivadas , Cerebelo/citologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Humanos , Proteína Antagonista do Receptor de Interleucina 1/química , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Lipopolissacarídeos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Comportamento Social , Transfecção , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
12.
Neurochem Res ; 38(12): 2550-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24132641

RESUMO

ErbB receptors not only function in cancer, but are also key developmental regulators in the nervous system. We previously identified an ErbB1 peptide antagonist, Inherbin3, that is capable of inhibiting tumor growth in vitro and in vivo. In this study, we found that inhibition of ErbB1 kinase activity and activation of ErbB4 by NRG-1ß induced neurite extension, suggesting that ErbB1 and ErbB4 act as negative and positive regulators, respectively, of the neuritogenic response. Inherbin3, inhibited activation not only of ErbB1 but also of ErbB4 in primary neurons, strongly induced neurite outgrowth in rat cerebellar granule neurons, indicating that this effect mainly was due to inhibition of ErbB1 activation.


Assuntos
Cerebelo/efeitos dos fármacos , Grânulos Citoplasmáticos/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Neuritos/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Sequência de Bases , Células Cultivadas , Cerebelo/citologia , Primers do DNA , Receptores ErbB/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Fosforilação , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
13.
Mol Med ; 19: 43-53, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23508572

RESUMO

We recently found that S100A4, a member of the multifunctional S100 protein family, protects neurons in the injured brain and identified two sequence motifs in S100A4 mediating its neurotrophic effect. Synthetic peptides encompassing these motifs stimulated neuritogenesis and survival in vitro and mimicked the S100A4-induced neuroprotection in brain trauma. Here, we investigated a possible function of S100A4 and its mimetics in the pathologies of the peripheral nervous system (PNS). We found that S100A4 was expressed in the injured PNS and that its peptide mimetic (H3) affected the regeneration and survival of myelinated axons. H3 accelerated electrophysiological, behavioral and morphological recovery after sciatic nerve crush while transiently delaying regeneration after sciatic nerve transection and repair. On the basis of the finding that both S100A4 and H3 increased neurite branching in vitro, these effects were attributed to the modulatory effect of H3 on initial axonal sprouting. In contrast to the modest effect of H3 on the time course of regeneration, H3 had a long-term neuroprotective effect in the myelin protein P0 null mice, a model of dysmyelinating neuropathy (Charcot-Marie-Tooth type 1 disease), where the peptide attenuated the deterioration of nerve conduction, demyelination and axonal loss. From these results, S100A4 mimetics emerge as a possible means to enhance axonal sprouting and survival, especially in the context of demyelinating neuropathies with secondary axonal loss, such as Charcot-Marie-Tooth type 1 disease. Moreover, our data suggest that S100A4 is a neuroprotectant in PNS and that other S100 proteins, sharing high homology in the H3 motif, may have important functions in PNS pathologies.


Assuntos
Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Proteínas S100/farmacologia , Nervo Isquiático/efeitos dos fármacos , Animais , Células Cultivadas , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Doença de Charcot-Marie-Tooth/fisiopatologia , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína P0 da Mielina/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Peptídeos/uso terapêutico , Ratos , Ratos Wistar , Proteínas S100/uso terapêutico , Nervo Isquiático/lesões , Nervo Isquiático/fisiopatologia , Nervo Tibial/efeitos dos fármacos , Nervo Tibial/fisiopatologia
14.
Nat Commun ; 3: 1197, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23149742

RESUMO

Identification of novel pro-survival factors in the brain is paramount for developing neuroprotective therapies. The multifunctional S100 family proteins have important roles in many human diseases and are also upregulated by brain injury. However, S100 functions in the nervous system remain unclear. Here we show that the S100A4 protein, mostly studied in cancer, is overexpressed in the damaged human and rodent brain and released from stressed astrocytes. Genetic deletion of S100A4 exacerbates neuronal loss after brain trauma or excitotoxicity, increasing oxidative cell damage and downregulating the neuroprotective protein metallothionein I+II. We identify two neurotrophic motifs in S100A4 and show that these motifs are neuroprotective in animal models of brain trauma. Finally, we find that S100A4 rescues neurons via the Janus kinase/STAT pathway and, partially, the interleukin-10 receptor. Our data introduce S100A4 as a therapeutic target in neurodegeneration, and raise the entire S100 family as a potentially important factor in central nervous system injury.


Assuntos
Citoproteção , Metástase Neoplásica/patologia , Neurônios/patologia , Proteínas S100/metabolismo , Motivos de Aminoácidos , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Morte Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Feminino , Deleção de Genes , Células HEK293 , Humanos , Janus Quinases/metabolismo , Ácido Caínico , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Ratos , Receptores de Interleucina-10/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/química , Fatores de Transcrição STAT/metabolismo , Convulsões/tratamento farmacológico , Convulsões/patologia , Regulação para Cima/efeitos dos fármacos
15.
J Neurochem ; 121(6): 915-23, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22469063

RESUMO

Apart from its hematopoietic activity, erythropoietin (EPO) is also known as a tissue-protective cytokine. In the brain, EPO and its receptor are up-regulated in response to insult and exert pro-survival effects. EPO binds to its receptor (EPOR) via high- and low-affinity binding sites (Sites 1 and 2, respectively), inducing conformational changes in the receptor, followed by the activation of downstream signaling cascades. Based on the crystal structure of the EPO:EPOR(2) complex, we designed a peptide, termed Epobis, whose sequence encompassed amino acids from binding Site 1. The present study shows that the Epobis peptide specifically binds to EPOR and induces neurite outgrowth from primary neurons in an EPOR-expression dependent manner. Furthermore, Epobis promoted the survival of hippocampal and cerebellar neuronal cultures after kainate treatment and KCl deprivation, respectively. Thus, we identified a new functional agonist of EPOR with the potential to promote neuroregeneration and neuroprotection.


Assuntos
Neuritos/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Receptores da Eritropoetina/agonistas , Receptores da Eritropoetina/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Eritropoetina/química , Eritropoetina/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Modelos Moleculares , Fármacos Neuroprotetores/metabolismo , Peptídeos/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Ressonância de Plasmônio de Superfície , Transfecção
16.
Eur J Cell Biol ; 90(12): 990-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22000729

RESUMO

Ciliary neurotrophic factor (CNTF) induces neuronal differentiation and promotes the survival of various neuronal cell types by binding to a receptor complex formed by CNTF receptor α (CNTFRα), gp130, and the leukemia inhibitory factor (LIF) receptor (LIFR). The CD loop-D helix region of CNTF has been suggested to be important for the cytokine interaction with LIFR. We designed a peptide, termed cintrofin, that encompasses this region. Surface plasmon resonance analysis demonstrated that cintrofin bound to LIFR and gp130, but not to CNTFRα, with apparent KD values of 35 nM and 1.1 nM, respectively. Cintrofin promoted the survival of cerebellar granule neurons (CGNs), in which cell death was induced either by potassium withdrawal or H2O2 treatment. Cintrofin induced neurite outgrowth from CGNs, and this effect was inhibited by specific antibodies against both gp130 and LIFR, indicating that these receptors are involved in the effects of cintrofin. The C-terminal part of the peptide, corresponding to the D helix region of CNTF, was shown to be essential for the neuritogenic action of the peptide. CNTF and LIF induced neurite outgrowth in CGNs plated on laminin-coated slides. On uncoated slides, CNTF and LIF had no neuritogenic effect but were able to inhibit cintrofin-induced neuronal differentiation, indicating that cintrofin and cytokines compete for the same receptors. In addition, cintrofin induced the phosphorylation of STAT3, Akt, and ERK, indicating that it exerts cell signaling properties similar to those induced by CNTF and may be a valuable survival agent with possible therapeutic potential.


Assuntos
Fator Neurotrófico Ciliar/farmacologia , Fator Inibidor de Leucemia/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Citocinas/metabolismo , Sequência de Aminoácidos , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fator Neurotrófico Ciliar/metabolismo , Humanos , Interleucina-6/metabolismo , Microscopia Confocal , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Transdução de Sinais
17.
J Neurosci Res ; 88(5): 1074-82, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19937811

RESUMO

Metallothioneins I and II (MTI/II) are metal-binding proteins overexpressed in response to brain injury. Recently, we have designed a peptide, termed EmtinB, which is modeled after the beta-domain of MT-II and mimics the biological effects of MTI/II in vitro. Here, we demonstrate the neuroprotective effect of EmtinB in the in vitro and in vivo models of kainic acid (KA)-induced neurotoxicity. We show that EmtinB passes the blood-brain barrier and is detectable in plasma for up to 24 hr. Treatment with EmtinB significantly attenuates seizures in C57BL/6J mice exposed to moderate (20 mg/kg) and high (30 mg/kg) KA doses and tends to decrease mortality induced by the high KA dose. Histopathological evaluation of hippocampal (CA3 and CA1) and cortical areas of mice treated with 20 mg/kg KA shows that EmtinB treatment reduces KA-induced neurodegeneration in the CA1 region. These findings establish EmtinB as a promising target for therapeutic development.


Assuntos
Ácido Caínico/antagonistas & inibidores , Metalotioneína/agonistas , Fármacos Neuroprotetores/farmacocinética , Neurotoxinas/antagonistas & inibidores , Peptídeos/farmacocinética , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular , Ácido Caínico/toxicidade , Masculino , Metalotioneína/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/toxicidade , Peptídeos/sangue , Peptídeos/uso terapêutico , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/fisiopatologia
18.
Int J Oncol ; 36(1): 29-38, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19956830

RESUMO

The Her-2 receptor coded for by the proto-oncogenic erbB-2 gene is a clinically validated target for treatment of a significant genetic subclass of breast cancers, and Her-2 is also overexpressed or mutated in a range of other cancers. In an approach to exploit antisense mediated splicing interference as a means of manipulating erbB-2 expression in a therapeutically relevant fashion, we have studied the effect on mRNA splicing of a series of peptide nucleic acid (PNA) oligomers targeting specific intron-exon junctions in the erbB-2 pre-mRNA. In particular, we are interested in identifying PNA oligomers that specifically induce skipping of exon 19 as this exon is coding for the ATP catalytic domain of Her-2, and if expressed such truncated version of the Her-2 protein should be functionally inactive in a dominant negative fashion. Therefore, antisense compounds having efficient erbB-2 exon 19 skipping activity could be very interesting in terms of drug discovery. In the present study we identified PNA oligomers having such activity in SK-BR-3 and HeLa cancer cells in culture.


Assuntos
Trifosfato de Adenosina/química , Genes erbB-2 , Ácidos Nucleicos Peptídicos/metabolismo , Splicing de RNA , Receptor ErbB-2/metabolismo , Processamento Alternativo , Domínio Catalítico , Linhagem Celular Tumoral , Éxons , Genes Dominantes , Células HeLa , Temperatura Alta , Humanos , Oligonucleotídeos Antissenso , Espectrofotometria Ultravioleta/métodos
19.
Bioconjug Chem ; 19(8): 1526-34, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18646838

RESUMO

Conjugation to cationic cell penetrating peptides (such as Tat, Penetratin, or oligo arginines) efficiently improves the cellular uptake of large hydrophilic molecules such as oligonucleotides and peptide nucleic acids, but the cellular uptake is predominantly via an unproductive endosomal pathway and therefore mechanisms that promote endosomal escape (or avoid the endosomal route) are required for improving bioavailability. A variety of auxiliary agents (chloroquine, calcium ions, or lipophilic photosensitizers) has this effect, but improved, unaided delivery would be highly advantageous in particular for future in vivo applications. We find that simply conjugating a lipid domain (fatty acid) to the cationic peptide (a CatLip conjugate) increases the biological effect of the corresponding PNA (CatLip) conjugates in a luciferase cellular antisense assay up to 2 orders of magnitude. The effect increases with increasing length of the fatty acid (C8-C16) but in parallel also results in increased cellular toxicity, with decanoic acid being optimal. Furthermore, the relative enhancement is significantly higher for Tat peptide compared to oligoarginine. Confocal microscopy and chloroquine enhancement indicates that the lipophilic domain increases the endosomal uptake as well as promoting significantly endosomal escape. These results provide a novel route for improving the (cellular) bioavailability of larger hydrophilic molecules.


Assuntos
Células/metabolismo , Lipídeos/química , Oligonucleotídeos Antissenso/metabolismo , Ácidos Nucleicos Peptídicos/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Arginina/metabolismo , Cátions/química , Células/citologia , Endossomos/metabolismo , Ácidos Graxos/metabolismo , Produtos do Gene tat/metabolismo , Células HeLa , Humanos , Luciferases/metabolismo , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/toxicidade , Ácidos Nucleicos Peptídicos/química , Ácidos Nucleicos Peptídicos/toxicidade , Peptídeos/química , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA