Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 851
Filtrar
1.
Pharmacol Ther ; 254: 108594, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38290651

RESUMO

Cerebral dopamine neurotrophic factor (CDNF) is an endogenous protein in humans and other vertebrates, and it has been shown to have protective and restorative effects on cells in various disease models. Although it is named as a neurotrophic factor, its actions are drastically different from classical neurotrophic factors such as neurotrophins or the glial cell line-derived neurotrophic family of proteins. Like all secreted proteins, CDNF has a signal sequence at the N-terminus, but unlike common growth factors it has a KDEL-receptor retrieval sequence at the C-terminus. Thus, CDNF is mainly located in the ER. In response to adverse effects, such as ER stress, the expression of CDNF is upregulated and can alleviate ER stress. Also different from other neurotrophic factors, CDNF reduces protein aggregation and inflammation in disease models. Although it is an ER luminal protein, it can surprisingly directly interact with alpha-synuclein, a protein involved in the pathogenesis of synucleinopathies e.g., Parkinson's disease. Pleiotropic CDNF has therapeutic potential and has been tested as a recombinant human protein and gene therapy. The neuroprotective and neurorestorative effects have been described in a number of preclinical studies of Parkinson's disease, stroke and amyotrophic lateral sclerosis. Currently, it was successfully evaluated for safety in a phase 1/2 clinical trial for Parkinson's disease. Collectively, based on recent findings on the mode of action and therapeutic potential of CDNF, its use as a drug could be expanded to other ER stress-related diseases.


Assuntos
Doença de Parkinson , Animais , Humanos , Doença de Parkinson/tratamento farmacológico , Fatores de Crescimento Neural/uso terapêutico , Fatores de Crescimento Neural/metabolismo , Proteínas Recombinantes/uso terapêutico
2.
Acta Neuropathol Commun ; 12(1): 10, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38229173

RESUMO

Mesencephalic astrocyte-derived neurotrophic factor (MANF) has cytoprotective effects on various injuries, including cerebral ischemia, and it can promote recovery even when delivered intracranially several days after ischemic stroke. In the uninjured rodent brain, MANF protein is expressed almost exclusively in neurons, but post-ischemic MANF expression has not been characterized. We aimed to investigate how endogenous cerebral MANF protein expression evolves in infarcted human brains and rodent ischemic stroke models. During infarct progression, the cerebral MANF expression pattern both in human and rat brains shifted drastically from neurons to expression in inflammatory cells. Intense MANF immunoreactivity took place in phagocytic microglia/macrophages in the ischemic territory, peaking at two weeks post-stroke in human and one-week post-stroke in rat ischemic cortex. Using double immunofluorescence and mice lacking MANF gene and protein from neuronal stem cells, neurons, astrocytes, and oligodendrocytes, we verified that MANF expression was induced in microglia/macrophage cells in the ischemic hemisphere. Embarking on the drastic expression transition towards inflammatory cells and the impact of blood-borne inflammation in stroke, we hypothesized that exogenously delivered MANF protein can modulate tissue recovery processes. In an attempt to enhance recovery, we designed a set of proof-of-concept studies using systemic delivery of recombinant MANF in a rat model of cortical ischemic stroke. Intranasal recombinant MANF treatment decreased infarct volume and reduced the severity of neurological deficits. Intravenous recombinant MANF treatment decreased the levels of pro-inflammatory cytokines and increased the levels of anti-inflammatory cytokine IL-10 in the infarcted cortex one-day post-stroke. In conclusion, MANF protein expression is induced in activated microglia/macrophage cells in infarcted human and rodent brains, and this could implicate MANF's involvement in the regulation of post-stroke inflammation in patients and experimental animals. Moreover, systemic delivery of recombinant MANF shows promising immunomodulatory effects and therapeutic potential in experimental ischemic stroke.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , Ratos , Camundongos , Animais , AVC Isquêmico/metabolismo , Ratos Sprague-Dawley , Encéfalo/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Acidente Vascular Cerebral/metabolismo , Infarto Cerebral/metabolismo , Inflamação/metabolismo
3.
Int Rev Neurobiol ; 171: 125-162, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37783554

RESUMO

Sleep deprivation is quite frequent in military during combat, intelligence gathering or peacekeeping operations. Even one night of sleep deprivation leads to accumulation of amyloid beta peptide burden that would lead to precipitation of Alzheimer's disease over the years. Thus, efforts are needed to slow down or neutralize accumulation of amyloid beta peptide (AßP) and associated Alzheimer's disease brain pathology including phosphorylated tau (p-tau) within the brain fluid environment. Sleep deprivation also alters serotonin (5-hydroxytryptamine) metabolism in the brain microenvironment and impair upregulation of several neurotrophic factors. Thus, blockade or neutralization of AßP, p-tau and serotonin in sleep deprivation may attenuate brain pathology. In this investigation this hypothesis is examined using nanodelivery of cerebrolysin- a balanced composition of several neurotrophic factors and active peptide fragments together with monoclonal antibodies against AßP, p-tau and serotonin (5-hydroxytryptamine, 5-HT). Our observations suggest that sleep deprivation induced pathophysiology is significantly reduced following nanodelivery of cerebrolysin together with monoclonal antibodies to AßP, p-tau and 5-HT, not reported earlier.


Assuntos
Doença de Alzheimer , Fármacos Neuroprotetores , Humanos , Peptídeos beta-Amiloides , Doença de Alzheimer/metabolismo , Serotonina/metabolismo , Privação do Sono/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Encéfalo/metabolismo , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico
4.
J Virol ; 97(10): e0069623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796129

RESUMO

IMPORTANCE: Human cytomegalovirus (HCMV) infection is the leading cause of non-heritable birth defects worldwide. HCMV readily infects the early progenitor cell population of the developing brain, and we have found that infection leads to significantly downregulated expression of key neurodevelopmental transcripts. Currently, there are no approved therapies to prevent or mitigate the effects of congenital HCMV infection. Therefore, we used human-induced pluripotent stem cell-derived organoids and neural progenitor cells to elucidate the glycoproteins and receptors used in the viral entry process and whether antibody neutralization was sufficient to block viral entry and prevent disruption of neurodevelopmental gene expression. We found that blocking viral entry alone was insufficient to maintain the expression of key neurodevelopmental genes, but neutralization combined with neurotrophic factor treatment provided robust protection. Together, these studies offer novel insight into mechanisms of HCMV infection in neural tissues, which may aid future therapeutic development.


Assuntos
Anticorpos Neutralizantes , Infecções por Citomegalovirus , Citomegalovirus , Expressão Gênica , Fatores de Crescimento Neural , Humanos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Organoides/citologia , Organoides/metabolismo , Organoides/virologia , Receptores Virais/antagonistas & inibidores , Receptores Virais/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos
5.
Int Rev Neurobiol ; 172: 103-143, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37833010

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder characterised by the loss of dopaminergic neurons in the substantia nigra pars compacta and the subsequent motor disability. The most frequently used treatments in clinics, such as L-DOPA, restore dopaminergic neurotransmission in the brain. However, these treatments are only symptomatic, have temporary efficacy, and produce side effects. Part of the side effects are related to the route of administration as the consumption of oral tablets leads to unspecific pulsatile activation of dopaminergic receptors. For this reason, it is necessary to not only find alternative treatments, but also to develop new administration systems with better security profiles. Nanoparticle delivery systems are new administration forms designed to reach the pharmacological target in a highly specific way, leading to better drug bioavailability, efficacy and safety. Some of these delivery systems have shown promising results in animal models of PD not only when dopaminergic drugs are administered, but even more when neurotrophic factors are released. These latter compounds promote maturation and survival of dopaminergic neurons and can be exogenously administered in the form of pharmacological therapy or endogenously generated by non-pharmacological methods. In this sense, experimental exposure to enriched environments, a non-invasive strategy based on the combination of social and inanimate stimuli, enhances the production of neurotrophic factors and produces a neuroprotective effect in parkinsonian animals. In this review, we will discuss new nanodelivery systems in PD with a special focus on therapies that increase the release of neurotrophic factors.


Assuntos
Pessoas com Deficiência , Transtornos Motores , Doença de Parkinson , Animais , Humanos , Doença de Parkinson/tratamento farmacológico , Levodopa/uso terapêutico , Fatores de Crescimento Neural/uso terapêutico
6.
Mov Disord ; 38(7): 1209-1222, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37212361

RESUMO

BACKGROUND: Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotrophic factor that protects dopamine neurons and improves motor function in animal models of Parkinson's disease (PD). OBJECTIVE: The primary objectives of this study were to assess the safety and tolerability of both CDNF and the drug delivery system (DDS) in patients with PD of moderate severity. METHODS: We assessed the safety and tolerability of monthly intraputamenal CDNF infusions in patients with PD using an investigational DDS, a bone-anchored transcutaneous port connected to four catheters. This phase 1 trial was divided into a placebo-controlled, double-blind, 6-month main study followed by an active-treatment 6-month extension. Eligible patients, aged 35 to 75 years, had moderate idiopathic PD for 5 to 15 years and Hoehn and Yahr score ≤ 3 (off state). Seventeen patients were randomized to placebo (n = 6), 0.4 mg CDNF (n = 6), or 1.2 mg CDNF (n = 5). The primary endpoints were safety and tolerability of CDNF and DDS and catheter implantation accuracy. Secondary endpoints were measures of PD symptoms, including Unified Parkinson's Disease Rating Scale, and DDS patency and port stability. Exploratory endpoints included motor symptom assessment (PKG, Global Kinetics Pty Ltd, Melbourne, Australia) and positron emission tomography using dopamine transporter radioligand [18 F]FE-PE2I. RESULTS: Drug-related adverse events were mild to moderate with no difference between placebo and treatment groups. No severe adverse events were associated with the drug, and device delivery accuracy met specification. The severe adverse events recorded were associated with the infusion procedure and did not reoccur after procedural modification. There were no significant changes between placebo and CDNF treatment groups in secondary endpoints between baseline and the end of the main and extension studies. CONCLUSIONS: Intraputamenally administered CDNF was safe and well tolerated, and possible signs of biological response to the drug were observed in individual patients. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Doença de Parkinson , Animais , Doença de Parkinson/tratamento farmacológico , Dopamina , Fatores de Crescimento Neural/fisiologia , Fatores de Crescimento Neural/uso terapêutico , Neurônios Dopaminérgicos , Sistemas de Liberação de Medicamentos , Método Duplo-Cego
7.
Curr Neurol Neurosci Rep ; 23(5): 201-212, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37014546

RESUMO

PURPOSE OF REVIEW: Recognition of node of Ranvier as the site of injury in inflammatory neuropathies contributed to discovery of antibodies against the nodal/paranodal structures. These antibodies mediate a unique type of inflammatory neuropathies that are different from typical chronic inflammatory demyelinating polyneuropathy. This review discusses the advancements made in the field of autoimmune neuropathies secondary to antibodies to nodal and paranodal proteins. RECENT FINDINGS: Neuropathies caused by antibodies to nodal-paranodal antigens including neurofascin 186, neurofascin 155, contactin1, and contactin-associated protein1 were termed as autoimmune nodopathies (AN) in 2021. Since the initial description almost a decade ago, newer cohorts have expanded the clinical spectrum of AN. In addition to IgG4, other subclasses of IgG such as IgG1/IgG3 have been identified, particularly in relation to acute presentations and anti-pan neurofascin antibody disease. In vitro and in vivo studies have also supported antibody-mediated pathogenicity of many of these biomarkers. Antibodies to nodal-paranodal antigens have emerged as a biomarker for a novel type of immune-mediated neuropathies. These antibodies have distinct pathogenic mechanisms and produce a unique set of clinicopathologic features. Their clinical profile and treatment may also vary depending on the antibody isotype. B cell depleting therapies are effective in managing some of these patients.


Assuntos
Polirradiculoneuropatia Desmielinizante Inflamatória Crônica , Nós Neurofibrosos , Humanos , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/patologia , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Moléculas de Adesão Celular/metabolismo , Moléculas de Adesão Celular/uso terapêutico , Autoanticorpos , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/tratamento farmacológico , Imunoglobulina G
8.
Cell Death Dis ; 14(2): 128, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36792604

RESUMO

During intracerebral hemorrhage (ICH), hematoma formation at the site of blood vessel damage results in local mechanical injury. Subsequently, erythrocytes lyse to release hemoglobin and heme, which act as neurotoxins and induce inflammation and secondary brain injury, resulting in severe neurological deficits. Accelerating hematoma resorption and mitigating hematoma-induced brain edema by modulating immune cells has potential as a novel therapeutic strategy for functional recovery after ICH. Here, we show that intracerebroventricular administration of recombinant human cerebral dopamine neurotrophic factor (rhCDNF) accelerates hemorrhagic lesion resolution, reduces peri-focal edema, and improves neurological outcomes in an animal model of collagenase-induced ICH. We demonstrate that CDNF acts on microglia/macrophages in the hemorrhagic striatum by promoting scavenger receptor expression, enhancing erythrophagocytosis and increasing anti-inflammatory mediators while suppressing the production of pro-inflammatory cytokines. Administration of rhCDNF results in upregulation of the Nrf2-HO-1 pathway, but alleviation of oxidative stress and unfolded protein responses in the perihematomal area. Finally, we demonstrate that intravenous delivery of rhCDNF has beneficial effects in an animal model of ICH and that systemic application promotes scavenging by the brain's myeloid cells for the treatment of ICH.


Assuntos
Edema Encefálico , Lesões Encefálicas , Animais , Humanos , Hemorragia Cerebral/complicações , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Inflamação/complicações , Hematoma/tratamento farmacológico , Hematoma/complicações , Hematoma/metabolismo , Imunidade Inata , Modelos Animais de Doenças , Edema Encefálico/complicações , Fatores de Crescimento Neural/uso terapêutico
9.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-36499693

RESUMO

Merkel cell carcinoma (MCC) is a rare and aggressive cutaneous malignant tumor with neuroendocrine differentiation, with a rapidly growing incidence rate, high risk of recurrence, and aggressive behavior. The available therapeutic options for advanced disease are limited and there is a pressing need for new treatments. Tumors harboring fusions involving one of the neurotrophin receptor tyrosine kinase (NTRK) genes are now actionable with targeted inhibitors. NTRK-fused genes have been identified in neuroendocrine tumors of other sites; thus, a series of 76 MCCs were firstly analyzed with pan-TRK immunohistochemistry and the positive ones with real-time RT-PCR, RNA-based NGS, and FISH to detect the eventual underlying gene fusion. Despite 34 MCCs showing pan-TRK expression, NTRK fusions were not found in any cases. As in other tumors with neural differentiation, TRK expression seems to be physiological and not caused by gene fusions.


Assuntos
Carcinoma de Célula de Merkel , Neoplasias , Neoplasias Cutâneas , Humanos , Receptor trkA/genética , Carcinoma de Célula de Merkel/genética , Fatores de Crescimento Neural/uso terapêutico , Receptor trkC/genética , Neoplasias/patologia , Neoplasias Cutâneas/genética , Proteínas de Fusão Oncogênica/genética , Biomarcadores Tumorais/genética
10.
J Agric Food Chem ; 70(42): 13602-13614, 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36239029

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease, characterized by memory loss and cognitive deficits accompanied by neuronal damage and cholinergic disorders. Sesamol, a lignan component in sesame oil, has been proven to have neuroprotective effects. This research aimed to investigate the preventive effects of sesamol on scopolamine (SCOP)-induced cholinergic disorders in C57BL/6 mice. The mice were pretreated with sesamol (100 mg/kg/d, p.o.) for 30 days. Behavioral tests indicated that sesamol supplement prevented SCOP-induced cognitive deficits. Sesamol enhanced the expression of neurotrophic factors and postsynaptic density (PSD) in SCOP-treated mice, reversing neuronal damage and synaptic dysfunction. Importantly, sesamol could balance the cholinergic system by suppressing the AChE activity and increasing the ChAT activity and M1 mAChR expression. Sesamol treatment also inhibited the expression of inflammatory factors and overactivation of microglia in SCOP-treated mice. Meanwhile, sesamol improved the antioxidant enzyme activity and suppressed oxidative stress in SCOP-treated mice and ameliorated the oxidized cellular status and mitochondrial dysfunction in SCOP-treated SH-SY5Y cells. In conclusion, these results indicated that sesamol attenuated SCOP-induced cognitive dysfunction via balancing the cholinergic system and reducing neuroinflammation and oxidative stress.


Assuntos
Disfunção Cognitiva , Lignanas , Neuroblastoma , Doenças Neurodegenerativas , Fármacos Neuroprotetores , Animais , Humanos , Camundongos , Antioxidantes/metabolismo , Colinérgicos , Cognição , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/genética , Lignanas/farmacologia , Aprendizagem em Labirinto , Transtornos da Memória/induzido quimicamente , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico , Doenças Neuroinflamatórias , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , Escopolamina , Óleo de Gergelim
11.
Psychiatr Danub ; 34(3): 390-397, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36256972

RESUMO

Major Depressive Disorder (MDD) is one of the leading causes of disability worldwide. The current pharmacological treatment options for MDD, which rely on the mono-amine hypothesis, has their limitations with respect to treatment non-response, partial response etc. This propels for a search for a novel neurobiological understanding of MDD that can lead to novel treatment options. A literature search strategy was thus employed using relevant keywords pertaining to the topic in PubMed, Embase and Google Scholar. Systematic reviews and meta-analyses, narrative reviews and clinical trials were reviewed to incorporate the most robust evidence-based literature available. A total of 37 publications were narrowed down based upon the topic. Alterations in brain neuroplasticity, as evidenced by changes in neurotrophic factors and from neuroimaging, has been found to be a strong patho-mechanism for MDD. This link has been exploited to stimulate psychopharmacological research to treat MDD.


Assuntos
Transtorno Depressivo Maior , Humanos , Transtorno Depressivo Maior/tratamento farmacológico , Depressão , Plasticidade Neuronal , Encéfalo/diagnóstico por imagem , Fatores de Crescimento Neural/uso terapêutico
12.
Ageing Res Rev ; 82: 101763, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36272696

RESUMO

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a novel evolutionarily conserved protein present in both vertebrate and invertebrate species. MANF shows distinct structural and functional properties than the traditional neurotrophic factors (NTF). MANF is composed of an N-terminal saposin-like lipid-binding domain and a C-terminal SAF-A/B, Acinus and PIAS (SAP) domain connected by a short linker. The two well-described activities of MANF include (1) role as a neurotrophic factor that plays direct neuroprotective effects in the nervous system and (2) cell protective effects in the animal models of non-neuronal diseases, including retinal damage, diabetes mellitus, liver injury, myocardial infarction, nephrotic syndrome, etc. The main objective of the current review is to provide up-to-date insights regarding the structure of MANF, mechanisms regulating its expression and secretion, physiological functions in various tissues and organs, protective effects during aging, and potential clinical applications. Together, this review highlights the importance of MANF in reversing age-related dysfunction and geroprotection.


Assuntos
Astrócitos , Fármacos Neuroprotetores , Animais , Humanos , Astrócitos/metabolismo , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Estresse do Retículo Endoplasmático
13.
Clin Transl Med ; 12(9): e1060, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36149789

RESUMO

BACKGROUND: Small-cell lung cancer (SCLC) molecular subtypes have been primarily characterized based on the expression pattern of the following key transcription regulators: ASCL1 (SCLC-A), NEUROD1 (SCLC-N), POU2F3 (SCLC-P) and YAP1 (SCLC-Y). Here, we investigated the proteomic landscape of these molecular subsets with the aim to identify novel subtype-specific proteins of diagnostic and therapeutic relevance. METHODS: Pellets and cell media of 26 human SCLC cell lines were subjected to label-free shotgun proteomics for large-scale protein identification and quantitation, followed by in-depth bioinformatic analyses. Proteomic data were correlated with the cell lines' phenotypic characteristics and with public transcriptomic data of SCLC cell lines and tissues. RESULTS: Our quantitative proteomic data highlighted that four molecular subtypes are clearly distinguishable at the protein level. The cell lines exhibited diverse neuroendocrine and epithelial-mesenchymal characteristics that varied by subtype. A total of 367 proteins were identified in the cell pellet and 34 in the culture media that showed significant up- or downregulation in one subtype, including known druggable proteins and potential blood-based markers. Pathway enrichment analysis and parallel investigation of transcriptomics from SCLC cell lines outlined unique signatures for each subtype, such as upregulated oxidative phosphorylation in SCLC-A, DNA replication in SCLC-N, neurotrophin signalling in SCLC-P and epithelial-mesenchymal transition in SCLC-Y. Importantly, we identified the YAP1-driven subtype as the most distinct SCLC subgroup. Using sparse partial least squares discriminant analysis, we identified proteins that clearly distinguish four SCLC subtypes based on their expression pattern, including potential diagnostic markers for SCLC-Y (e.g. GPX8, PKD2 and UFO). CONCLUSIONS: We report for the first time, the protein expression differences among SCLC subtypes. By shedding light on potential subtype-specific therapeutic vulnerabilities and diagnostic biomarkers, our results may contribute to a better understanding of SCLC biology and the development of novel therapies.


Assuntos
Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Biomarcadores , Linhagem Celular Tumoral , Meios de Cultura , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Peroxidases/genética , Peroxidases/metabolismo , Peroxidases/uso terapêutico , Proteômica , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo
14.
Biomed Pharmacother ; 155: 113664, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36095961

RESUMO

Mitochondrial abnormalities and a defective expression of neurotrophic factors contribute to neuronal damage in Huntington's disease (HD). HD patients showed a reduction in transforming growth factor-ß1 (TGF-ß1) levels in the peripheral blood and in cortical neurons. 3-n-butylphthalide (NBP) is first isolated from the seeds of celery, treats ischemic stroke in China. NBP could attenuate cognitive and motor impairments in the experimental models of Parkinson's disease and Alzheimer's disease, reduce mitochondrial oxidative stress and increase the expression of TGF-ß1 in rats with focal cerebral ischemia. To our knowledge, the effect of NBP on Huntington's disease has not been reported. We proposed the hypothesis that whether NBP could protect mitochondria and regulate TGF-ß1 and its downstream signaling in a HD animal model, further prevents motor dysfunction. Malonic acid is a reversible inhibitor of mitochondrial enzyme complex-II, induces energy crisis and free radical generation. In this study, we used intrastriatal injections of malonic acid in rats to mimic mitochondrial abnormalities and the other HD like symptoms. We found that treatment with NBP significantly attenuated malonic acid-induced motor and cognitive dysfunction in locomotor behaviour test, rotarod test, novel object recognition test and morris water maze test, prevented neurotoxicity and mitochondrial damage, activated TGF-ß1/Akt/Wnt/ß-Catenin pathway in striatum, but didn't regulate mitochondrial fusion and fission. The above effect was partly reversed by a PI3K/Akt inhibitor. Our data support NBP as a potential candidate for the treatment of HD.


Assuntos
Doença de Huntington , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Animais , Ratos , Fator de Crescimento Transformador beta1 , Doença de Huntington/induzido quimicamente , beta Catenina , Fármacos Neuroprotetores/uso terapêutico , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Síndromes Neurotóxicas/tratamento farmacológico , Fatores de Crescimento Neural/uso terapêutico
15.
Int J Mol Sci ; 23(16)2022 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-36012320

RESUMO

Prenatal hypoxic−ischemic (HI) injury inflicts severe damage on the developing brain provoked by a pathophysiological response that leads to neural structural lesions, synaptic loss, and neuronal death, which may result in a high risk of permanent neurological deficits or even newborn decease. It is known that growth hormone (GH) can act as a neurotrophic factor inducing neuroprotection, neurite growth, and synaptogenesis after HI injury. In this study we used the chicken embryo to develop both in vitro and in vivo models of prenatal HI injury in the cerebral pallium, which is the equivalent of brain cortex in mammals, to examine whether GH exerts neuroprotective and regenerative effects in this tissue and the putative mechanisms involved in these actions. For the in vitro experiments, pallial cell cultures obtained from chick embryos were incubated under HI conditions (<5% O2, 1 g/L glucose) for 24 h and treated with 10 nM GH, and then collected for analysis. For the in vivo experiments, chicken embryos (ED14) were injected in ovo with GH (2.25 µg), exposed to hypoxia (12% O2) for 6 h, and later the pallial tissue was obtained to perform the studies. Results show that GH exerted a clear anti-apoptotic effect and promoted cell survival and proliferation in HI-injured pallial neurons, in both in vitro and in vivo models. Neuroprotective actions of GH were associated with the activation of ERK1/2 and Bcl-2 signaling pathways. Remarkably, GH protected mature neurons that were particularly harmed by HI injury, but was also capable of stimulating neural precursors. In addition, GH stimulated restorative processes such as the number and length of neurite outgrowth and branching in HI-injured pallial neurons, and these effects were blocked by a specific GH antibody, thus indicating a direct action of GH. Furthermore, it was found that the local expression of several synaptogenic markers (NRXN1, NRXN3, GAP-43, and NLG1) and neurotrophic factors (GH, BDNF, NT-3, IGF-1, and BMP4) were increased after GH treatment during HI damage. Together, these results provide novel evidence supporting that GH exerts protective and restorative effects in brain pallium during prenatal HI injury, and these actions could be the result of a joint effect between GH and endogenous neurotrophic factors. Also, they encourage further research on the potential role of GH as a therapeutic complement in HI encephalopathy treatments.


Assuntos
Hormônio do Crescimento Humano , Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Animais , Animais Recém-Nascidos , Embrião de Galinha , Galinhas/metabolismo , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/uso terapêutico , Hipóxia/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Isquemia/tratamento farmacológico , Mamíferos/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
16.
Yakugaku Zasshi ; 142(3): 241-277, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35228379

RESUMO

Neurotrophic factors have been shown to potentially be beneficial for the treatment of neurodegenerative diseases such as Alzheimer's disease, because endogenous neurotrophic factors (NGF, BDNF) have been recognized to play critical roles in the promotion of neurogenesis, differentiation, and neuroprotection throughout the development of the central nervous system. However, high-molecular-weight proteins are unable to cross the blood-brain barrier and are easily decomposed under physiological conditions. Thus, small molecules that can mimic the functions of neurotrophic factors are promising alternatives for the treatment of neurodegenerative disease. Since 1990, the author has been involved in searching for natural products with typical neurotrophic properties that can cause neurogenesis, enhance neurite outgrowth, and protect against neuronal death by using three cellular systems (PC12, rat cortical neurons, and MEB5 cells). Through these research activities on neurotrophic natural products, the author has tried to induce a paradigm shift from the discipline of natural products chemistry to science disciplines. This review focuses on our independent synthetic studies of the neurotrophic natural products discovered in the plants. The following synthetic elaborations are described: syntheses of dimeric isocuparane-type sesquiterpenes mastigophorenes A and B, macrocyclic bis-bibenzyls plagiochins A-D and cavicularin through a Pd-catalyzed Stille-Kelly reaction; the formal synthesis of merrilactone A and jiadifenin, which are seco-prezizaane-type sesquiterpenes, through intramolecular Pd-catalyzed Mizoroki-Heck and Tsuji-Trost reactions; and finally the first enantioselective synthesis of neovibsanin B, a vibsane-type diterpene, through a Pd-catalyzed cyclic carbopalladation-carbonyl tandem reaction.


Assuntos
Produtos Biológicos/síntese química , Fatores de Crescimento Neural/síntese química , Animais , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Barreira Hematoencefálica/metabolismo , Hidrocarbonetos Aromáticos com Pontes/síntese química , Ciclopentanos/síntese química , Diterpenos/síntese química , Lactonas/síntese química , Camundongos , Peso Molecular , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Fenômenos de Química Orgânica , Ratos , Sesquiterpenos/síntese química , Estereoisomerismo
17.
Trends Endocrinol Metab ; 33(4): 236-246, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35135706

RESUMO

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum-resident protein and a secretory factor and has beneficial effects in multiple diseases. Recent evidence shows that its circulating levels in humans are dynamically regulated under various metabolic diseases, including diabetes, obesity, fatty liver, and cardiovascular diseases, suggesting that MANF may play a role in these pathological states. Also, its downregulation in mice impairs glucose homeostasis, promotes lipid accumulation in the liver, reduces energy expenditure, and induces inflammation. Conversely, MANF overexpression prevents or mitigates some of these metabolic disturbances. In particular, systemic MANF administration alleviates dietary obesity and related metabolic disorders in obese mice. We therefore propose that MANF might be a promising target for treating chronic metabolic diseases.


Assuntos
Doenças Metabólicas , Fatores de Crescimento Neural , Animais , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Humanos , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Camundongos , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Obesidade/metabolismo
18.
Cell Death Dis ; 13(2): 100, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35110525

RESUMO

Acetaminophen (APAP)-induced liver injury (AILI) is the most frequent cause of acute liver failure; but the underlying mechanisms still remain obscure. Macrophages and endoplasmic reticulum (ER) stress play an important role in the pathogenesis of AILI. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a newly identified 18-kDa soluble protein, whose expression and secretion are stimulated by ER stress. To investigate the role of myeloid cell MANF in the pathogenesis of AILI, we assayed serum and liver samples from AILI model mice and patients with drug-induced liver injury (DILI). We demonstrated that the levels of MANF were elevated in patients with DILI and in mice with AILI. Moreover, myeloid-specific MANF knockout mice were generated and used. It was observed that a delayed liver recovery from myeloid-specific MANF gene knockout mice following APAP overdose compared to that from wild-type mice. MANF deficiency in myeloid cells resulted in increased infiltrating monocyte-derived macrophages (MoMFs) but reduced restorative Ly6Clow macrophages after APAP treatment. MANF supplementation increased restorative Ly6Clow macrophages and subsequently alleviated liver injury. Moreover, MANF could enhance IL-10 expression and phagocytosis in macrophages via p38 MAPK pathway. Altogether, MANF seems to be a critical immune modulator in promoting liver repair via reducing and reprogramming MoMFs. MANF perhaps promoted the phenotype conversion of pro-inflammatory MoMFs to pro-restorative Ly6Clow MoMFs via p38 MAPK pathway, particularly through enhancing IL-10 and phagocytosis.


Assuntos
Acetaminofen/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Macrófagos/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Interleucina-10/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Células Mieloides/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Fagocitose , Transdução de Sinais
19.
Drug Chem Toxicol ; 45(6): 2769-2779, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34674598

RESUMO

Chemotherapy-induced cognitive impairment (CICI) comprises different neurological problems, including difficulty in learning new things, concentrating and making decisions that affect daily life activities. Clinical reports indicate that around 70% of cancer patients receiving chemotherapy suffer from cognitive impairment. The purpose of the present study is to examine the effects of widely used anticancer medication (Carmustine) on cognitive function using mice model and investigation of the neuroprotective effects of Cerebrolysin (CBN). Cerebrolysin (CBN) is a mixture of several neurotrophic factors and active peptides with anti-inflammatory, antioxidant, and neuroprotective actions. Our study aimed to establish a mice model of Carmustine (BCNU)-induced cognitive deficits and determine the protective effects of CBN. BCNU (10 mg/kg, i.v.) was administered to mice for 28 days, and behavioral parameters were measured on a weekly basis. CBN (44 and 88 mg/kg, i.p.) was administered daily from day 1 to 28 to BCNU treatment mice. All animals were sacrificed on day 29 and brain hippocampus tissues were used for biochemical, neuroinflammatory, neurotransmitters analysis. BCNU administration animals showed impaired cognition and memory, confirmed from behavioral analysis. Further, BCNU increased oxidative stress, inflammatory cytokines release and altered neurotransmitters concentration as compared to the control group (p < 0.01). However, mice treated with CBN (44 and 88 mg/kg, i.p.) significantly and dose-dependently improved cognitive functions, reduced oxidative stress markers, inflammatory cytokines and restored neurotransmitters concentration as compared to BCNU administered mice (p < 0.05). The finding of current study suggested that CBN could be the promising compound to reverse cognitive impairment associated with use of chemotherapy.


Assuntos
Disfunção Cognitiva , Fármacos Neuroprotetores , Animais , Camundongos , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Carmustina/efeitos adversos , Carmustina/uso terapêutico , Carmustina/toxicidade , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Citocinas , Modelos Animais de Doenças , Fatores de Crescimento Neural/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Neurotransmissores
20.
Mol Biol Rep ; 49(3): 2345-2357, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34826049

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia among the elderly population. AD is accompanied with the dysregulation of specific neurotrophic factors (NTFs) and their receptors, which plays a critical role in neuronal degeneration. NTFs are small proteins with therapeutic potential for human neurodegenerative diseases. These growth factors are categorized into four families: neurotrophins, neurokines, the glial cell line-derived NTF family of ligands, and the newly discovered cerebral dopamine NTF/mesencephalic astrocyte-derived NTF family. NTFs are capable of preventing cell death in degenerative conditions and can increase the neuronal growth and function in these disorders. Nevertheless, the adverse side effects of NTFs delivery and poor diffusion of these factors in the brain restrict the efficacy of NTFs therapy in clinical situations. MATERIALS AND METHODS: In this review, we focus on the current advances in the use of NTFs to treat AD and summarize previous experimental and clinical studies for supporting the protective and therapeutic effects of these factors. CONCLUSION: Based on reports, NTFs are considered as new and promising candidates for treating AD and AD-associated cognitive impairment.


Assuntos
Doença de Alzheimer , Fatores de Crescimento Neural , Idoso , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Humanos , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...