Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
2.
Signal Transduct Target Ther ; 8(1): 450, 2023 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-38086815

RESUMO

With the development and regulatory approval of immune checkpoint inhibitors and adoptive cell therapies, cancer immunotherapy has undergone a profound transformation over the past decades. Recently, therapeutic cancer vaccines have shown promise by eliciting de novo T cell responses targeting tumor antigens, including tumor-associated antigens and tumor-specific antigens. The objective was to amplify and diversify the intrinsic repertoire of tumor-specific T cells. However, the complete realization of these capabilities remains an ongoing pursuit. Therefore, we provide an overview of the current landscape of cancer vaccines in this review. The range of antigen selection, antigen delivery systems development the strategic nuances underlying effective antigen presentation have pioneered cancer vaccine design. Furthermore, this review addresses the current status of clinical trials and discusses their strategies, focusing on tumor-specific immunogenicity and anti-tumor efficacy assessment. However, current clinical attempts toward developing cancer vaccines have not yielded breakthrough clinical outcomes due to significant challenges, including tumor immune microenvironment suppression, optimal candidate identification, immune response evaluation, and vaccine manufacturing acceleration. Therefore, the field is poised to overcome hurdles and improve patient outcomes in the future by acknowledging these clinical complexities and persistently striving to surmount inherent constraints.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Antígenos de Neoplasias , Imunoterapia , Imunidade , Microambiente Tumoral
3.
Adv Healthc Mater ; 12(28): e2301328, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37392128

RESUMO

To strengthen the antitumor efficacy and avoid toxicity to normal cells of cisplatin and triptolide, herein, an acid and glutathione (GSH) dual-controlled nanoplatform for enhanced cancer treatment through the synergy of both "1+1" apoptosis and "1+1" ferroptosis is designed. Remarkably, ZIF8 in response to tumor microenvironment enhances drug targeting and protects drugs from premature degradation. Meanwhile, the PtIV  center can be easily reduced to cisplatin because of the large amount of GSH, thus liberating the triptolide as the coordinated ligand. The released cisplatin and hemin in turn boost the tumor cell "1+1" apoptosis through chemotherapy and photodynamic therapy, respectively. Furthermore, GSH reduction through PtIV  weakens the activation of glutathione peroxidase 4 (GPX4) effectively. The released triptolide can inhibit the expressions of GSH by regulating nuclear factor E2 related factor 2 (Nrf2), further promoting membrane lipid peroxidation, thus "1+1" ferroptosis can be achieved. Both in vitro and in vivo results demonstrate that the nanosystem can not only perform superior specificity and therapeutic outcomes but also reduce the toxicity to normal cells/tissues of cisplatin and triptolide effectively. Overall, the prodrug-based smart system provides an efficient therapeutic strategy for cancer treatment by virtue of the effect of enhanced "1+1" apoptosis and "1+1" ferroptosis therapies.


Assuntos
Neoplasias da Mama , Diterpenos , Pró-Fármacos , Humanos , Feminino , Cisplatino/farmacologia , Neoplasias da Mama/tratamento farmacológico , Pró-Fármacos/farmacologia , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Mater Today Bio ; 16: 100375, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35983175

RESUMO

Severely hypoxic condition of tumour represents a notable obstacle against the efficiency of photodynamic therapy (PDT). While mitochondria targeted therapy by metformin has been considered as a promising strategy for reducing oxygen consumption in tumours, its low treatment sensitivity, short half-life and narrow absorption window in vivo remain the intractable challenges. In this report, 5'-guanosine monophosphate (5'GMP), indocyanine green (ICG), hemin and metformin, were combined to construct a smart G-quadruplex (G4) hydrogel named HMI@GEL for breast cancer (BC) treatment. Benefiting from the photothermal (PTT) effect of ICG, HMI@GEL exhibited excellent characteristics of NIR-light-triggered and persistent drug delivery to maintain high intratumoral concentration of metformin. Furthermore, drug loading concentration of metformin reached an amazing 300 â€‹mg â€‹mL-1 in HMI@GEL. To our knowledge, it might be the highest loading efficiency in the reported literatures. With the combination of catalase-mimicking Hemin@mil88, metformin could inhibit tumour mitochondrial respiratory significantly, which sequentially permitted in situ efficient oxygen generation. Remarkable apoptosis and necrosis were achieved by the combination of PTT and synergistically enhanced PDT as well as the activated tumour immunotherapy. Collectively, the HMI@GEL in situ injectable platform showed a promising strategy for enhanced PDT by metformin, and opened new perspectives for treating BC versatilely.

5.
Chin J Nat Med ; 20(7): 494-505, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35907648

RESUMO

Impaired immunomodulatory capacity and oxidative stress are the key factors limiting the effectiveness of mesenchymal stem cell transplantation therapy. The present study was aimed to investigate the effects of jujuboside A (JuA) on the protective effect and immunomodulatory capacity of human umbilical cord mesenchymal stem cells (hUC-MSCs). Hydrogen peroxide was used to establish an oxidative damage model of hUC-MSCs, while PBMCs isolated from rats were used to evaluate the effect of JuA pre-treatment on the immunomodulatory capacity of hUC-MSCs. Furthermore, Hoechst 33258 staining, lactate dehydrogenase test, measurement of malondialdehyde, Western blot, high-performance liquid chromatography; and flow cytometry were performed. Our results indicated that JuA (25 µmol·L-1) promoted the proliferation of hUC-MSCs, but did not affect the differentiating capability of these cells. JuA pre-treatment inhibited apoptosis, prevented oxidative damage, and up-regulated the protein expression of nuclear factor-erythroid factor 2-related factor 2 and heme oxygenase 1 in hUC-MSCs in which oxidative stress was induced with H2O2. In addition, JuA pre-treatment enhanced the inhibitory effect of hUC-MSCs against abnormally activated PBMCs, which was related to stimulation of the expression and activity of indoleamine 2,3-dioxygenase. In conclusion, our results demonstrate that JuA pre-treatment can enhance the survival and immunomodulatory ability through pathways related to oxidative stress, providing a new option for the improvement of hUC-MSCs in the clinical setting.


Assuntos
Células-Tronco Mesenquimais , Cordão Umbilical , Animais , Diferenciação Celular , Humanos , Peróxido de Hidrogênio/metabolismo , Estresse Oxidativo , Ratos , Saponinas , Cordão Umbilical/metabolismo
6.
Neurosci Lett ; 772: 136473, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35077846

RESUMO

Mobilization of hippocampal neurogenesis has been considered as a potential strategy for the treatment of neurodegenerative diseases, including Alzheimer's disease (AD). In present study, we evaluated both the neuroprotective effects and the effects on the proliferation and differentiation of APP-overexpressing neural stem cells (APP-NSCs) by Jujuboside A (JuA) in vitro. Our results demonstrated that JuA (50 µM) decreased apoptosis and suppressed oxidative stress damage of APP-NSCs. JuA (50 µM) upregulated the secretion of brain-derived neurotrophic factor and promoted the proliferation and neuronal differentiation of APP-NSCs. Moreover, JuA (50 µM) upregulated Wnt-3a and ß-catenin protein expression, and enhanced the expression of downstream genes Ccnd1, Neurod1 and Prox1. However, XAV-939, an inhibitor of the Wnt/ß-catenin signaling pathway, inhibited these positive effects of JuA. Taken together, these findings suggest that JuA promote proliferation and neuronal differentiation of APP-NSCs partly by activating the Wnt/ß-catenin signaling pathway. We hope that this study will provide a viable strategy for the treatment of AD.


Assuntos
Proliferação de Células , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese , Saponinas/farmacologia , Via de Sinalização Wnt , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Compostos Heterocíclicos com 3 Anéis/farmacologia , Hipocampo/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , beta Catenina/metabolismo
7.
Acta Biomater ; 138: 463-477, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34718179

RESUMO

Photodynamic therapy (PDT) is an effective noninvasive therapeutic strategy that can convert oxygen to highly cytotoxic singlet oxygen (1O2) through the co-localization of excitation light and photosensitizers. However, compromised by the hypoxic tumor microenvironment, the therapeutic efficacy of PDT is reduced seriously. Herein, to overcome tumor-associated hypoxia, and further achieve tumor-targeted synergistic chemotherapy/PDT/photothermal therapy (PTT), we have constructed a biodegradable oxygen-producing nanoplatform (named Ini@PM-HP), which was composed of the porous metal-organic framework (PCN-224(Mn)), the poly (ADP-ribose) polymerase (PARP) inhibitor (Iniparib), and the polydopamine-modified hyaluronic acid (HA-PDA). Since HA can specifically bind to the overexpressed HA receptors (cluster determinant 44, CD44) on tumor cell, Ini@PM-HP prefers to accumulate at the tumor site once injected intravenously. Then iniparib can be released in tumor environment (TME), thereby dysfunctioning DNA damage repair and promoting cell apoptosis. At the same time, the chelating of Mn and tetrakis(4-carboxyphenyl) porphyrin (Mn-TCPP) can generate O2 in situ by reacting with endogenous H2O2, relieving the hypoxic TME and achieving enhanced PDT. Moreover, owing to the high photothermal conversion efficiency of PDA, PTT can be driven by the 808 nm laser irradiation. As systematically demonstrated in vitro and in vivo, this nanotherapeutic approach enables the combined therapy with great inhibition on tumor. Overall, the as-prepared nanoplatform provide a promising strategy to overcome tumor-associated hypoxia, and shows great potential for combination tumor therapy. STATEMENT OF SIGNIFICANCE: A delicately designed biodegradable oxygen-producing nanoplatform Ini@PM-HP is constructed to achieve combination therapy of solid tumors. Taking advantage of the active-targeting, PTT, enhanced PDT and PARPi, this nanotherapeutic approach successfully enables the combined chemo/photothermal/photodynamic therapy with great inhibition of solid tumors.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Fotoquimioterapia , Linhagem Celular Tumoral , Peróxido de Hidrogênio , Manganês , Estruturas Metalorgânicas/farmacologia , Oxigênio , Fármacos Fotossensibilizantes/farmacologia , Terapia Fototérmica
8.
ACS Appl Mater Interfaces ; 14(1): 57-68, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34935343

RESUMO

Integrating chemodynamic therapy (CDT) and photodynamic therapy (PDT) into one nanoplatform can produce much more reactive oxygen species (ROS) for tumor therapy. Nevertheless, it is still a great challenge to selectively generate sufficient ROS in tumor regions. Meanwhile, CDT and PDT are restricted by insufficient H2O2 content in the tumor as well as by the limited tumor tissue penetration of the light source. In this study, a smart pH/ROS-responsive nanoplatform, Fe2+@UCM-BBD, is rationally designed for tumor combination therapy. The acidic microenvironment can induce the pH-responsive release of doxorubicin (DOX), which can induce tumor apoptosis through DNA damage. Beyond that, DOX can promote the production of H2O2, providing sufficient materials for CDT. Of note, upconversion nanoparticles at the core can convert the 980 nm light to red and green light, which are used to activate Ce6 to produce singlet oxygen (1O2) and achieve upconversion luminescence imaging, respectively. Then, the ROS-responsive linker bis-(alkylthio)alkene is cleaved by 1O2, resulting in the release of Fenton reagent (Fe2+) to realize CDT. Taken together, Fe2+@UCM-BBD exhibits on-demand therapeutic reagent release capability, excellent biocompatibility, and remarkable tumor inhibition ability via synergistic chemo/photodynamic/chemodynamic combination therapy.


Assuntos
Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Clorofilídeos/química , Clorofilídeos/efeitos da radiação , Clorofilídeos/uso terapêutico , Terapia Combinada , Doxorrubicina/química , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Tratamento Farmacológico , Érbio/química , Érbio/efeitos da radiação , Érbio/uso terapêutico , Feminino , Fluoretos/química , Fluoretos/efeitos da radiação , Fluoretos/uso terapêutico , Humanos , Ferro/química , Ferro/efeitos da radiação , Ferro/uso terapêutico , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Camundongos Endogâmicos BALB C , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Itérbio/química , Itérbio/efeitos da radiação , Itérbio/uso terapêutico , Ítrio/química , Ítrio/efeitos da radiação , Ítrio/uso terapêutico
9.
Front Immunol ; 12: 682262, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34305915

RESUMO

Immunotherapy begins to be widely used due to the increasing exploration and gratifying effects in multiple cancers. Chordoma, as a rare bone malignant tumor, often recurs and metastasizes after undergoing surgery and radiotherapy. Therefore, immunotherapy can be explored as an emerging, potentially effective treatment to improve the survival rate and clinical benefit of patients. However, a variety of immune-related adverse events (irAEs) cannot be avoided completely. And the immunotherapy-induced myocarditis, as a rare but fatal irAE, has been increasingly reported. Understanding the mechanism involved in irAEs can inform best practices for side effects management. Here, we firstly reported a case of immune myocarditis and subsequent myasthenia gravis (MG) following anti-PD-1 treatment for chordoma.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Cordoma/complicações , Miastenia Gravis/complicações , Miocardite/diagnóstico , Miocardite/etiologia , Idoso , Antineoplásicos Imunológicos/uso terapêutico , Biomarcadores , Cordoma/diagnóstico , Cordoma/tratamento farmacológico , Testes de Função Cardíaca , Humanos , Imunossupressores/uso terapêutico , Testes de Função Renal , Testes de Função Hepática , Imageamento por Ressonância Magnética , Masculino , Terapia de Alvo Molecular/efeitos adversos , Miocardite/tratamento farmacológico , Tomografia por Emissão de Pósitrons , Receptor de Morte Celular Programada 1/antagonistas & inibidores
10.
Int J Hematol ; 113(4): 508-517, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33387294

RESUMO

In this study, we aimed to improve understanding of the clinical manifestations, laboratory findings, and risk factors of Clostridium perfringens sepsis in patients with acute leukemia and to analyze treatment strategies for improving prognosis. We analyzed clinical manifestations, laboratory data, diagnosis, and treatment strategies in three cases of C. perfringens sepsis in patients with acute leukemia. We also reviewed and analyzed the relevant literature, incorporating our findings into the discussion. All three patients developed septic shock with neutropenia following chemotherapy. Analysis of blood samples confirmed the presence of C. perfringens, and two patients had fulminant intravascular hemolysis and developed multiple organ dysfunction syndrome. Two patients survived and one died despite timely and full-dose antibacterial treatments, blood purification, and noninvasive positive pressure ventilation. Overall, our findings showed that C. perfringens sepsis is rare in patients with acute leukemia but progresses rapidly. A high mortality rate was observed, and patients often experienced refractory shock and intravascular hemolysis. This demonstrates the importance of early detection and diagnosis. Multimodal treatments, including fluid resuscitation, antibiotics, organ support, and blood purification, are essential for success.


Assuntos
Infecções por Clostridium/diagnóstico , Infecções por Clostridium/etiologia , Clostridium perfringens , Leucemia/complicações , Sepse/diagnóstico , Sepse/etiologia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores , Biópsia , Neutropenia Febril Induzida por Quimioterapia/diagnóstico , Neutropenia Febril Induzida por Quimioterapia/etiologia , Feminino , Humanos , Leucemia/diagnóstico , Leucemia/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Prognóstico , Resultado do Tratamento , Adulto Jovem
11.
Immunopharmacol Immunotoxicol ; 41(2): 349-360, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31056982

RESUMO

Traumatic brain injury (TBI) is a common neurotrosis disorder of the central nervous system (CNS), which has dramatic consequences on the integrity of damaged tissue. In this study, we investigated the neuroprotective effect and anti-inflammatory actions of osthole, a natural coumarin derivative, in both in vivo and in vitro TBI models. We first prepared a mouse model of cortical stab wound brain injury, investigated the capacity for osthole to prevent secondary brain injury and further examined the underlying mechanism. We revealed that osthole significantly improved the neurological function, increased the number of neurons beside injured site. Additionally, osthole treatment reduced the expression of microglia and glial scar, lowered the level of the proinflammatory cytokines interleukin (IL)-6, IL-1ß, and tumor necrosis factor-α (TNF-α), and blocked the activation of nuclear factor kappa B (NF-κB). Furthermore, the protective effect of osthole was also examined in SH-SY5Y cells subjected to scratch injury. Treatment of osthole prominently suppressed cell apoptosis and inflammatory factors release by blocking injury-induced IκB-α phosphorylation and NF-κB translocation, and upregulated the IκB-α which functions in the NF-κB signaling pathway of SH-SY5Y cells. However, NF-κB signaling pathway was inhibited by pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor, the anti-inflammatory effect of osthole was abolished. In conclusion, our findings demonstrated that osthole attenuated inflammatory response by inhibiting the NF-κB pathway in TBI.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Cumarínicos/farmacologia , Regulação para Baixo/efeitos dos fármacos , NF-kappa B/imunologia , Transdução de Sinais/efeitos dos fármacos , Animais , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/patologia , Linhagem Celular , Citocinas/imunologia , Regulação para Baixo/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Camundongos , Transdução de Sinais/imunologia
12.
Life Sci ; 225: 117-131, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30951743

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of Editor-in-Chief. The journal was initially contacted by the corresponding author to request the retraction of the article because the "same pictures were confused" in Figure 3B. The further investigation of the editor found that Dr. Elisabeth Bik has pointed out some problems of this article, including similarities between features and sections of panels within Figures 3A and 3B, as well as between Western Blots within Figures 6A,B, 8C and 10A,B, and therefore the Editor has decided to retract the article.


Assuntos
Doença de Alzheimer/prevenção & controle , Precursor de Proteína beta-Amiloide/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Cumarínicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , MicroRNAs/genética , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Animais , Modelos Animais de Doenças , Humanos , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Presenilina-1/metabolismo , Células Tumorais Cultivadas , Regulação para Cima
13.
Life Sci ; 221: 35-46, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30735733

RESUMO

AIM: Alzheimer's disease (AD), a neurodegenerative disease, is characterized by memory loss and synaptic damage. Up to now, there are limited drugs to cure or delay the state of this illness. Recently, the Fyn tyrosine kinase is implicated in AD pathology triggered by synaptic damage. Thus, Fyn inhibition may prevent or delay the AD progression. Therefore, in this paper, we investigated whether Panaxadiol could decrease synaptic damage in AD and the underlying mechanism. MAIN METHODS: The ability of learning and memory of mice has detected by Morris Water Maze. The pathological changes detected by H&E staining and Nissl staining. The percentage of cell apoptosis and the calcium concentration were detected by Flow Cytometry in vitro. The amount of synaptic protein and related proteins in the Fyn/GluN2B/CaMKIIα signaling pathway were detected by Western Blot. KEY FINDINGS: In the present article, Panaxadiol could significantly improve the ability of learning and memory of mice and reduce its synaptic dysfunction. Panaxadiol could down-regulate GluN2B's phosphorylation level by inhibition Fyn kinase activity, Subsequently, decrease Ca2+-mediated synaptic damage, reducing LDH leakage, inhibiting apoptosis in AD, resulting in facilitating the cells survival. For the underlying molecular mechanism, we used PP2 to block the Fyn/GluN2B/CaMKIIα signaling pathway. The results from WB showed that the expression of related proteins in the Fyn signaling pathway decreased with PP2 treated. SIGNIFICANCE: Our results indicate that Panaxadiol could decrease synaptic damage, which will cause AD via inhibition of the Fyn/GluN2B/CaMKIIα signaling pathway. Thus, the Panaxadiol is a best promising candidate to test as a potential therapy for AD.


Assuntos
Ginsenosídeos/metabolismo , Proteínas Proto-Oncogênicas c-fyn/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide , Animais , Linhagem Celular , Modelos Animais de Doenças , Ginsenosídeos/farmacologia , Humanos , Memória , Transtornos da Memória , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Receptores de N-Metil-D-Aspartato , Transdução de Sinais
14.
Life Sci ; 217: 16-24, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30471283

RESUMO

AIM: Alzheimer's disease (AD), a neurodegenerative disease, was characterized by the loss of memory and progressive cognitive deterioration. Up to now, there has no effective drugs to cure or delay the state of illness. Increasing evidence indicates that hyperphosphorylated tau protein plays a pivotal role in the occurrence and development of AD. Therefore, in present study, we aim to investigate whether osthole (OST) could decrease hyperphosphorylated tau protein in AD and the underlying mechanism. MAIN METHODS: The ability of learning and memory was detected by Morris Water Maze. The pathological changes were detected by H&E staining. The percentage of cells apoptosis was detected by TUNEL assay in vivo and Flow Cytometry in intro. The expressions of tau protein and related proteins in PI3K/Akt/GSK-3ß signaling pathway were detected by Western Blot. KEY FINDINGS: We found that OST could significantly improve learning and memory dysfunction, ameliorate the histology structure of damaged neural cells in hippocampal area. Moreover, we also found that OST could decrease tau protein phosphorylation as well as inhibit cells apoptosis. To explore the underlying mechanism, we used LY294002 to block PI3K/Akt/GSK-3ß signaling pathway, the results from Western bolt showed that the expression of related proteins in PI3K signaling pathway were decreased with LY294002 treated. SIGNIFICANCE: Taken together, the results indicated that OST could decrease phosphorylated tau levels via activation of PI3K/Akt/GSK-3ß signaling pathway. Thus, this study demonstrated that OST might be a potential candidate for the treatment of AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Cumarínicos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Memória/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
15.
Biol Pharm Bull ; 40(7): 1043-1054, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28674247

RESUMO

Neuroendoscopy processes can cause severe traumatic brain injury. Existing therapeutic methods, such as neural stem cell transplantation and osthole have not been proven effective. Therefore, there is an emerging need on the development of new techniques for the treatment of brain injuries. In this study we propose to combine the above stem cell based methods and then evaluate the efficiency and accuracy of the new method. Mice were randomly divided into four groups: group 1 (brain injury alone); group 2 (osthole); group 3 (stem cell transplantation); and group 4 (osthole combined with stem cell transplantation). We carried out water maze task to exam spatial memory. Immunocytochemistry was used to test the inflammatory condition of each group, and the differentiation of stem cells. To evaluate the condition of the damaged blood brain barrier restore, we detect the Evans blue (EB) extravasation across the blood brain barrier. The result shows that osthole and stem cell transplantation combined therapeutic method has a potent effect on improving the spatial memory. This combined method was more effective on inhibiting inflammation and preventing neuronal degeneration than the single treated ones. In addition, there was a distinct decline of EB extravasation in the combined treatment groups, which was not observed in single treatment groups. Most importantly, the combined usage of osthole and stem cell transplantation provide a better treatment for the traumatic brain injury caused by neuroendoscopy. The collective evidence indicates osthole combined with neural stem cell transplantation is superior than either method alone for the treatment of traumatic brain injury caused by neuroendoscopy.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Cumarínicos/uso terapêutico , Neuroendoscopia/efeitos adversos , Transplante de Células-Tronco , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/transplante
16.
Neurochem Res ; 42(2): 398-405, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27734182

RESUMO

In recent years, neural stem cell (NSC) transplantation has been widely explored as a treatment for neurodegenerative diseases. NSCs are special cells that have some capacity for self-renewal and the potential to differentiate into multiple cell types. However, the inflammatory environment of diseased tissue is not conducive to the survival of transplanted cells. Osthole (Ost) is a principal bioactive component of Fructus Cnidii, Radix Angelicae Pubescentis and other traditional Chinese medicines. Ost has a wide range of pharmacological activities, such as anti-inflammation, immunomodulation, and neuroprotection. In the present study, we assessed the protective effects of Ost on bone marrow-derived-NSCs (BM-NSCs) against injury induced by hydrogen peroxide (H2O2). BM-NSCs were pre-treated with different doses of Ost and treated with H2O2. The cell counting kit-8 (CCK-8) method and lactate dehydrogenase (LDH) leakage assay were used to determine cell viability. Using the TUNEL assay and RT-PCR, we evaluated the effect of Ost on cell apoptosis. The results showed that Ost had protective effects against H2O2-induced cell damage, and the number of apoptotic cells was significantly decreased in the Ost pre-treated groups compared to the H2O2 group. The expression ratio of Bax/Bcl-2 mRNA was also decreased. Furthermore, western blotting was used to analyze levels of proteins related to PI3K/Akt-1 signaling pathway, and results indicated that ost can increase p-Akt and PI3K. Our findings suggested that Ost protects BM-NSCs against oxidative stress injury, and it can be used to improve the inflammatory environment of neurodegenerative diseases so and promote the survival rate of transplanted NSCs.


Assuntos
Cumarínicos/farmacologia , Citoproteção/fisiologia , Células-Tronco Neurais/metabolismo , Estresse Oxidativo/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Citoproteção/efeitos dos fármacos , Relação Dose-Resposta a Droga , Peróxido de Hidrogênio/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Life Sci ; 166: 131-138, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27720999

RESUMO

AIMS: Recently, the potential for neural stem cells (NSCs) to be used in the treatment of Alzheimer's disease (AD) has been reported; however, the therapeutic effects are modest by virtue of the low neural differentiation rate. In our study, we transfected bone marrow-derived NSCs (BM-NSCs) with Neurotrophin-3 (NT-3), a superactive neurotrophic factor that promotes neuronal survival, differentiation, and migration of neuronal cells, to investigate the effects of NT-3 gene overexpression on the proliferation and differentiation into cholinergic neuron of BM-NSCs in vitro and its possible molecular mechanism. MAIN METHODS: BM-NSCs were generated from BM mesenchymal cells of adult C57BL/6 mice and cultured in vitro. After transfected with NT-3 gene, immunofluorescence and RT-PCR method were used to determine the ability of BM-NSCs on proliferation and differentiation into cholinergic neuron; Acetylcholine Assay Kit was used for acetylcholine (Ach). RT-PCR and WB analysis were used to characterize mRNA and protein level related to the Notch signaling pathway. KEY FINDINGS: We found that NT-3 can promote the proliferation and differentiation of BM-NSCs into cholinergic neurons and elevate the levels of acetylcholine (ACh) in the supernatant. Furthermore, NT-3 gene overexpression increase the expression of Hes1, decreased the expression of Mash1 and Ngn1 during proliferation of BM-NSCs. Whereas, the expression of Hes1 was down-regulated, and Mash1 and Ngn1 expression were up-regulated during differentiation of BM-NSCs. SIGNIFICANCE: Our findings support the prospect of using NT-3-transduced BM-NSCs in developing therapies for AD due to their equivalent therapeutic potential as subventricular zone-derived NSCs (SVZ-NSCs), greater accessibility, and autogenous attributes.


Assuntos
Células da Medula Óssea/citologia , Neurônios Colinérgicos/citologia , Células-Tronco Neurais/citologia , Neurogênese , Neurotrofina 3/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Células da Medula Óssea/metabolismo , Proliferação de Células , Células Cultivadas , Neurônios Colinérgicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurotrofina 3/genética , Transdução Genética , Regulação para Cima
18.
J Mol Neurosci ; 60(1): 115-29, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27389368

RESUMO

Mechanical trauma injury is a severe insult to neural cells. Subsequent secondary injury involves the release of inflammatory factors that have dramatic consequences for undamaged cells, leading to normal cell death after the initial injury. The present study investigated the capacity for arctigenin (ARC) to prevent secondary effects and evaluated the mechanism underlying the action of microRNA (miRNA)-199a and miRNA-16 in a mechanical trauma injury (MTI) model using SH-SY5Y cells in vitro. SH-SY5Y cells are often applied to in vitro models of neuronal function and differentiation. Recently, miRNAs have been demonstrated to play a crucial role in NF-κB and cholinergic signaling, which can regulate inflammation. The cell model was established by scratch-induced injury of human SH-SY5Y cells, which mimics the characteristics of MTI. A cell counting kit-8 (CCK-8), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and immunocytochemistry were used to measure cell viability. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate the inflammatory cytokine and cholinesterase (CHE) content. The lactate dehydrogenase (LDH) content was measured to assess the degree of cell injury. The mRNA levels were measured by RT-PCR to analyze ARC's mechanism of action. miRNA inhibitors and mimics were used to inhibit and strengthen the expression of miRNAs. Protein expression was detected by western blotting analysis. ARC treatment reduced the TNF-α and IL-6 levels as well as the number of TUNEL+ apoptotic SH-SY5Y cells surrounding the scratch and increased the IL-10 level compared to the controls. ARC attenuated the increase of the cell damage degree and LDH content induced by scratching, indicating increased cell survival. Mechanistic studies showed that ARC upregulated the miRNA-16 and miRNA-199a levels to reduce upstream protein (IKKα and IKKß) expression and inhibit NF-κB signaling pathway activity; moreover, the increased miRNA-199a suppresses cholinesterases to increase cholinergic signaling, resulting in decreased expression of proinflammatory cytokines. ARC treatment confers protection for SH-SY5Y cells through positive regulation of miRNA expression, thereby reducing the inflammatory response. In turn, these effects accelerate injury repair in the scratch-induced injury model. These results might provide insights into the pharmacological role of ARC in anti-inflammation and neuroprotection in neural cells.


Assuntos
Anti-Inflamatórios/farmacologia , Furanos/farmacologia , Lignanas/farmacologia , MicroRNAs/genética , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Apoptose , Lesões Encefálicas Traumáticas/metabolismo , Linhagem Celular Tumoral , Humanos , MicroRNAs/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Neurônios/metabolismo
19.
J Mol Neurosci ; 60(1): 71-81, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27394443

RESUMO

Accumulation of ß-amyloid peptide (Aß) in the brain plays an important role in the pathogenesis of Alzheimer's disease (AD). It has been reported that osthole exerts its neuroprotective effect on neuronal synapses, but its exact mechanism is obscure. Recently, microRNAs have been demonstrated to play a crucial role in inducing synaptotoxicity by Aß, implying that targeting microRNAs could be a therapeutic approach of AD. In the present study, we investigated the neuroprotective effects of osthole on a cell model of AD by transducing APP695 Swedish mutant (APP695swe, APP) into mouse cortical neurons and human SH-SY5Y cells. In this study, the cell counting kit CCK-8, apoptosis assay, immunofluorescence analysis, enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction, and Western blot assay were used. We found that osthole could enhance cell viability, prevent cell death, and reverse the reduction of synaptic proteins (synapsin-1, synaptophysin, and postsynaptic density-95) in APP-overexpressed cells, which was attributed to increases in microRNA-9 (miR-9) expression and subsequent decreases in CAMKK2 and p-AMPKα expressions. These results demonstrated that osthole plays a neuroprotective activity role in part through upregulating miR-9 in AD.


Assuntos
Doença de Alzheimer/metabolismo , Cumarínicos/farmacologia , MicroRNAs/genética , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sinapses/efeitos dos fármacos , Quinases Proteína-Quinases Ativadas por AMP , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Sinapses/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Sinaptofisina/genética , Sinaptofisina/metabolismo
20.
Front Pharmacol ; 7: 182, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27445818

RESUMO

UNLABELLED: Convection enhanced delivery (CED) infuses drugs directly into brain tissue. Needle insertion is required and results in a stab wound injury (SWI). Subsequent secondary injury involves the release of inflammatory and apoptotic cytokines, which have dramatic consequences on the integrity of damaged tissue, leading to the evolution of a pericontusional-damaged area minutes to days after in the initial injury. The present study investigated the capacity for arctigenin (ARC) to prevent secondary brain injury and the determination of the underlying mechanism of action in a mouse model of SWI that mimics the process of CED. After CED, mice received a gavage of ARC from 30 min to 14 days. Neurological severity scores (NSS) and wound closure degree were assessed after the injury. Histological analysis and immunocytochemistry were used to evaluated the extent of brain damage and neuroinflammation. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) was used to detect universal apoptosis. Enzyme-linked immunosorbent assays (ELISA) was used to test the inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-10) and lactate dehydrogenase (LDH) content. Gene levels of inflammation (TNF-α, IL-6, and IL-10) and apoptosis (Caspase-3, Bax and Bcl-2) were detected by reverse transcription-polymerase chain reaction (RT-PCR). Using these, we analyzed ARC's efficacy and mechanism of action. RESULTS: ARC treatment improved neurological function by reducing brain water content and hematoma and accelerating wound closure relative to untreated mice. ARC treatment reduced the levels of TNF-α and IL-6 and the number of allograft inflammatory factor (IBA)- and myeloperoxidase (MPO)-positive cells and increased the levels of IL-10. ARC-treated mice had fewer TUNEL+ apoptotic neurons and activated caspase-3-positive neurons surrounding the lesion than controls, indicating increased neuronal survival. CONCLUSIONS: ARC treatment confers neuroprotection of brain tissue through anti-inflammatory and anti-apoptotic effects in a mouse model of SWI. These results suggest a new strategy for promoting neuronal survival and function after CED to improve long-term patient outcome.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA