Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Chem Commun (Camb) ; 60(17): 2301-2319, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38251733

RESUMO

The emerging field of liquid biopsy has garnered significant interest in precision diagnostics, offering a non-invasive and repetitive method for analyzing bodily fluids to procure real-time diagnostic data. The precision and accuracy offered by the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (CRISPR/Cas) technology have advanced and broadened the applications of liquid biopsy. Significantly, when combined with swiftly advancing nanotechnology, CRISPR/Cas-mediated nanodevices show vast potential in precise liquid biopsy applications. However, persistent challenges are still associated with off-target effects, and the current platforms also constrain the performance of the assays. In this review, we highlight the merits of CRISPR/Cas systems in liquid biopsy, tracing the development of CRISPR/Cas systems and their current applications in disease diagnosis particularly in liquid biopsies. We also outline ongoing efforts to design nanoscale devices with improved sensing and readout capabilities, aiming to enhance the performance of CRISPR/Cas detectors in liquid biopsy. Finally, we identify the critical obstacles hindering the widespread adoption of CRISPR/Cas liquid biopsy and explore potential solutions. This feature article presents a comprehensive overview of CRISPR/Cas-mediated liquid biopsies, emphasizing the progress in integrating nanodevices to improve specificity and sensitivity. It also sheds light on future research directions in employing nanodevices for CRISPR/Cas-based liquid biopsies in the realm of precision medicine.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Medicina de Precisão
2.
Adv Mater ; 36(13): e2300665, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37437039

RESUMO

Clustered regularly interspaced short palindromic repeats/associated protein 9 (CRISPR/Cas9) gene-editing technology shows promise for manipulating single or multiple tumor-associated genes and engineering immune cells to treat cancers. Currently, most gene-editing strategies rely on viral delivery; yet, while being efficient, many limitations, mainly from safety and packaging capacity considerations, hinder the use of viral CRISPR vectors in cancer therapy. In contrast, recent advances in non-viral CRISPR/Cas9 nanoformulations have paved the way for better cancer gene editing, as these nanoformulations can be engineered to improve safety, efficiency, and specificity through optimizing the packaging capacity, pharmacokinetics, and targetability. In this review, the advance in non-viral CRISPR delivery is highlighted, and there is a discussion on how these approaches can be potentially used to treat cancers in addressing the aforementioned limitations, followed by the perspectives in designing a proper CRISPR/Cas9-based cancer nanomedicine system with translational potential.


Assuntos
Edição de Genes , Neoplasias , Humanos , Sistemas CRISPR-Cas/genética , Terapia Genética , Vetores Genéticos , Neoplasias/genética , Neoplasias/terapia
3.
Biomaterials ; 302: 122349, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37844429

RESUMO

Targeting the activated epidermal growth factor receptor (EGFR) via clustered regularly interspaced short palindromic repeat (CRISPR) technology is appealing to overcome the drug resistance of hepatocellular carcinoma (HCC) towards tyrosine kinase inhibitor (TKI) therapy. However, combining these two distinct drugs using traditional liposomes results in a suboptimal synergistic anti-HCC effect due to the limited CRISPR/Cas9 delivery efficiency caused by lysosomal entrapment after endocytosis. Herein, we developed a liver-targeting gene-hybridizing-TKI fusogenic liposome (LIGHTFUL) that can achieve high CRISPR/Cas9 expression to reverse the EGFR-mediated drug resistance for enhanced TKI-based HCC therapy efficiently. Coated with a galactose-modified membrane-fusogenic lipid layer, LIGHTFUL reached the targeting liver site to fuse with HCC tumor cells, directly and efficiently transporting interior CDK5- and PLK1-targeting CRISPR/Cas9 plasmids (pXG333-CPs) into the HCC cell cytoplasm and then the cell nucleus for efficient expression. Such membrane-fusion-mediated pXG333-CP delivery resulted in effective downregulation of both CDK5 and PLK1, sufficiently inactivating EGFR to improve the anti-HCC effects of the co-delivered TKI, lenvatinib. This membrane-fusion-participant codelivery strategy optimized the synergetic effect of CRISPR/Cas9 and TKI combinational therapy as indicated by the 0.35 combination index in vitro and the dramatic reduction of subcutaneous and orthotopic TKI-insensitive HCC tumor growth in mice. Therefore, the established LIGHTFUL provides a unique co-delivery platform to combine gene editing and TKI therapies for enhanced synergetic therapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Humanos , Camundongos , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Neoplasias Hepáticas/terapia , Nanomedicina , Tirosina
4.
Sci Adv ; 9(32): eadh2413, 2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37556535

RESUMO

Equipping multiple functionalities on adoptive effector cells is essential to overcome the complex immunological barriers in solid tumors for superior antitumor efficacy. However, current cell engineering technologies cannot endow these functionalities to cells within a single step because of the different spatial distributions of targets in one cell. Here, we present a core-shell anti-phagocytosis-blocking repolarization-resistant membrane-fusogenic liposome (ARMFUL) to achieve one-step multiplexing cell engineering for multifunctional cell construction. Through fusing with the M1 macrophage membrane, ARMFUL inserts an anti-CD47 (aCD47)-modified lipid shell onto the surface and simultaneously delivers colony-stimulating factor 1 receptor inhibitor BLZ945-loaded core into the cytoplasm. The surface-presenting aCD47 boosts macrophage's phagocytosis against the tumor by blocking CD47. The cytoplasm-located BLZ945 prompts its polarization resistance to M2 phenotype in the immunosuppressive microenvironment via inactivating the intracellular M2 polarization signaling pathway. This ARMFUL provides a versatile cell engineering platform to customize multimodal cellular functions for enhanced adoptive cell therapy.


Assuntos
Lipossomos , Neoplasias , Humanos , Lipossomos/metabolismo , Imunoterapia Adotiva , Linhagem Celular Tumoral , Fagocitose , Macrófagos/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral
5.
Adv Mater ; 35(14): e2206989, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36566024

RESUMO

Natural killer (NK) cell therapies show potential for tumor treatment but are immunologically resisted by the overexpressed immunosuppressing tumor cell surface glycans. To reverse this glycan-mediated immunosuppression, the surface NK-inhibitory glycan expressions need to be downregulated and NK-activating glycan levels should be elevated synchronously with optimal efficiency. Here, a core-shell membrane-fusogenic liposome (MFL) is designed to simultaneously achieve the physical modification of NK-activating glycans and biological inhibition of immunosuppressing glycans on the tumor cell surface via a membrane-fusion manner. Loaded into a tumor-microenvironment-triggered-degradable thermosensitive hydrogel, MFLs could be conveniently injected and controllably released into local tumor. Through fusion with tumor cell membrane, the released MFLs could simultaneously deliver sialyltransferase-inhibitor-loaded core into cytoplasm, and anchor NK-activating-glycan-modified shell onto tumor surface. This spatially-differential distribution of core and shell in one cell ensures the effective inhibition of intracellular sialyltransferase to downregulate immunosuppressing sialic acid, and direct presentation of NK-activating Lewis X trisaccharide (LeX) on tumor surface simultaneously. Consequentially, the sialic acid-caused immunosuppression of tumor surface is reprogrammed to be LeX-induced NK activation, resulting in sensitive susceptibility to NK-cell-mediated recognition and lysis for improved tumor elimination. This MFL provides a novel platform for multiplex cell engineering and personalized regulation of intercellular interactions for enhanced cancer immunotherapy.


Assuntos
Ácido N-Acetilneuramínico , Neoplasias , Humanos , Neoplasias/terapia , Membrana Celular/metabolismo , Polissacarídeos , Sialiltransferases , Terapia Baseada em Transplante de Células e Tecidos , Microambiente Tumoral
6.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(6): 1679-1687, 2022 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-36476889

RESUMO

OBJECTIVE: To explore the hematopoiesis protection effect of Danggui Buxue Tang (DBT) and its main components Angelica polysaccharide (APS) and Astragalus polysaccharide (ASPS) on myelosuppression mice, and the mechanism of anti-apoptosis of Meg-01 cells. METHODS: Mice were radiated with 4 Gy of 137Csγ ray to establish the model of radiation-induced myelosuppression. DBT, APS or ASPS (10 mg/kg) were injected into irradiated mice. Peripheral blood cell counts were performed on mice before radiation (day 0) and day 7, 14 and 21 after radiation. On the 21st day, poor plasma platelets were collected from mice to detect TPO concentration and then the mice were sacrificed. The femoral bone marrow cells were cultured for colony cell forming units (CFU). Meg-01 cells were cultured without FBS for 24 h to induce apoptosis, and then treated with DBT/APS/ASPS for 72 h. Flow cytometry (FCM) was used to detect early apoptosis (Annexin V), mitochondrial membrane potential (JC-1) and the expression of Caspase-3 to analyze the effect of DBT/APS/ASPS on cell apoptosis. RESULTS: DBT can stimulate the recovery of white blood cells (WBC), red blood cells (RBC) and platelets (PLT) of myelosuppression mice, especially for WBC and PLT (P<0.01, P<0.05). Compared with the control group, the number of BFU-E, CFU-MK and CFU-GM increased after adding DBT (BFU-E & CFU-GM: P<0.05; CFU-MK: P<0.01). The effect of DBT on blood TPO concentration in mice was not obvious (P=0.89). RBC, WBC and PLT were increased in APS group compared with control group (P<0.05). WBC increased after the treatment of ASPS (P<0.05). APS stimulated the formation of CFU-F, CFU-MK and CFU-GM (P<0.05). Only CFU-GM increased in ASPS group(P<0.05). Besides, DBT decreased the apoptosis of Meg-01 cells (P<0.05). The early apoptosis rate and total death rate in APS (100 µg/ml) group were lower than that of control group (P<0.01, P<0.05). The early apoptosis rate of ASPS (100 µg/ml) group was lower than that of control group (P<0.05). JC-1 and Caspase-3 showed that APS (100 µg/ml) significantly reduced apoptosis rate (P<0.01, P<0.05). CONCLUSION: DBT has protective effect on hematopoietic system, especially WBC and PLT, and has anti-apoptotic effect on Meg-01. It was found that the above effects of DBT were mainly caused by APS, and its anti-apoptosis mechanism was carried out mainly through JC-1 and Caspase-3 pathways.


Assuntos
Medula Óssea , Polissacarídeos , Camundongos , Animais , Caspase 3
7.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(6): 1873-1880, 2022 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-36476919

RESUMO

OBJECTIVE: To investigate the hematopoietic protective effect of platelet-derived growth factor (PDGF)-BB on radiation-induced myelosuppression model mice and effect of anti-apoptosis of megakaryocyte line Meg-01 cells, and its possible mechanism. METHODS: Mice were radiated with 4 Gy of 137Csγ ray to establish the model of myelosuppression. Mice were weighed and peripheral blood cell were counted before radiation (day 0) and day 7, 14 and 21 after radiation. On the 21 st day, the mice were killed. The sternal tissues of the mice were taken for morphological observation, and the femoral bone marrow cells were cultured for the assay of colony cell forming units (CFU). Meg-01 cells were cultured without FBS for 24 h to induce apoptosis, and then treated with PDGF-BB for 48 h. The effects of PDGF-BB on the proliferation were investigated by cell counting. Flow cytometry was used to detect early apoptosis (Annexin V), mitochondrial membrane potential (JC-1) and the expression of caspase-3. RESULTS: Peripheral blood cell counts of mice showed that PDGF-BB stimulated the recovery of white blood cells, red blood cells and platelets after radiation (P<0.05), especially for white blood cells. Morphological examination showed bone marrow hyperplasia in PDGF-BB group, the numbers of megakaryocytes and their progenitor cells were higher than those in the control group. PDGF-BB significantly stimulated the formation of CFU-MK, CFU-GM, BFU-E and CFU-F. PDGF-BB showed a strong proliferation effect in the concentration range of 5-50 ng/ml (P<0.001). PDGF-BB (50 ng/ml) significantly reduced the positive expression of Annexin V (P<0.01). The mitochondrial membrane potential in the control group was decreased when compared with PDGF-BB group, which indicated that the number of apoptotic cells was increased (P<0.01). Besides, the expression of caspase-3 in PDGF-BB group was significantly lower than that in control group (P<0.05). CONCLUSION: PDGF-BB has a protective effect on the hematopoietic system of myelosuppression model mice, especially megakaryocytes and their progenitor cells. PDGF-BB has pro-proliferative and anti-apoptotic effects on Meg-01 cells, and the mechanism may be mediated through JC-1 and caspase-3 pathway.


Assuntos
Sistema Hematopoético , Animais , Camundongos , Becaplermina , Caspase 3 , Apoptose
8.
Gen Physiol Biophys ; 41(6): 499-509, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36454111

RESUMO

The ubiquitin-editing enzyme TNF alpha-induced protein 3 (TNFAIP3) emerges protective roles in neurological disorder, such as cerebral trauma. However, the molecular mechanisms of TNFAIP3 in epilepsy are not very clear. Hereon, the epileptic mouse models and BV2 microglial cellular models were established by kainic acid (KA) and lipopolysaccharide (LPS) respectively. We found that TNFAIP3 was highly expressed in the hippocampus of epileptic mice. Besides, TNFAIP3 overexpression relieved the spatial learning and memory, reduced the hot plate latency, as well as inhibited neuronal apoptosis in KA-treated mice. In vivo and in vitro experiments indicated that inflammation, a key characteristic of epilepsy, was inhibited by TNFAIP3 upregulation, as evidenced by the downregulated expression of pro-inflammatory cytokine interleukin (IL)-1ß and inducible NO synthase (iNOS), along with the decreased levels of NLRP3 inflammasome, which could activate inflammation. Collectively, we infer that TNFAIP3 relieves neuronal injury in epilepsy by suppressing inflammation.


Assuntos
Epilepsia , Neuroproteção , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Animais , Camundongos , Epilepsia/induzido quimicamente , Hipocampo , Inflamação , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética
9.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(4): 998-1004, 2022 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-35981353

RESUMO

OBJECTIVE: To investigate the effect of thrombospondin-1 (TSP-1) on apoptosis of human megakaryocytic leukemia cell line Meg-01 and its possible mechanism. METHODS: The expression of CD36 antigen in Meg-01 cells was detected by flow cytometry and immunocytochemistry. Meg-01 cells were cultured for 48 hours with TSP-1 and CD36 antibody FA6-152 at different concentrations. The early apoptosis and activity of caspase-3 were detected by flow cytometry. The effect of TSP-1 on the growth and differentiation of megakaryocytes was investigated by cell counting and CFU-MK culture. RESULTS: The flow cytometry and immunocytochemistry showed that CD36 antigen was expressed on the surface of Meg-01 cells. TSP-1 (5 µg/ml) inhibited the growth of Meg-01 cells, but had unobvious effect on M-07e cells. After addition of CD36 antibody FA6-152 (5, 10, and 25 µg/ml), the inhibition effect of TSP-1 was significantly reduced. TSP-1 (2.5, 5, and 7.5 µg/ml) increased the positive expression of Annexin V (P<0.01) and caspase-3 activity (P<0.01), which indicated that TSP-1 had a significant effect on inducing apoptosis. After addition of CD36 antibody FA6-152 (25 µg/ml), the apoptosis induced by TSP-1 in Meg-01 cells was significantly reduced. TSP-1 (5, 10, and 25 µg/ml) could significantly inhibit the formation of CFU-MK in mouse bone marrow cells, while ß-TG could not. CD36 antibody FA6-152 (25 µg/ml) could significantly reduce the inhibition of TSP-1 on CFU-MK. CONCLUSION: TSP-1 may induce apoptosis of megakaryocytic leukemia cell line Meg-01 cells via CD36/caspase-3, which provides a potential new drug development and treatment target for clinical treatment of megakaryocytic leukemia.


Assuntos
Leucemia Megacarioblástica Aguda , Trombospondina 1 , Animais , Apoptose , Antígenos CD36/metabolismo , Caspase 3/metabolismo , Linhagem Celular , Humanos , Camundongos , Trombospondina 1/metabolismo , Trombospondina 1/farmacologia
10.
Adv Sci (Weinh) ; 8(24): e2102051, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34665528

RESUMO

Liver disease, particularly viral hepatitis and hepatocellular carcinoma (HCC), is a global healthcare burden and leads to more than 2 million deaths per year worldwide. Despite some success in diagnosis and vaccine development, there are still unmet needs to improve diagnostics and therapeutics for viral hepatitis and HCC. The emerging clustered regularly interspaced short palindromic repeat/associated proteins (CRISPR/Cas) technology may open up a unique avenue to tackle these two diseases at the genetic level in a precise manner. Especially, liver is a more accessible organ over others from the delivery point of view, and many advanced strategies applied for nanotheranostics can be adapted in CRISPR-mediated diagnostics or liver gene editing. In this review, the focus is on these two aspects of viral hepatitis and HCC applications. An overview on CRISPR editor development and current progress in clinical trials is first given, followed by highlighting the recent advances integrating the merits of gene editing and nanotheranostics. The promising systems that are used in other applications but may hold potentials in liver gene editing are also discussed. This review concludes with the perspectives on rationally designing the next-generation CRISPR approaches and improving the editing performance.


Assuntos
Sistemas CRISPR-Cas/genética , Carcinoma Hepatocelular/terapia , Edição de Genes/métodos , Terapia Genética/métodos , Hepatite Viral Humana/terapia , Neoplasias Hepáticas/terapia , Nanomedicina Teranóstica/métodos , Carcinoma Hepatocelular/genética , Hepatite Viral Humana/genética , Humanos , Neoplasias Hepáticas/genética
11.
Int J Biol Sci ; 17(2): 562-573, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33613113

RESUMO

Identifying high specificity and sensitivity biomarkers has always been the focus of research in the field of non-invasive cancer diagnosis. Exosomes are extracellular vesicles with a lipid bilayer membrane that can be released by all types of cells, which contain a variety of proteins, lipids, and a variety of non-coding RNAs. Increasing research has shown that the lipid bilayer can effectively protect the nucleic acid in exosomes. In cancers, tumor cell-derived exosomal circRNAs can act on target cells or organs through the transport of exosomes, and then participate in the regulation of tumor development and metastasis. Since exosomes exist in various body fluids and circRNAs in exosomes exhibit high stability, exosomal circRNAs have the potential as biomarkers for early and minimally invasive cancer diagnosis and prognosis judgment. In this review, we summarized circRNAs and their biological roles in cancers, with the emerging value biomarkers in cancer diagnosis, disease judgment, and prognosis observation. In addition, we briefly compared the advantages of exosomal circRNAs as biomarkers and the current obstacles in the exosome isolation technology, shed light to the future development of this technology.


Assuntos
Biomarcadores Tumorais/metabolismo , Exossomos/metabolismo , Neoplasias/metabolismo , RNA Circular/metabolismo , Animais , Humanos
12.
Onco Targets Ther ; 13: 6681-6697, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32764959

RESUMO

PURPOSE: Pediatric acute promyelocytic leukemia (APL) accounts for 10% of pediatric acute myelogenous leukemia (AML) case and is accompanied by a tendency to hemorrhage. miR-188-5p plays an important role in adult AML. Therefore, the purpose of this study was to explore the effects of miR-188-5p on cell proliferation and apoptosis and tumor growth, and its mechanism in pediatric APL patients. MATERIALS AND METHODS: Survival-associated miRNAs or mRNAs from TCGA database associated with AML were identified via using the "survival R" package in R language. CCK8, clone formation, flow cytometry, RT-PCR, immunohistochemistry and Western blot assays were used to detect the viability, proliferation, apoptosis, cell cycle, and related gene expression in APL cell lines. The prognostic value of miR-188-5p was evaluated using a ROC curve. The tumorigenic ability of APL cell lines was determined using a nude mouse transplantation tumor experiment. Tumor cell apoptosis was determined by TUNEL assay in vivo. The target genes of miR-188-5p were predicted using the miRDB, miRTarBase, and TargetScan databases. A PPI network was constructed using STRING database and the hub gene was identified using the MCODE plug-in of the Cytoscape software. The DAVID database was used to perform GO and KEGG pathway enrichment analyses. A luciferase reporter assay was used to demonstrate the binding of miR-188-5p to CD2AP. RESULTS: miR-188-5p overexpression or CD2 associated protein (CD2AP) inhibition was significantly associated with poor survival in pediatric APL patients. Upregulation of miR-188-5p was identified in the blood of pediatric APL patients and cell lines. Increased expression of miR-188-5p also promoted the viability, proliferation, and cell cycle progression, and reduced the apoptosis of APL cells. Additionally, upregulation of miR-188-5p regulated the expressions of cyclinD1, p53, Bax, Bcl-2 and cleaved caspase-3. The area under the ROC curve (AUC) of miR-188-5p was 0.661. miR-188-5p overexpression increased the tumorigenic ability of APL and Ki67 expression, and reduced cell apoptosis in vivo. CD2AP was identified as the only overlapping gene from the list of miR-188-5p target genes and survival-related mRNAs of the TCGA database. It was mainly enriched in the "biological process (BP)" and "cellular component (CC)" terms, and was downregulated in the blood of pediatric APL patients and cell lines. The luciferase reporter, RT-PCR, and Western blot assays demonstrated that the binding of miR-188-5p to CD2AP. CD2AP inhibition promoted the proliferation and inhibited the apoptosis of APL cells. Rescue experiments showed that inhibition of miR-188-5p inhibited cell proliferation, activated the PI3K/AKT/mTOR signaling pathway, induced G0/G1 phase arrest, regulated gene expression, and promoted cell apoptosis, which were reversed by CD2AP inhibition. CONCLUSION: miR-188-5p, an oncogene, promoted tumor growth and progression of pediatric APL in vitro and in vivo via targeting CD2AP and activating the PI3K/AKT/mTOR signaling pathway.

13.
Drug Des Devel Ther ; 14: 1813-1823, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32494123

RESUMO

INTRODUCTION: Berberine has been reported to inhibit cancer cell growth by apoptosis induction and exhibits a protective role against cancer progression. The current study aims to investigate the effects of berberine on acute lymphoblastic leukemia (ALL) and the mechanism beyond apoptosis. METHODS: Cell viability was determined in ALL cell lines EU-6 and SKW-3 using trypan blue staining. Cell autophagy was determined by immunofluorescence and Western blot. ALL xenograft mice were established to investigate the anti-tumor effects of BBR. The molecular mechanism was explored in ALL cell lines using siRNA and signaling inhibitors. RESULTS: Herein, we show that berberine treatment significantly inhibits ALL cell viability and promotes cell death by inducing autophagy in a dose-dependent manner. Moreover, berberine significantly alleviates the aggressive pathological condition in ALL xenograft mice. Mechanistic studies exhibit that berberine induces autophagic death in ALL cells by inactivating AKT/mTORC1 signaling. Chemically targeting AKT/mTORC1 signaling controls berberine-induced cell autophagy in vitro, and blockade of autophagic process blunts berberine-alleviated pathological condition in vivo. DISCUSSION: In conclusion, our study reveals that berberine could induce ALL cell autophagic death by inactivating AKT/mTORC1 signaling that could be used to develop small molecule drug for ALL treatment.


Assuntos
Antineoplásicos/farmacologia , Berberina/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Adolescente , Adulto , Idoso , Morte Celular Autofágica/efeitos dos fármacos , Criança , Pré-Escolar , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Pessoa de Meia-Idade , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem
14.
J Mol Neurosci ; 57(1): 28-37, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25957996

RESUMO

Astrocyte activation, associated with the release of pro-inflammatory cytokines interleukin 1-ß (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α), is a hallmark of multiple brain diseases, including mesial temporal lobe epilepsy. In recent years, several microRNAs have emerged as important controllers of Toll-like receptor (TLR) signaling. In this study, we investigated the effect of miR-132, miR-146a, and miR-155 on myeloid-related protein-8 (MRP8) induced astrocyte-related inflammation. Using quantitative polymerase chain reaction (qPCR) and western blot, we found clear upregulation of TLR4 and downstream inflammatory cytokines, along with dysregulation of miR-132, miR-146a, and miR-155 in in vitro astrocytes after exposing them to different concentrations of MRP8. In addition, we focused on the effect of miR-132 on astrocyte-related inflammation induced by MRP8 via lentiviral infection then evaluated the expression of its possible target genes: acetylcholinesterase (AChE) and interleukin-1 receptor-associated kinase (IRAK4). Our results show that miR-132 is a negative feedback regulator of IL-1ß and IL-6, but not TNF-α, by targeting IRAK4. Together, our findings demonstrate the novel role of TLR4-related microRNAs, especially miR-132, in the regulation of MRP8-induced astrocyte activation and highlight the importance of miR-132 in the modulation of innate immune response induced by endogenous ligands in neurological diseases.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Astrócitos/metabolismo , MicroRNAs/genética , Receptor 4 Toll-Like/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Acetilcolinesterase/genética , Acetilcolinesterase/metabolismo , Linhagem Celular Tumoral , Humanos , Inflamação/metabolismo , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
J Neuroimmunol ; 282: 110-7, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25903737

RESUMO

Recently, the role of inflammation in the pathogenesis of mesial temporal lobe epilepsy (MTLE) has garnered great attention. Increasing evidence indicated that interleukin-1ß (IL-1ß) plays a critical role in the pathogenesis of MTLE. In this study, we cultured primary hippocampal neurons, using IL-1ß to mimic the process of inflammatory reaction in neurons, then inhibited the inflammation using inhibitors of the PI3K/Akt/mTOR signaling pathway. The expression of proteins related to the PI3K/Akt/mTOR signaling pathway in rat hippocampal neurons was detected by western blot, and similar methods were applied to the hippocampi obtained from children with MTLE and normal controls. Neuronal somatic size and dendritic length were measured by immunohistochemistry and digital imaging. We observed that stimulation with IL-1ß in neuron led to the up-regulation of p-Akt and p70S6K and promoted the growth of cell somatic size and dendritic length via the PI3K/Akt/mTOR signaling pathway. Pre-treatment with inhibitors of the pathway, LY294002 and rapamycin, decreased the expression of p-Akt and p70S6K and alleviated the morphological changes induced by IL-1ß in hippocampal neurons. We further verified the increasement of P-Akt and p70S6K in the hippocampi of children with MTLE. These data are the first to demonstrate that the inflammatory response induced by IL-1ß promotes seizures and plays an important role in the pathogenesis of MTLE via the PI3K/Akt/mTOR signaling pathway. Therefore, modulation of the PI3K/Akt/mTOR signaling pathway may be a novel therapeutic target for the treatment of MTLE.


Assuntos
Epilepsia do Lobo Temporal/patologia , Hipocampo/citologia , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Criança , Cromonas/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Humanos , Interleucina-1beta/farmacologia , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
16.
Physiol Behav ; 143: 104-12, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25700894

RESUMO

Although the pathogenesis and epileptogenesis of mesial temporal lobe epilepsy (MTLE) have been studied for years, many questions remain. The ubiquitin-proteasome system (UPS) is one factor that might regulate ion channels, inflammation and neuron excitability. Nedd4-2 is an E3 ubiquitin ligase linked with ion channels and synaptic vesicle recycling. Here, we explore the role of the UPS and its E3 ligase Nedd4-2 in the pathogenesis of MTLE. Our western blot results revealed that ubiquitin and Nedd4-2 were expressed differentially in different stages of MTLE. Co-immunoprecipitation and double immunostaining results indicated that Nedd4-2 was the substrate protein of ubiquitin both in vivo and in vitro. Inhibition of the UPS aggravated the epileptogenesis of MTLE, causing early and frequent spontaneous seizures, more obvious neuron loss and aberrant mossy fiber sprouting. Inhibition of ubiquitin also enhanced the activation of Nedd4-2, and switched ion channel α-ENaC downstream. Our study is the first to report that the UPS participates in the pathogenesis of MTLE, inhibition of UPS could aggravate the epileptogenesis, and that Nedd4-2 is a critical E3 ligase involved in this process.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Regulação da Expressão Gênica , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Monofosfato de Adenosina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Antimaníacos/uso terapêutico , Células Cultivadas , Inibidores de Cisteína Proteinase/uso terapêutico , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Imunoprecipitação , Leupeptinas/uso terapêutico , Cloreto de Lítio/uso terapêutico , Masculino , Agonistas Muscarínicos/toxicidade , Ubiquitina-Proteína Ligases Nedd4 , Neurônios/efeitos dos fármacos , Pilocarpina/toxicidade , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção , Ubiquitina/genética , Ubiquitinas/farmacologia
17.
Mol Neurobiol ; 49(1): 337-51, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23982744

RESUMO

The role of Toll-like receptor 4 (TLR4) in the activation of innate immunity has been extensively studied in the past several years. Here, we are the first to report that myeloid-related protein 8 (MRP8), an endogenous TLR4 ligand, is involved in the epileptogenesis of mesial temporal lobe epilepsy (MTLE). We find that the expression of MRP8, TLR4, and interleukin 1-ß (IL-1ß) was upregulated in a MTLE model during both acute and chronic disease stages. We next investigated the possible roles played by astrocytes, which have been shown to be the major source of IL-1ß during epilepsy. Stimulation via MRP8 led to the induction of IL-1ß in astrocytes in vitro, accompanied by the activation of Nuclear Factor-κB, while knockdown of TLR4 or inhibition of NF-κB in astrocytes prevented this IL-1ß induction. Thus, MRP8 may potentiate the perpetuation of MTLE by activating the NF-κB pathway in astrocytes, and could be a new target for anticonvulsant therapies.


Assuntos
Astrócitos/metabolismo , Calgranulina A/biossíntese , Epilepsia do Lobo Temporal/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/biossíntese , Animais , Astrócitos/patologia , Células Cultivadas , Criança , Epilepsia do Lobo Temporal/patologia , Feminino , Humanos , Ligantes , Masculino , Ratos , Ratos Sprague-Dawley
18.
ScientificWorldJournal ; 2013: 208309, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24170980

RESUMO

Astrocytes are now recognized as a heterogeneous class of cells with many important and diverse functions in healthy and diseased central nervous system (CNS). MicroRNAs (miRNAs) are small, noncoding RNAs which may have key roles in astrocytes activation in response to various stimuli. We performed quantitative real-time PCR (qPCR) to detect changes in the expressions of brain-enriched miRNAs (124, 134, 9, 132, and 138), inflammation-related miRNAs (146a, 21, 181a, 221, and 222), and tumor necrosis factor alpha (TNF- α ) in the rat primary astrocyte cultures after stimulation with myeloid-related protein 8 (MRP8) and lipopolysaccharides (LPS). Further, we inhibited the expression of TNF- α in the astrocytes by using TNF- α inhibitor (lenalidomide) and tested for the first time the effect of this inhibition on the expressions of the same tested miRNAs. Stimulation of the astrocytes with MRP8 or LPS leads to significant upregulation of miRNAs (124, 134, 9, 132, 146a, 21, 181a, 221, and 222), while miRNA-138 was downregulated. TNF- α inhibition with lenalidomide leads to opposite expressions of the tested miRNAs. These miRNAs may play an important role in activation of the astrocytes and may be a novel target for cell-specific therapeutic interventions in multiple CNS diseases.


Assuntos
Astrócitos/efeitos dos fármacos , Calgranulina A , Lipopolissacarídeos/farmacologia , MicroRNAs , Talidomida/análogos & derivados , Fator de Necrose Tumoral alfa/genética , Animais , Astrócitos/citologia , Encéfalo/fisiologia , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Lenalidomida , Masculino , Ratos , Ratos Sprague-Dawley , Talidomida/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
19.
J Mol Neurosci ; 51(3): 950-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23636891

RESUMO

Recently, the role of inflammation has attracted great attention in the pathogenesis of mesial temporal lobe epilepsy (MTLE), and microRNAs start to emerge as promising new players in MTLE pathogenesis. In this study, we investigated the dynamic expression patterns of tumor necrosis factor alpha (TNF-α) and microRNA-155 (miR-155) in the hippocampi of an immature rat model of status epilepticus (SE) and children with MTLE. The expressions of TNF-α and miR-155 were significantly upregulated in the seizure-related acute and chronic stages of MTLE in the immature rat model and also in children with MTLE. Modulation of TNF-α expression, either by stimulation using myeloid-related protein (MRP8) or lipopolysaccharide or inhibition using lenalidomide on astrocytes, leads to similar dynamic changes in miR-155 expression. Our study is the first to focus on the dynamic expression pattern of miR-155 in the immature rat of SE lithium-pilocarpine model and children with MTLE and to detect their relationship at the astrocyte level. TNF-α and miR-155, having similar expression patterns in the three stages of MTLE development, and their relationship at the astrocyte level may suggest a direct interactive relationship during MTLE development. Therefore, modulation of the TNF-α/miR-155 axis may be a novel therapeutic target for the treatment of MTLE.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , MicroRNAs/metabolismo , Estado Epiléptico/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adolescente , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Estudos de Casos e Controles , Criança , Epilepsia do Lobo Temporal/patologia , Feminino , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Humanos , Lenalidomida , Lipopolissacarídeos/farmacologia , Cloreto de Lítio , Masculino , MicroRNAs/genética , Pilocarpina , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente , Talidomida/análogos & derivados , Talidomida/farmacologia , Fator de Necrose Tumoral alfa/genética
20.
J Med Virol ; 82(7): 1143-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20513076

RESUMO

Chronic hepatitis B virus (CHBV) infection causes cirrhosis and hepatocellular carcinoma. Lamivudine (LAM) has been successfully used to treat CHBV infections but prolonged use leads to the emergence of drug-resistant variants. This is primarily linked to a mutation in the tyrosine-methionine-aspartate-aspartate (YMDD) motif of the HBV polymerase gene at position 204. Rapid diagnosis of drug-resistant HBV is necessary for a prompt treatment response. Common diagnostic methods such as sequencing and restriction fragment length polymorphism (RFLP) analysis lack sensitivity and require significant processing. The aim of this study was to demonstrate the usefulness of a novel diagnostic method that combines polymerase chain reaction (PCR), ligase detection reaction (LDR) and a nucleic acid detection strip (NADS) in detecting site-specific mutations related to HBV LAM resistance. We compared this method (PLNA) to direct sequencing and RFLP analysis in 50 clinical samples from HBV infected patients. There was 90% concordance between all three results. PLNA detected more samples containing mutant variants than both sequencing and RFLP analysis and was more sensitive in detecting mixed variant populations. Plasmid standards indicated that the sensitivity of PLNA is at or below 3,000 copies per ml and that it can detect a minor variant at 5% of the total viral population. This warrants its further development and suggests that the PLNA method could be a useful tool in detecting LAM resistance.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/genética , Vírus da Hepatite B/efeitos dos fármacos , Hepatite B Crônica/virologia , Lamivudina/uso terapêutico , Reação em Cadeia da Polimerase/métodos , Inibidores da Transcriptase Reversa/uso terapêutico , Antivirais/uso terapêutico , DNA Viral/genética , Vírus da Hepatite B/genética , Hepatite B Crônica/tratamento farmacológico , Humanos , Reação em Cadeia da Ligase , Mutação/efeitos dos fármacos , Polimorfismo de Fragmento de Restrição , Fitas Reagentes , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA