Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cancer Res ; 84(1): 84-100, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-37874330

RESUMO

Expanding the utility of chimeric antigen receptor (CAR)-T cells in solid tumors requires improving their efficacy and safety. Hypoxia is a feature of most solid tumors that could be used to help CAR-T cells discriminate tumors from normal tissues. In this study, we developed hypoxia-responsive CAR-T cells by engineering the CAR to be under regulation of hypoxia-responsive elements and selected the optimal structure (5H1P-CEA CAR), which can be activated in the tumor hypoxic microenvironment to induce CAR-T cells with high polyfunctionality. Hypoxia-responsive CAR T cells were in a "resting" state with low CAR expression under normoxic conditions. Compared with conventional CAR-T cells, hypoxia-responsive CAR-T cells maintained lower differentiation and displayed enhanced oxidative metabolism and proliferation during cultivation, and they sowed a capacity to alleviate the negative effects of hypoxia on T-cell proliferation and metabolism. Furthermore, 5H1P-CEA CAR-T cells exhibited decreased T-cell exhaustion and improved T-cell phenotype in vivo. In patient-derived xenograft models, hypoxia-responsive CAR-T cells induced more durable antitumor activity than their conventional counterparts. Overall, this study provides an approach to limit CAR expression to the hypoxic tumor microenvironment that could help to enhance CAR T-cell efficacy and safety in solid tumors. SIGNIFICANCE: Engineering CAR-T cells to upregulate CAR expression under hypoxic conditions induces metabolic reprogramming, reduces differentiation, and increases proliferation to enhance their antitumor activity, providing a strategy to improve efficacy and safety.


Assuntos
Imunoterapia Adotiva , Neoplasias , Humanos , Neoplasias/metabolismo , Linfócitos T , Hipóxia/metabolismo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Front Oncol ; 11: 596789, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996539

RESUMO

Circulating tumor DNA (ctDNA) is a promising biomarker for accurate monitoring and less invasive assessment of tumor burden and treatment response. Here, targeted next-generation sequencing (NGS) with a designed gene panel of 176 cancer-relevant genes was used to assess mutations in 90 ctDNA samples from 90 patients with multiple types of liver disease and 10 healthy donor samples for control. Using our ctDNA detection panel, we identified mutations in 98.89% (89/90) of patient plasma biopsy samples, and 19 coding variants located in 10 cancer-related genes [ACVR2A, PCLO, TBCK, adhesion G protein-coupled receptor (ADGRV1), COL1A1, GABBR1, MUC16, MAGEC1, FASLG, and JAK1] were identified in 96.7% of patients (87/90). The 10 top mutated genes were tumor protein p53 (TP53), ACVR2A, ADGRV1, MUC16, TBCK, PCLO, COL11A1, titin (TTN), DNAH9, and GABBR1. TTN and TP53 and TTN and DNAH9 mutations tended to occur together in hepatocellular carcinoma samples. Most importantly, we found that most of those variants were insertions (frameshift insertions) and deletions (frameshift deletions and in-frame deletions), such as insertion variants in ACVR2A, PCLO, and TBCK; such mutations were detected in almost 95% of patients. Our study demonstrated that the targeted NGS-based ctDNA mutation profiling was a useful tool for hepatocellular carcinoma (HCC) monitoring and could potentially be used to guide treatment decisions in HCC.

3.
Stem Cell Res Ther ; 12(1): 86, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33494824

RESUMO

BACKGROUND: Leukemia stem cells (LSCs) are responsible for the initiation, progression, and relapse of acute myeloid leukemia (AML). Therefore, a therapeutic strategy targeting LSCs is a potential approach to eradicate AML. In this study, we aimed to identify LSC-specific surface markers and uncover the underlying mechanism of AML LSCs. METHODS: Microarray gene expression data were used to investigate candidate AML-LSC-specific markers. CD9 expression in AML cell lines, patients with AML, and normal donors was evaluated by flow cytometry (FC). The biological characteristics of CD9-positive (CD9+) cells were analyzed by in vitro proliferation, chemotherapeutic drug resistance, migration, and in vivo xenotransplantation assays. The molecular mechanism involved in CD9+ cell function was investigated by gene expression profiling. The effects of alpha-2-macroglobulin (A2M) on CD9+ cells were analyzed with regard to proliferation, drug resistance, and migration. RESULTS: CD9, a cell surface protein, was specifically expressed on AML LSCs but barely detected on normal hematopoietic stem cells (HSCs). CD9+ cells exhibit more resistance to chemotherapy drugs and higher migration potential than do CD9-negative (CD9-) cells. More importantly, CD9+ cells possess the ability to reconstitute human AML in immunocompromised mice and promote leukemia growth, suggesting that CD9+ cells define the LSC population. Furthermore, we identified that A2M plays a crucial role in maintaining CD9+ LSC stemness. Knockdown of A2M impairs drug resistance and migration of CD9+ cells. CONCLUSION: Our findings suggest that CD9 is a new biomarker of AML LSCs and is a promising therapeutic target.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Neoplásicas , Animais , Biomarcadores , Resistência a Medicamentos , Células-Tronco Hematopoéticas , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Camundongos , Tetraspanina 29/genética
4.
Gastroenterology ; 158(3): 664-678.e24, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31678303

RESUMO

BACKGROUND & AIMS: Immune checkpoint inhibitors have some efficacy in the treatment of hepatocellular carcinoma (HCC). Programmed cell death 1 ligand 1 (PD-L1), expressed on some cancer cells, binds to the receptor programmed cell death 1 (PDCD1, also called PD1) on T cells to prevent their proliferation and reduce the antigen-tumor immune response. Immune cells that infiltrate some types of HCCs secrete interferon gamma (IFNG). Some HCC cells express myocyte enhancer factor 2D (MEF2D), which has been associated with shorter survival times of patients. We studied whether HCC cell expression of MEF2D regulates expression of PD-L1 in response to IFNG. METHODS: We analyzed immune cells from 20 fresh HCC tissues by flow cytometry. We analyzed 225 fixed HCC tissues (from 2 cohorts) from patients in China by immunohistochemistry and obtained survival data. We created mice with liver-specific knockout of MEF2D (MEF2DLPC-KO mice). We knocked out or knocked down MEF2D, E1A binding protein p300 (p300), or sirtuin 7 (SIRT7) in SMMC-7721, Huh7, H22, and Hepa1-6 HCC cell lines, some incubated with IFNG. We analyzed liver tissues from mice and cell lines by RNA sequencing, immunoblot, dual luciferase reporter, and chromatin precipitation assays. MEF2D protein acetylation and proteins that interact with MEF2D were identified by coimmunoprecipitation and pull-down assays. H22 cells, with MEF2D knockout or without (controls), were transplanted into BALB/c mice, and some mice were given antibodies to deplete T cells. Mice bearing orthotopic tumors grown from HCC cells, with or without knockout of SIRT7, were given injections of an antibody against PD1. Growth of tumors was measured, and tumors were analyzed by immunohistochemistry and flow cytometry. RESULTS: In human HCC specimens, we found an inverse correlation between level of MEF2D and numbers of CD4+ and CD8+ T cells; level of MEF2D correlated with percentages of PD1-positive or TIM3-positive CD8+ T cells. Knockout of MEF2D from H22 cells reduced their growth as allograft tumors in immune-competent mice but not in immune-deficient mice or mice with depletion of CD8+ T cells. When MEF2D-knockout cells were injected into immune-competent mice, they formed smaller tumors that had increased infiltration and activation of T cells compared with control HCC cells. In human and mouse HCC cells, MEF2D knockdown or knockout reduced expression of PD-L1. MEF2D bound the promoter region of the CD274 gene (encodes PD-L1) and activated its transcription. Overexpression of p300 in HCC cells, or knockout of SIRT7, promoted acetylation of MEF2D and increased its binding, along with acetylated histones, to the promoter region of CD274. Exposure of HCC cells to IFNG induced expression of p300 and its binding MEF2D, which reduced the interaction between MEF2D and SIRT7. MEF2D-induced expression of PD-L1 upon IFNG exposure was independent of interferon-regulatory factors 1 or 9. In HCC cells not exposed to IFNG, SIRT7 formed a complex with MEF2D that attenuated expression of PD-L1. Knockout of SIRT7 reduced proliferation of HCC cells and growth of tumors in immune-deficient mice. Compared with allograft tumors grown from control HCC cells, in immune-competent mice, tumors grown from SIRT7-knockout HCC cells expressed higher levels of PD-L1 and had reduced infiltration and activation of T cells. In immune-competent mice given antibodies to PD1, allograft tumors grew more slowly from SIRT7-knockout HCC cells than from control HCC cells. CONCLUSIONS: Expression of MEF2D by HCC cells increases their expression of PD-L1, which prevents CD8+ T-cell-mediated antitumor immunity. When HCC cells are exposed to IFNG, p300 acetylates MEF2D, causing it to bind the CD274 gene promoter and up-regulate PD-L1 expression. In addition to promoting HCC cell proliferation, SIRT7 reduced acetylation of MEF2D and expression of PD-L1 in HCC cells not exposed to IFNG. Strategies to manipulate this pathway might increase the efficacy of immune therapies for HCC.


Assuntos
Antígeno B7-H1/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Sirtuínas/genética , Adolescente , Adulto , Idoso , Animais , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Feminino , Técnicas de Inativação de Genes , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Imunocompetência , Interferon gama/farmacologia , Neoplasias Hepáticas/patologia , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Transplante de Neoplasias , Receptor de Morte Celular Programada 1/metabolismo , Sirtuínas/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Adulto Jovem
5.
Cancer Lett ; 454: 78-89, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-30980868

RESUMO

Patients with advanced hepatocellular carcinoma (HCC) will almost always develop acquired tolerance after sorafenib therapy, and the molecular mechanism of sorafenib tolerance remains poorly characterized. Here, using our established sorafenib-resistant HCC cell and xenograft models, we identified a novel gene, KIAA1199, which was markedly elevated among the differentially expressed genes involved in sorafenib tolerance. Moreover, elevated expression of KIAA1199 was positively correlated with a high risk of recurrence and metastasis and advanced TNM stage in HCC patients. Functionally, loss- and gain-of-function studies showed that KIAA1199 promoted the migration, invasion, and metastasis of sorafenib-resistant HCC cells. Mechanistically, KIAA1199 is required for EGF-induced epithelial-mesenchymal transition (EMT) in sorafenib-resistant HCC cells by aiding in EGFR phosphorylation. In summary, our data uncover KIAA1199 as a novel sorafenib-tolerant promoting gene that plays an indispensable role in maintaining sorafenib-resistant HCC cell metastasis.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Fator de Crescimento Epidérmico/metabolismo , Hialuronoglucosaminidase/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Sorafenibe/farmacologia , Animais , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Feminino , Células Hep G2 , Xenoenxertos , Humanos , Hialuronoglucosaminidase/biossíntese , Hialuronoglucosaminidase/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica , Fosforilação
6.
Hepatology ; 70(4): 1197-1213, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30901096

RESUMO

Metabolic reprogramming endows cancer cells with the ability to adjust metabolic pathways to support heterogeneously biological processes. However, it is not known how the reprogrammed activities are implemented during differentiation of cancer stem cells (CSCs). In this study, we demonstrated that liver CSCs relied on the enhanced mitochondrial function to maintain stemness properties, which is different from aerobic glycolysis playing main roles in the differentiated non-CSCs. We found that liver CSCs exhibit increased mitochondrial respiratory capacity and that complex-I of mitochondria was necessary for stemness properties of liver CSCs through regulation of mitochondrial respiration. Bioinformatics analysis reveals that mitochondrial ribosomal protein S5 (MRPS5) is closely related with the function of complex-I. Further experiments confirmed that MRPS5 promoted the production of nicotinamide adenine dinucleotide (NAD+ ), which is necessary for enhanced mitochondrial function in liver CSCs. MRPS5 played a critical role for liver CSCs to maintain stemness properties and to participate in tumor progression. Mechanistically, the acetylation status of MRPS5 is directly regulated by NAD+ dependent deacetylase sirtuin-1 (SIRT1), which is abundant in liver CSCs and decreased during differentiation. Deacetylated MRPS5 locates in mitochondria to promote the function complex-I and the generation of NAD+ to enhance mitochondrial respiration. Conversely, the acetylated MRPS5 gathered in nuclei leads to increased expression of glycolytic proteins and promotion of the Warburg Effect. Therefore, liver CSCs transform mitochondrial-dependent energy supply to a Warburg phenotype by the dual function of MRPS5. Clinical analysis of SIRT1 and MRPS5 expression in tumor tissues showed the SIRT1High /Cytoplasmic-MRPS5High profile was associated with patients with hepatocellular carcinoma with poor prognosis. Conclusion: SIRT1/MRPS5 axis participates in metabolic reprogramming to facilitate tumor progression and may serve as a promising therapeutic target of liver cancer.


Assuntos
Carcinoma Hepatocelular/genética , Reprogramação Celular/genética , Neoplasias Hepáticas/genética , Proteínas Mitocondriais/genética , NAD/metabolismo , Proteínas Ribossômicas/genética , Sirtuína 1/metabolismo , Acetilação/efeitos dos fármacos , Animais , Carcinoma Hepatocelular/patologia , Diferenciação Celular/genética , Metilação de DNA/genética , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Sensibilidade e Especificidade , Células Tumorais Cultivadas
7.
Cancer Lett ; 375(2): 390-399, 2016 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27000993

RESUMO

Within the cancer stem cell (CSC) niche, non-CSCs play an indispensable role in facilitating a microenvironment capable of maintaining CSC properties. Non-CSCs contribute to not only the structure and topology of the tumor microenvironment but also the maintenance of the dynamic state of CSCs. Interleukin-17E (IL-17E/IL-25) is important in allergic inflammation and protection against parasitic infection. Moreover, it has also been demonstrated that IL-17E takes part in different cancers recently. Here, for the first time we demonstrate that discrepant expression of IL-17E and the IL-17 receptor B (IL-17RB) exists in Nanog positive (Nanog(Pos)) CSCs and Nanog negative (Nanog(Neg)) non-CSCs in hepatocellular carcinoma (HCC). Moreover, we further demonstrate that IL-17E binding to IL-17RB activates NF-κB and JAK/Stat3 pathways to promote proliferation and sustain self-renewal of CSCs in HCC. Meanwhile, the beneficial effect of IL-17E on Nanog(Pos) CSCs could be blocked by specific inhibitors of JAK and NF-κB signaling. All the findings indicated that non-CSC-derived secreted IL-17E binds IL-17RB on CSCs to signal via JAK/Stat3 and NF-κB pathways to mediate crosstalk between CSCs and non-CSCs. Therefore, IL-17E/IL-17RB signaling represents a potential therapeutic target for treatment of HCC.


Assuntos
Carcinoma Hepatocelular/genética , Interleucina-17/genética , Neoplasias Hepáticas/genética , Fator de Transcrição STAT3/biossíntese , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Janus Quinases/biossíntese , Janus Quinases/genética , Neoplasias Hepáticas/patologia , NF-kappa B/biossíntese , NF-kappa B/genética , Proteína Homeobox Nanog , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Fator de Transcrição STAT3/genética , Microambiente Tumoral/genética
8.
Mol Biosyst ; 10(6): 1524-37, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24691568

RESUMO

The constitutively active fusion protein BCR-ABL1 is the major cause of chronic myeloid leukemia (CML), and selective inhibition of ABL1 is a promising approach for the treatment of CML. Reported drugs worked well in clinical practice, such as imatinib, dasatinib, nilotinib and bosutinib. However, resistance arises due to ABL1 mutation in patients, especially the T315I gate-keeper mutation. Thus, wide spectrum drugs targeting ABL1 are urgently needed. In order to screen potential drugs targeting wild-type ABL1 and T315I mutant ABL1, 1408 FDA approved small molecule drugs were subjected to molecular docking. With subsequent molecular dynamic (MD) simulation and MM/GBSA binding free energy calculation and energy decomposition, we identified chlorhexidine and sorafenib as potential "new use" drugs targeting wild-type ABL1, while nicergoline and plerixafor targeted T315I ABL1. Meanwhile, we also found that residues located in the ATP-binding site and A-loop motif played key roles in drug discovery towards ABL1. These findings may not only serve as a paradigm for the repositioning of existing approved drugs, but also instill new vitality to ABL1-targeted anti-CML therapeutics.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas/métodos , Proteínas de Fusão bcr-abl/química , Proteínas de Fusão bcr-abl/genética , Inibidores de Proteínas Quinases/farmacologia , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Bases de Dados de Produtos Farmacêuticos , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Estados Unidos , United States Food and Drug Administration
9.
Comput Biol Chem ; 47: 56-65, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23891721

RESUMO

Non-specific lipid transfer proteins (ns-LTPs), ubiquitously found in various types of plants, have been well-known to transfer amphiphilic lipids and promote the lipid exchange between mitochondria and microbody. In this study, an in silico analysis was proposed to study ns-LTP in Peganum harmala L., which may belong to ns-LTP1 family, aiming at constructing its three-dimensional structure. Moreover, we adopted MEGA to analyze ns-LTPs and other species phylogenetically, which brought out an initial sequence alignment of ns-LTPs. In addition, we used molecular docking and molecular dynamics simulations to further investigate the affinities and stabilities of ns-LTP with several ligands complexes. Taken together, our results about ns-LTPs and their ligand-binding activities can provide a better understanding of the lipid-protein interactions, indicating some future applications of ns-LTP-mediated transport.


Assuntos
Antígenos de Plantas/química , Antígenos de Plantas/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Peganum/química , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Filogenia
10.
Acta Pharmacol Sin ; 34(5): 612-24, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23564085

RESUMO

Autophagy, an evolutionarily conserved catabolic process involving the engulfment and degradation of non-essential or abnormal cellular organelles and proteins, is crucial for homeostatic maintenance in living cells. This highly regulated, multi-step process has been implicated in diverse diseases including cancer. Autophagy can function as either a promoter or a suppressor of cancer, which makes it a promising and challenging therapeutic target. Herein, we overview the regulatory mechanisms and dual roles of autophagy in cancer. We also describe some of the representative agents that exert their anticancer effects by regulating autophagy. Additionally, some emerging strategies aimed at modulating autophagy are discussed as having the potential for future anticancer drug discovery. In summary, these findings will provide valuable information to better utilize autophagy in the future development of anticancer therapeutics that meet clinical requirements.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Descoberta de Drogas/métodos , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Humanos , MicroRNAs/genética , Neoplasias/genética , Neoplasias/metabolismo
11.
Appl Biochem Biotechnol ; 167(3): 621-31, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22581077

RESUMO

MicroRNAs (miRNAs), highly conserved, non-coding endogenous RNA and nearly ~22 nucleotides (nt) in length, are well-known to regulate several apoptotic pathways in cancer. In this study, we computationally constructed the initial human apoptotic PPI network by several online databases, and further integrated these high-throughput datasets into a Naïve Bayesian model to predict protein functional connections. Based on the modified apoptotic network, we identified several apoptotic hub proteins such as TP53, SRC, M3K3/5/8, cyclin-dependent kinase2/6, TNFR16/19, and TGF-ß receptor 1/2. Subsequently, we identified some microRNAs that could target the aforementioned apoptotic hub proteins by using TargetScan, PicTar, and Diana-MicroH. In conclusion, these results demonstrate the PPI network-based identification of new connections amongst apoptotic pathways in cancer, which may shed new light on the intricate relationships between core apoptotic pathways and some targeted miRNAs in human cancers.


Assuntos
Apoptose , Biologia Computacional/métodos , Neoplasias/metabolismo , Neoplasias/patologia , Mapas de Interação de Proteínas , Transdução de Sinais , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Humanos , MicroRNAs/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
J Mol Model ; 18(1): 27-37, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21445708

RESUMO

The Galanthus nivalis agglutinin (GNA)-related lectin family exhibit significant anti-HIV and anti-HSV properties that are closely related to their carbohydrate-binding activities. However, there is still no conclusive evidence that GNA-related lectins possess anti-influenza properties. The hemagglutinin (HA) of influenza virus is a surface protein that is involved in binding host cell sialic acid during the early stages of infection. Herein, we studied the 3D-QSARs (three-dimensional quantitative structure-activity relationships) of lectin- and HA-sialic acid by molecular modeling. The affinities and stabilities of lectin- and HA-sialic acid complexes were also assessed by molecular docking and molecular dynamics simulations. Finally, anti-influenza GNA-related lectins that possess stable conformations and higher binding affinities for sialic acid than HAs of human influenza virus were screened, and a possible mechanism was proposed. Accordingly, our results indicate that some GNA-related lectins, such as Yucca filamentosa lectin and Polygonatum cyrtonema lectin, could act as drugs that prevent influenza virus infection via competitive binding. In conclusion, the GNA-related lectin family may be helpful in the design of novel candidate agents for preventing influenza A infection through the use of competitive combination against sialic acid specific viral infection.


Assuntos
Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Lectinas de Ligação a Manose/química , Modelos Moleculares , Simulação de Dinâmica Molecular , Lectinas de Plantas/química , Sequência de Aminoácidos , Antivirais/química , Ligação de Hidrogênio , Modelos Químicos , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Relação Quantitativa Estrutura-Atividade , Alinhamento de Sequência , Análise de Sequência de Proteína
13.
Biochem Biophys Res Commun ; 414(2): 282-6, 2011 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-21951850

RESUMO

Concanavalin A (ConA), a Ca(2+)/Mn(2+)-dependent and mannose/glucose-binding legume lectin, has drawn a rising attention for its remarkable anti-proliferative and anti-tumor activities to a variety of cancer cells. ConA induces programmed cell death via mitochondria-mediated, P73-Foxo1a-Bim apoptosis and BNIP3-mediated mitochondrial autophagy. Through IKK-NF-κB-COX-2, SHP-2-MEK-1-ERK, and SHP-2-Ras-ERK anti-angiogenic pathways, ConA would inhibit cancer cell survival. In addition, ConA stimulates cell immunity and generates an immune memory, resisting to the same genotypic tumor. These biological findings shed light on new perspectives of ConA as a potential anti-neoplastic agent targeting apoptosis, autophagy and anti-angiogenesis in pre-clinical or clinical trials for cancer therapeutics.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Concanavalina A/farmacologia , Neoplasias/tratamento farmacológico , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Concanavalina A/química , Concanavalina A/uso terapêutico , Humanos , Sistema de Sinalização das MAP Quinases , Dados de Sequência Molecular , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas ras/metabolismo
14.
Phytomedicine ; 18(8-9): 748-55, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21146383

RESUMO

Polygonatum odoratum lectin (POL), a novel mannose-binding lectin with anti-viral and apoptosis-inducing activities, was isolated from rhizomes of Polygonatum odoratum (Mill.) Druce. POL was a homo-tetramer with molecular weight of 11953.623Da per subunits as determined by gel filtration, SDS-PAGE and mass spectrometry. Based on its N-terminal 29-amino acid sequence the full-length cDNA sequence of POL was cloned. Subsequent phylogenetic analysis and molecular modeling revealed that POL belonged to the Galanthus nivalis agglutinin (GNA)-related lectin family, which acquired unique mannose-binding specificity. The hemagglutinating activities of POL were metal ion-independent, and were stable within certain range of pH and temperature alterations. Moreover, POL showed remarkable anti-HSV-II activity towards Vero cells, cytotoxicity towards human melanoma A375 cells and induced apoptosis in a caspase-dependent manner.


Assuntos
Herpesvirus Humano 2/efeitos dos fármacos , Lectinas de Ligação a Manose/química , Melanoma/tratamento farmacológico , Polygonatum/química , Sequência de Aminoácidos , Antivirais/química , Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Sequência de Bases , Clonagem Molecular , Citotoxinas/química , Citotoxinas/farmacologia , Humanos , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/farmacologia , Dados de Sequência Molecular , Fitoterapia , Extratos Vegetais/farmacologia , Lectinas de Plantas/química , Lectinas de Plantas/genética , Lectinas de Plantas/farmacologia , Rizoma/química
15.
Biochimie ; 92(12): 1934-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20713122

RESUMO

Polygonatum cyrtonema lectin (PCL), a mannose/sialic acid-binding lectin, has been reported to display remarkable anti-proliferative and apoptosis-inducing activities toward a variety of cancer cells; however, the precise molecular mechanisms by which PCL induces cancer cell death are still elusive. In the current study, we found that PCL could induce apoptosis and autophagy in murine fibrosarcoma L929 cells. Subsequently, we demonstrated that inhibition of Ras could promote L929 cell death, suggesting that Ras-Raf signaling pathway plays the key negative regulator in PCL-induced apoptosis. And, we showed that Ras-Raf signaling pathway was also involved in PCL-induced autophagy as the negative regulator. In addition, we found that class I phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway could play the negative regulator in PCL-induced apoptosis and autophagy. Taken together, these results demonstrate that PCL induces murine fibrosarcoma L929 cell apoptosis and autophagy via blocking Ras-Raf and PI3K-Akt signaling pathways.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Lectinas de Plantas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Fibrossarcoma/ultraestrutura , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Indóis/farmacologia , Camundongos , Microscopia Eletrônica de Transmissão , Fenóis/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Polienos/farmacologia , Polygonatum/metabolismo , Alcamidas Poli-Insaturadas/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tetrazóis/farmacologia , Fatores de Tempo , Wortmanina , Quinases raf/antagonistas & inibidores , Quinases raf/metabolismo , Proteínas ras/antagonistas & inibidores , Proteínas ras/metabolismo
16.
Curr Mol Pharmacol ; 3(3): 123-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20533895

RESUMO

Plant lectins, carbohydrate-binding proteins distributed widely in a variety of plant species, have been well-known to possess a broad range of significant biological functions such as anti-tumor, anti-fungal and anti-viral activities. Amongst the seven major lectin families, legume lectins have been drawing a rising attention for cancer biologists due to their remarkable anti-tumor properties compared to other lectin families. In this review, we mainly focus on analyzing the anti-tumor activities of Concanavalin A (ConA), the first and most typical representative of legume lectin family, and its related mechanisms of cell death implicated in apoptosis and autophagy. We present the up-to-date experimental advancements that ConA is able to induce cancer cell apoptosis through mitochondria-dependent and p73-mediated pathways, as well as ConA can induce cancer cell autophagy through a mitochondria-dependent signaling pathway. In addition, we further discuss the pre-clinical studies of ConA for its potential cancer therapeutic applications. In conclusion, these findings may shed light on the complicated molecular mechanisms of ConA-induced cancer cell death, thereby opening a new perspective for plant lectins as potential anti-neoplastic drugs in future cancer therapeutics.


Assuntos
Antineoplásicos/uso terapêutico , Concanavalina A/uso terapêutico , Antineoplásicos/química , Apoptose , Autofagia , Concanavalina A/química , Proteínas de Ligação a DNA/metabolismo , Humanos , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Proteínas Nucleares/metabolismo , Lectinas de Plantas/uso terapêutico , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA