Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Adv Exp Med Biol ; 1164: 11-34, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31576537

RESUMO

Of the ~129,079 new cases of nasopharyngeal carcinoma (NPC) and 72,987 associated deaths estimated for 2018, the majority will be geographically localized to South East Asia, and likely to show an upward trend annually. It is thought that disparities in dietary habits, lifestyle, and exposures to harmful environmental factors are likely the root cause of NPC incidence rates to differ geographically. Genetic differences due to ethnicity and the Epstein Barr virus (EBV) are likely contributing factors. Pertinently, NPC is associated with poor prognosis which is largely attributed to lack of awareness of the salient symptoms of NPC. These include nose hemorrhage and headaches and coupled with detection and the limited therapeutic options. Treatment options include radiotherapy or chemotherapy or combination of both. Surgical excision is generally the last option considered for advanced and metastatic disease, given the close proximity of nasopharynx to brain stem cell area, major blood vessels, and nerves. To improve outcome of NPC patients, novel cellular and in vivo systems are needed to allow an understanding of the underling molecular events causal for NPC pathogenesis and for identifying novel therapeutic targets and effective therapies. While challenges and gaps in current NPC research are noted, some advances in targeted therapies and immunotherapies targeting EBV NPCs are discussed in this chapter, which may offer improvements in outcome of NPC patients.


Assuntos
Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Sudeste Asiático/epidemiologia , Infecções por Vírus Epstein-Barr/complicações , Humanos , Carcinoma Nasofaríngeo/complicações , Carcinoma Nasofaríngeo/epidemiologia , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/complicações , Neoplasias Nasofaríngeas/epidemiologia , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/terapia , Fatores de Risco
2.
N Engl J Med ; 370(17): 1615-1625, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24716661

RESUMO

Genetic defects in MOGS, the gene encoding mannosyl-oligosaccharide glucosidase (the first enzyme in the processing pathway of N-linked oligosaccharide), cause the rare congenital disorder of glycosylation type IIb (CDG-IIb), also known as MOGS-CDG. MOGS is expressed in the endoplasmic reticulum and is involved in the trimming of N-glycans. We evaluated two siblings with CDG-IIb who presented with multiple neurologic complications and a paradoxical immunologic phenotype characterized by severe hypogammaglobulinemia but limited clinical evidence of an infectious diathesis. A shortened immunoglobulin half-life was determined to be the mechanism underlying the hypogammaglobulinemia. Impaired viral replication and cellular entry may explain a decreased susceptibility to infections.


Assuntos
Agamaglobulinemia/genética , Defeitos Congênitos da Glicosilação/imunologia , Resistência à Doença/genética , Viroses/imunologia , alfa-Glucosidases/genética , Agamaglobulinemia/imunologia , Anticorpos Antivirais/sangue , Criança , Defeitos Congênitos da Glicosilação/genética , Defeitos Congênitos da Glicosilação/metabolismo , Feminino , Glicosilação , Humanos , Imunoglobulinas/metabolismo , Masculino
3.
Invest New Drugs ; 35(2): 166-179, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28058624

RESUMO

Zebrafish represents a powerful in vivo model for phenotype-based drug discovery to identify clinically relevant small molecules. By utilizing this model, we evaluated natural product derived compounds that could potentially modulate Notch signaling that is important in both zebrafish embryogenesis and pathogenic in human cancers. A total of 234 compounds were screened using zebrafish embryos and 3 were identified to be conferring phenotypic alterations similar to embryos treated with known Notch inhibitors. Subsequent secondary screens using HEK293T cells overexpressing truncated Notch1 (HEK293TΔE) identified 2 compounds, EDD3 and 3H4MB, to be potential Notch antagonists. Both compounds reduced protein expression of NOTCH1, Notch intracellular domain (NICD) and hairy and enhancer of split-1 (HES1) in HEK293TΔE and downregulated Notch target genes. Importantly, EDD3 treatment of human oral cancer cell lines demonstrated reduction of Notch target proteins and genes. EDD3 also inhibited proliferation and induced G0/G1 cell cycle arrest of ORL-150 cells through inducing p27KIP1. Our data demonstrates the utility of the zebrafish phenotypic screen and identifying EDD3 as a promising Notch antagonist for further development as a novel therapeutic agent.


Assuntos
Antineoplásicos/farmacologia , Curcumina/análogos & derivados , Curcumina/farmacologia , Receptores Notch/antagonistas & inibidores , Triterpenos/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Embrião não Mamífero/anormalidades , Embrião não Mamífero/efeitos dos fármacos , Células HEK293 , Humanos , Fenótipo , Receptores Notch/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/metabolismo
4.
J Cell Sci ; 127(Pt 24): 5261-72, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25344255

RESUMO

The basal layer of the epidermis contains stem cells and transit amplifying cells that rapidly proliferate and differentiate further into the upper layers of the epidermis. A number of molecules have been identified as regulators of this process, including p63 (also known as tumor protein 63) and Notch1. However, little is known about the mechanisms that regulate the transitions from stem cell to proliferating or differentiating transit amplifying cell. Here, we demonstrate that epiprofin (Epfn, also known as Sp6) plays crucial distinct roles in these transition stages as a cell cycle regulator and a transcription factor. Epfn knockout mice have a thickened epidermis, in which p63-expressing basal cells form multiple layers owing to the accumulation of premature transit amplifying cells with reduced proliferation and a reduction in the number of differentiating keratinocytes expressing Notch1. We found that low levels of Epfn expression increased the proliferation of human immortalized keratinocyte (HaCaT) cells by increasing EGF responsiveness and superphosphorylation of Rb. By contrast, high levels of Epfn expression promoted cell cycle exit and differentiation, by reducing E2F transactivation and inducing Notch1 expression. Our findings identify multiple novel functions of Epfn in epidermal development.


Assuntos
Diferenciação Celular , Epiderme/patologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Sp/metabolismo , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Epiderme/crescimento & desenvolvimento , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/patologia , Humanos , Queratinócitos/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/deficiência , Camundongos , Modelos Biológicos , Morfogênese/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Receptores Notch/metabolismo , Proteína do Retinoblastoma/metabolismo
5.
Microcirculation ; 23(6): 389-405, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27177346

RESUMO

Cancer metastasis which predominantly occurs through blood and lymphatic vessels, is the leading cause of death in cancer patients. Consequently, several anti-angiogenic agents have been approved as therapeutic agents for human cancers such as metastatic renal cell carcinoma. Also, anti-lymphangiogenic drugs such as monoclonal antibodies VGX-100 and IMC-3C5 have undergone phase I clinical trials for advanced and metastatic solid tumors. Although anti-tumor-associated angiogenesis has proven to be a promising therapeutic strategy for human cancers, this approach is fraught with toxicities and development of drug resistance. This emphasizes the need for alternative anti-(lymph)angiogenic drugs. The use of zebrafish has become accepted as an established model for high-throughput screening, vascular biology, and cancer research. Importantly, various zebrafish transgenic lines have now been generated that can readily discriminate different vascular compartments. This now enables detailed in vivo studies that are relevant to both human physiological and tumor (lymph)angiogenesis to be conducted in zebrafish. This review highlights recent advancements in the zebrafish anti-vascular screening platform and showcases promising new anti-(lymph)angiogenic compounds that have been derived from this model. In addition, this review discusses the promises and challenges of the zebrafish model in the context of anti-(lymph)angiogenic compound discovery for cancer treatment.


Assuntos
Inibidores da Angiogênese/química , Modelos Animais de Doenças , Peixe-Zebra , Animais , Descoberta de Drogas/métodos , Humanos , Linfangiogênese/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico
6.
FASEB J ; 29(3): 1056-68, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25466898

RESUMO

Multiple human malignancies rely on C-X-C motif chemokine receptor type 4 (CXCR4) and its ligand, SDF-1/CXCL12 (stroma cell-derived factor 1/C-X-C motif chemokine 12), to metastasize. CXCR4 inhibitors promote the mobilization of bone marrow stem cells, limiting their clinical application for metastasis prevention. We investigated the CXCR4-initiated signaling circuitry to identify new potential therapeutic targets. We used HeLa human cancer cells expressing high levels of CXCR4 endogenously. We found that CXCL12 promotes their migration in Boyden chamber assays and single cell tracking. CXCL12 activated mTOR (mechanistic target of rapamycin) potently in a pertussis-sensitive fashion. Inhibition of mTOR complex 1 (mTORC1) by rapamycin [drug concentration causing 50% inhibition (IC50) = 5 nM] and mTORC1/mTORC2 by Torin2 (IC50 = 6 nM), or by knocking down key mTORC1/2 components, Raptor and Rictor, respectively, decreased directional cell migration toward CXCL12. We developed a CXCR4-mediated spontaneous metastasis model by implanting HeLa cells in the tongue of SCID-NOD mice, in which 80% of the animals develop lymph node metastasis. It is surprising that mTORC1 disruption by Raptor knockdown was sufficient to reduce tumor growth by 60% and spontaneous metastasis by 72%, which were nearly abolished by rapamycin. In contrast, disrupting mTORC2 had no effect in tumor growth or metastasis compared with control short hairpin RNAs. These data suggest that mTORC1 may represent a suitable therapeutic target in human malignancies using CXCR4 for their metastatic spread. .


Assuntos
Movimento Celular , Quimiocina CXCL12/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Receptores CXCR4/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Neoplasias do Colo do Útero/secundário , Animais , Apoptose , Western Blotting , Proliferação de Células , Feminino , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transdução de Sinais , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/metabolismo
7.
Cancer Sci ; 106(10): 1333-40, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26250467

RESUMO

Human papillomavirus (HPV) oncoproteins drive distinctive promoter methylation patterns in cancer. However, the underlying mechanism remains to be elucidated. Cyclin A1 (CCNA1) promoter methylation is strongly associated with HPV-associated cancer. CCNA1 methylation is found in HPV-associated cervical cancers, as well as in head and neck squamous cell cancer. Numerous pieces of evidence suggest that E7 may drive CCNA1 methylation. First, the CCNA1 promoter is methylated in HPV-positive epithelial lesions after transformation. Second, the CCNA1 promoter is methylated at a high level when HPV is integrated into the human genome. Finally, E7 has been shown to interact with DNA methyltransferase 1 (Dnmt1). Here, we sought to determine the mechanism by which E7 increases methylation in cervical cancer by using CCNA1 as a gene model. We investigated whether E7 induces CCNA1 promoter methylation, resulting in the loss of expression. Using both E7 knockdown and overexpression approaches in SiHa and C33a cells, our data showed that CCNA1 promoter methylation decreases with a corresponding increase in expression in E7 siRNA-transfected cells. By contrast, CCNA1 promoter methylation was augmented with a corresponding reduction in expression in E7-overexpressing cells. To confirm whether the binding of the E7-Dnmt1 complex to the CCNA1 promoter induced methylation and loss of expression, ChIP assays were carried out in E7-, del CR3-E7 and vector control-overexpressing C33a cells. The data showed that E7 induced CCNA1 methylation by forming a complex with Dnmt1 at the CCNA1 promoter, resulting in the subsequent reduction of expression in cancers. It is interesting to further explore the genome-wide mechanism of E7 oncoprotein-mediated DNA methylation.


Assuntos
Ciclina A1/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Papillomavirus Humano 16/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Regiões Promotoras Genéticas/genética , Linhagem Celular Tumoral , Ciclina A1/biossíntese , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA Metiltransferase 3A , Feminino , Papillomavirus Humano 16/genética , Humanos , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus/genética , Interferência de RNA , RNA Interferente Pequeno , Neoplasias do Colo do Útero/genética , DNA Metiltransferase 3B
8.
Bioconjug Chem ; 26(3): 396-404, 2015 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-25642999

RESUMO

A simple and effective method for synthesizing highly fluorescent, protein-based nanoparticles (Prodots) and their facile uptake into the cytoplasm of cells is described here. Prodots made from bovine serum albumin (nBSA), glucose oxidase (nGO), horseradish peroxidase (nHRP), catalase (nCatalase), and lipase (nLipase) were found to be 15-50 nm wide and have been characterized by gel electrophoresis, transmission electron microscopy (TEM), circular dichroism (CD), fluorescence spectroscopy, dynamic light scattering (DLS), and optical microscopic methods. Data showed that the secondary structure of the protein in Prodots is retained to a significant extent and specific activities of nGO, nHRP, nCatalase, and nLipase were 80%, 70%, 65%, and 50% of their respective unmodified enzyme activities. Calorimetric studies indicated that the denaturation temperatures of nGO and nBSA increased while those of other Prodots remained nearly unchanged, and accelerated storage half-lives of Prodots at 60 °C increased by 4- to 8-fold. Exposure of nGO and nBSA+ nGO to cells indicated rapid uptake within 1-3 h, accompanied by significant blebbing of the plasma membrane, but no uptake has been noted in the absence of nGO. The presence of nGO/glucose in the media facilitated the uptake, and hydrogen peroxide induced membrane permeability could be responsible for this rapid uptake of Prodots. In control studies, FITC alone did not enter the cell, BSA-FITC was not internalized even in the presence of nGO, and there has been no uptake of nBSA-FITC in the absence of nGO. These are the very first examples of very rapid cellular uptake of fluorescent nanoparticles into cells, particularly nanoparticles made from pure proteins. The current approach is a simple and efficient method for the preparation of bioactive, fluorescent protein nanoparticles of controllable size for cellular imaging, and cell uptake is under the control of two separate chemical triggers.


Assuntos
Membrana Celular , Fluoresceína-5-Isotiocianato/química , Corantes Fluorescentes/química , Nanopartículas/química , Soroalbumina Bovina/química , Animais , Bovinos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Corantes Fluorescentes/metabolismo , Humanos , Nanopartículas/metabolismo , Tamanho da Partícula , Soroalbumina Bovina/metabolismo , Espectrometria de Fluorescência/métodos
9.
Immunology ; 142(3): 396-405, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24673602

RESUMO

We have previously reported a novel method for the production of tumour-antigen-presenting cells (referred to as TAPCells) that are currently being used in cancer therapy, using an allogeneic melanoma-derived cell lysate (referred to as TRIMEL) as an antigen provider and activation factor. It was recently demonstrated that TAPCell-based immunotherapy induces T-cell-mediated immune responses resulting in improved long-term survival of stage IV melanoma patients. Clinically, dendritic cell (DC) migration from injected sites to lymph nodes is an important requirement for an effective anti-tumour immunization. This mobilization of DCs is mainly driven by the C-C chemokine receptor type 7 (CCR7), which is up-regulated on mature DCs. Using flow cytometry and immunohistochemistry, we investigated if TRIMEL was capable of inducing the expression of the CCR7 on TAPCells and enhancing their migration in vitro, as well as their in vivo relocation to lymph nodes in an ectopic xenograft animal model. Our results confirmed that TRIMEL induces a phenotypic maturation and increases the expression of surface CCR7 on melanoma patient-derived DCs, and also on the monocytic/macrophage cell line THP-1. Moreover, in vitro assays showed that TRIMEL-stimulated DCs and THP-1 cells were capable of migrating specifically in the presence of the CCR7 ligand CCL19. Finally, we demonstrated that TAPCells could migrate in vivo from the injection site into the draining lymph nodes. This work contributes to an increased understanding of the biology of DCs produced ex vivo allowing the design of new strategies for effective DC-based vaccines for treating aggressive melanomas.


Assuntos
Extratos Celulares/farmacologia , Movimento Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Linfonodos/imunologia , Melanoma , Receptores CCR7/genética , Animais , Linhagem Celular Tumoral , Células Dendríticas/citologia , Humanos , Linfonodos/citologia , Linfonodos/efeitos dos fármacos , Masculino , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores CCR7/imunologia , Receptores CCR7/metabolismo
10.
Mol Ther ; 21(3): 680-7, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23295951

RESUMO

Recombinant adeno-associated viruses (AAV) have been used for therapeutic gene transfer. These vectors offer a number of advantages including resistance to the effects of pH, a broad cellular tropism, efficient gene transfer, persistence of gene expression, and little toxicity. AAV vectors; however, at high doses can induce humoral and cellular immune responses. While potentially problematic for replacement gene therapy, this effect may be advantageous for antitumor vaccination. We examined the activity of an oral and intramuscular antitumor vaccination using AAV serotypes 5 and 6 expressing a truncated neu oncogene in a neu-positive murine TUBO breast cancer model. Mice receiving a single oral administration of AAV5-neu or AAV6-neu demonstrated improved survival. Oral vaccination significantly improved survivals compared with intramuscular vaccination. Mice vaccinated with AAV6-neu survived longer than those treated with AAV5-neu. Vaccination with AAV5-neu or AAV6-neu induced both humoral and cellular immune responses against the NEU antigen. These responses were more robust in the mice undergoing oral vaccination compared with mice receiving the intramuscular vaccination. Protection from tumor was long lasting with 80% of the animals treated with oral AAV6-neu surviving a re-challenge with TUBO cells at 120 and 320 days post-vaccination. Further evaluation of AAV-based vectors as tumor vaccines is warranted.


Assuntos
Neoplasias da Mama/terapia , Vacinas Anticâncer/uso terapêutico , Dependovirus/genética , Vetores Genéticos , Receptor ErbB-2/uso terapêutico , Administração Oral , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Receptor ErbB-2/genética , Transdução Genética
11.
Pharmaceuticals (Basel) ; 17(1)2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38256941

RESUMO

Tumour-associated angiogenesis play key roles in tumour growth and cancer metastasis. Consequently, several anti-angiogenic drugs such as sunitinib and axitinib have been approved for use as anti-cancer therapies. However, the majority of these drugs target the vascular endothelial growth factor A (VEGFA)/VEGF receptor 2 (VEGFR2) pathway and have shown mixed outcome, largely due to development of resistances and increased tumour aggressiveness. In this study, we used the zebrafish model to screen for novel anti-angiogenic molecules from a library of compounds derived from natural products. From this, we identified canthin-6-one, an indole alkaloid, which inhibited zebrafish intersegmental vessel (ISV) and sub-intestinal vessel development. Further characterisation revealed that treatment of canthin-6-one reduced ISV endothelial cell number and inhibited proliferation of human umbilical vein endothelial cells (HUVECs), suggesting that canthin-6-one inhibits endothelial cell proliferation. Of note, canthin-6-one did not inhibit VEGFA-induced phosphorylation of VEGFR2 in HUVECs and downstream phosphorylation of extracellular signal-regulated kinase (Erk) in leading ISV endothelial cells in zebrafish, suggesting that canthin-6-one inhibits angiogenesis independent of the VEGFA/VEGFR2 pathway. Importantly, we found that canthin-6-one impairs tumour-associated angiogenesis in a zebrafish B16F10 melanoma cell xenograft model and synergises with VEGFR inhibitor sunitinib malate to inhibit developmental angiogenesis. In summary, we showed that canthin-6-one exhibits anti-angiogenic properties in both developmental and pathological contexts in zebrafish, independent of the VEGFA/VEGFR2 pathway and demonstrate that canthin-6-one may hold value for further development as a novel anti-angiogenic drug.

12.
Dev Cell ; 14(1): 25-36, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18194650

RESUMO

Vascular endothelial growth factor (VEGF) and Angiopoietin 1 (Ang1) are both potent proangiogenic factors, but, whereas VEGF causes vascular permeability, Ang1 stabilizes blood vessels and protects them from VEGF-induced plasma leakage. The antivascular permeability mechanisms deployed by Ang1 are still undefined. Here, we demonstrate that Ang1 halts the ability of VEGF to induce the phosphorylation-dependent redistribution of the adhesion molecule VE-cadherin, thereby rescuing the endothelial barrier function. Ang1 inhibits the activation of Src by VEGF, the most upstream component of the pathway linking VEGF receptors to VE-cadherin internalization. Indeed, Ang1 promotes the activation of mDia through RhoA, resulting in the association of mDia with Src. This ultimately deprives VEGF receptors of an essential molecule required for promoting the disruption of endothelial cell-cell contacts and paracellular permeability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Angiopoietina-1/farmacologia , Proteínas de Transporte/fisiologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Antígenos CD/fisiologia , Caderinas/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Endocitose/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Transferência Ressonante de Energia de Fluorescência , Forminas , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-38357015

RESUMO

Background: Increasing use prevalence of waterpipe tobacco smoking raises concerns about environmental impacts from waterpipe waste disposal. The U.S. Food and Drug Administration (FDA) is required to assess the environmental impact of its tobacco regulatory actions per the National Environmental Policy Act. This study builds on FDA's efforts characterizing the aquatic toxicity of waterpipe wastewater chemicals. Methods: We compiled a comprehensive list of waterpipe wastewater chemical concentrations from literature. We then selected chemicals for risk assessment by estimating persistence, bioaccumulation, and aquatic toxicity characteristics (PBT; U.S. Environmental Protection Agency), and hazardous concentration values (concentration affecting specific proportion of species). Results: Of 38 chemicals in waterpipe wastewater with concentration data, 20 are listed as harmful or potentially harmful constituents (HPHCs) in tobacco smoke and tobacco products by FDA, and 15 are hazardous waste per U. S. Environmental Protection Agency. Among metals, six (cadmium, chromium, lead, mercury, nickel and selenium) are included in both HPHC and hazardous waste lists and were selected for future risk assessments. Among non-metals, nicotine, and 4-methylnitrosamino-1-(3-pyridyl)-1-butanone (NNK) were shortlisted, as they are classified as persistent and toxic. Further, N-nitrosonornicotine (NNN), with a low HC50 value for chronic aquatic toxicity, had high aquatic toxicity concern and is selected. Conclusions: The presence of multiple hazardous compounds in waterpipe wastewater highlights the importance of awareness on the proper disposal of waterpipe wastewater in residential and retail settings. Future studies can build on the hazard characterization provided in this study through fate and transport modeling, exposure characterization and risk assessments of waterpipe wastewater chemicals.

14.
Anal Chem ; 84(14): 6249-55, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22697359

RESUMO

Multiplexed biomarker protein detection holds unrealized promise for clinical cancer diagnostics due to lack of suitable measurement devices and lack of rigorously validated protein panels. Here we report an ultrasensitive electrochemical microfluidic array optimized to measure a four-protein panel of biomarker proteins, and we validate the protein panel for accurate oral cancer diagnostics. Unprecedented ultralow detection into the 5-50 fg·mL(-1) range was achieved for simultaneous measurement of proteins interleukin 6 (IL-6), IL-8, vascular endothelial growth factor (VEGF), and VEGF-C in diluted serum. The immunoarray achieves high sensitivity in 50 min assays by using off-line protein capture by magnetic beads carrying 400,000 enzyme labels and ~100,000 antibodies. After capture of the proteins and washing to inhibit nonspecific binding, the beads are magnetically separated and injected into the array for selective capture by antibodies on eight nanostructured sensors. Good correlations with enzyme-linked immunosorbent assays (ELISA) for protein determinations in conditioned cancer cell media confirmed the accuracy of this approach. Normalized means of the four protein levels in 78 oral cancer patient serum samples and 49 controls gave clinical sensitivity of 89% and specificity of 98% for oral cancer detection, demonstrating high diagnostic utility. The low-cost, easily fabricated immunoarray provides a rapid serum test for diagnosis and personalized therapy of oral cancer. The device is readily adaptable to clinical diagnostics of other cancers.


Assuntos
Biomarcadores Tumorais/sangue , Análise Química do Sangue/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Nanotecnologia/instrumentação , Animais , Estudos de Casos e Controles , Bovinos , Hipóxia Celular , Humanos , Imunoensaio , Neoplasias Bucais/sangue , Neoplasias Bucais/patologia , Proteínas de Neoplasias/sangue
16.
Exp Mol Pathol ; 93(2): 237-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22580187

RESUMO

The expression of heme oxygenase-1 (HO-1) was shown to be increased in multiple tumors compared with their surrounding healthy tissues and was also observed to be up-regulated in oral squamous cell carcinomas (OSCC). However, conflicting results were obtained and little information is available regarding HO-1 significance in head and neck squamous cell carcinoma (HNSCC). Therefore, the aim of the present study was to perform a wide screening of HO-1 expression in a large collection of human primary HNSCCs and to correlate the results with clinical and pathological parameters. For this purpose, we investigated the expression of this protein by immunohistochemistry (IHC) in tissue microarrays (TMAs) of HNSCC and in an independent cohort of paraffin-embedded tumor specimens. HO-1 expression was further validated by real-time qPCR performed on selected laser capture-microdissected (LCM) oral tissue samples. Both the number of HO-1-positive samples and HO-1 immunoreactivity in the cancerous tissues were significantly higher than those in the non-tumor tissues. These results were confirmed at the mRNA level. Interestingly, HO-1 localization was observed in the nucleus, and the rate of nuclear HO-1 in HNSCC was higher than that in non-malignant tissues. Nuclear HO-1 was observed in HNSCC cell lines and increased even further following hemin treatment. Analysis of HO-1 expression and sub-cellular localization in a mouse model of squamous cell carcinoma (SCC) and in human HNSCC revealed that nuclear HO-1 increases with tumor progression. Taken together, these results demonstrate that HO-1 is up-regulated in HNSCC and that nuclear localization of HO-1 is associated with malignant progression in this tumor type.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Neoplasias de Cabeça e Pescoço/enzimologia , Heme Oxigenase-1/metabolismo , Idoso , Animais , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Modelos Animais de Doenças , Progressão da Doença , Feminino , Neoplasias de Cabeça e Pescoço/diagnóstico , Neoplasias de Cabeça e Pescoço/genética , Heme Oxigenase-1/genética , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Inclusão em Parafina , Reação em Cadeia da Polimerase , Prognóstico , RNA Mensageiro/metabolismo , Análise Serial de Tecidos
17.
Biochem Biophys Res Commun ; 406(2): 171-6, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21291860

RESUMO

Ligand engagement by integrins induces receptor clustering and formation of complexes at the integrin cytoplasmic face that controls cell signaling and cytoskeletal dynamics critical for adhesion-dependent processes. This study searches for a subset of integrin effectors that coordinates both tumor cell invasion and resistance to the chemotherapeutic drug cisplatin in oral carcinomas. Candidate integrin effectors were identified in a proteomics screen of proteins recruited to clustered integrin αß1, α(v)ß or α(6)ß receptors in oral carcinomas. Proteins with diverse functions including microtubule and actin binding proteins, and factors involved in trafficking, transcription and translation were identified in oral carcinoma integrin complexes. Knockdown of effectors in the oral carcinoma HN12 cells revealed that p130Cas, Dek, Src and talin were required for invasion through Matrigel. Disruption of talin or p130Cas by RNA interference increased resistance to cisplatin, whereas targeting Dek, Src or zyxin reduced HN12 resistance to cisplatin. Analysis of the spreading of HN12 cells on collagen I and laminin I revealed that a decrease in p130Cas or talin expression inhibited spreading on both matrices. Interestingly, a reduction in zyxin expression enhanced spreading on laminin I and inhibited spreading on collagen I. Reduction of Dek, Src, talin or zyxin expression reduced HN12 proliferation by 30%. Proliferation was not affected by a reduction in p130Cas expression. We conclude that p130Cas, Src and talin function in both oral carcinoma invasion and resistance to cisplatin.


Assuntos
Carcinoma/patologia , Proteína Substrato Associada a Crk/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias Bucais/patologia , Talina/metabolismo , Quinases da Família src/metabolismo , Carcinoma/tratamento farmacológico , Carcinoma/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/farmacologia , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Proteína Substrato Associada a Crk/genética , Combinação de Medicamentos , Humanos , Integrina alfa1beta1/metabolismo , Integrina alfa5beta1/metabolismo , Integrina alfa6beta1/metabolismo , Laminina/metabolismo , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/metabolismo , Invasividade Neoplásica , Proteoglicanas/metabolismo , Proteômica , RNA Interferente Pequeno/genética , Talina/genética , Quinases da Família src/genética
18.
Breast Cancer Res Treat ; 129(1): 49-67, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20890655

RESUMO

Over the past several years, we have been interested in understanding the mechanisms by which mammary carcinomas acquire hormone independence. We demonstrated that carcinoma associated fibroblasts participate in the ligand-independent activation of progesterone receptors inducing tumor growth. In this study, we used DNA microarrays to compare the gene expression profiles of tumors from the MPA mouse breast cancer model, one hormone-dependent (C4-HD) and one hormone-independent (C4-HI), using whole tumor samples or laser-captured purified stromal and epithelial cells obtained from the same tumors. The expression of selected genes was validated by immunohistochemistry and immunofluorescence assays. We identified 413 genes specifically expressed in tumor stroma. Eighty-five percent of these genes were upregulated, whereas the remaining 15% were downregulated in C4-HI relative to their expression in the C4-HD tumor stroma. Several matrix metallopeptidases were overexpressed in the C4-HI tumor microenvironment. On the other hand, 1100 genes were specifically expressed in the tumor parenchyma. Among them, the 29% were upregulated, whereas the remaining 71% were downregulated in C4-HI relative to C4-HD tumor epithelium. Steap, Pdgfc, Runx2, Cxcl9, and Sdf2 were among the genes with high expression in the C4-HI tumor parenchyma. Interestingly, Fgf2 was one of the few genes upregulated by MPA in C4-HD tumors, confirming its pivotal role in regulating tumor growth in this model. In conclusion, we demonstrate herein a gene expression profile that distinguishes both the epithelial and the stromal cells in mammary tumors with different hormone dependence, supporting the hypothesis that the tumor-associated stroma may contribute to hormone-independent tumor growth.


Assuntos
Antineoplásicos Hormonais/farmacologia , Carcinoma/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Mamárias Experimentais/genética , Acetato de Medroxiprogesterona/farmacologia , Animais , Carcinoma/metabolismo , Carcinoma/patologia , Análise por Conglomerados , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reprodutibilidade dos Testes , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
19.
Adv Exp Med Biol ; 720: 27-38, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21901616

RESUMO

The human oral squamous epithelium plays an important role in maintaining a barrier function against mechanical, physical, and pathological injury. However, the self-renewing cells residing on the basement membrane of the epithelium can give rise to oral squamous cell carcinomas (OSCC), now the sixth most common cancer in the developed world, which is still associated with poor prognosis. This is due, in part, to the limited availability of well-defined culture systems for studying oral epithelial cell biology, which could advance our understanding of the molecular basis of OSCC. Here, we describe methods to successfully isolate large cultures of human oral epithelial cells and fibroblasts from small pieces of donor tissues for use in techniques such as three-dimensional cultures and animal grafts to validate genes suspected of playing a role in OSCC development and progression. Finally, the use of isolated oral epithelial cells in generating iPS cells is discussed which holds promise in the field of oral regenerative medicine.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/patologia , Separação Celular , Células Cultivadas , Células Epiteliais/fisiologia , Humanos , Queratinócitos/citologia , Mucosa Bucal/citologia , Células-Tronco/citologia
20.
J AOAC Int ; 104(2): 485-497, 2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-33259591

RESUMO

BACKGROUND: Phyllanthus species exhibit a wide range of in vitro and in vivo pharmacological activities; however, little is known about the compounds present in the extracts that are responsible for such actions. OBJECTIVE: Development and validation of a simple reversed phase HPLC-PDA method for profiling of phyllanthin, hypophyllanthin, nirtetralin, and niranthin in extracts of Phyllanthus species was carried out. METHODS: Separation was achieved using an XBridge column® (150 × 4.6 mm, 5.0 µm id) in an isocratic elution mode with mobile phase comprising of a mixture of acetonitrile and water with TFA (0.05%, v/v, pH = 2.15) at ambient temperature with a flow rate of 1 mL/min. RESULTS: Phyllanthin, hypophyllanthin, nirtetralin, and niranthin were eluted at mean retention times of 10.47, 11.10, 13.67, and 14.53 min, respectively. LOD and LOQ for all four analytes were 0.75 and 3.00 µg/mL, respectively. RSDr values for intraday and interday precision for phyllanthin, hypophyllanthin, nirtetralin, and niranthin were 0.38-1.32 and 0.45-1.77%; 0.22-3.69 and 0.24-3.04%, 0.73-2.37 and 0.09-0.31%, and 1.56-2.77 and 0.12-0.68%, respectively. CONCLUSIONS: The developed and validated HPLC-PDA method was applied for identification and quantification of phyllanthin, hypophyllanthin, nirtetralin, and niranthin in extracts of different plant parts of selected Phyllanthus species. The outcome of the present investigation could be useful for selection of best species to promote its commercial cultivation and suitable extraction solvent for preparation of lignan-enriched fractions. This HPLC-PDA method could be useful for quality control of herbal formulations containing plants from Phyllanthus species.


Assuntos
Lignanas , Phyllanthus , Anisóis , Cromatografia Líquida de Alta Pressão , Dioxóis , Índia , Extratos Vegetais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa