Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Res ; 248: 118304, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38295979

RESUMO

The complexity of micro-tidal inlets arises from the combined action of littoral drift and tidal range on their stability. Consequently, understanding and evaluating their stability poses a significant challenge. This study aims to shed some insight on the assessment of inlet stability by employing Delft 3D model. The stability of the inlet between the ocean and estuary relies on the balance between the longshore transport rate and the spring tidal prism. Disrupting this equilibrium results in the closure of the inlets. The movement of sediments in the surf zone is primarily driven by longshore velocity, which acts as the driving force for littoral drift, which is estimated using Delft 3D wave model. The longshore transport rate is estimated by employing empirical relationships and numerical codes based on the obtained driving force. Subsequently, the stability of the inlet is assessed based on these estimations. The spring tidal prism refers to the discharge of water flowing into the ocean from inlets and estuaries. Flow velocity is determined using Delft 3D flow model. The input data for nearshore circulation resulting from waves and currents is primarily collected through field measurements and data collected from Indian National Centre for Ocean Information Services (INCOIS). For the current study, Muttukadu (12°47'13″N, 80°15'01″E) inlet, Kovalam along the East Coast of the Indian Peninsula is investigated by assessing its seasonal variations. This study contributes to the management of marine biological ecology, the expansion of small-scale artisanal fishing, the promotion of water sports-related tourism, the advancement of fishing harbor development, and the execution of coastal engineering projects.


Assuntos
Baías , Estuários , Água , Estações do Ano , Monitoramento Ambiental/métodos
2.
Environ Res ; 249: 118246, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38278509

RESUMO

The Earth's history is documented in human civilizations, soil layers, river movement, and quiet sediments throughout millennia. This investigation explores the significant legacy of environmental toxins in these key planet components. Understanding how ancient activity shaped the terrain is crucial as mankind faces environmental issues. This interdisciplinary study uses environmental science, archaeology, and geology to uncover Earth's mysteries. It illuminates the dynamic processes that have built our globe by studying pollutants and soil, water, and sediments. This research follows human actions, both intentional and unintentional, from ancient civilizations through contemporary industrialization and their far-reaching effects. Environmental destiny examines how contaminants affect ecosystems and human health. This study of past contamination helps solve modern problems including pollution cleanup, sustainable land management, and water conservation. This review studies reminds us that our previous activities still affect the ecosystem in a society facing rapid urbanisation and industrialization. It emphasises the importance of environmental stewardship and provides a framework for making educated choices to reduce toxins in soil, water, and sediments. Discovery of Earth's secrets is not only a historical curiosity; it's a necessary step towards a sustainable and peaceful cohabitation with our home planet.


Assuntos
Sedimentos Geológicos , Sedimentos Geológicos/química , Sedimentos Geológicos/análise , Poluentes do Solo/análise , Humanos , Planeta Terra , Solo/química , Monitoramento Ambiental/história , Monitoramento Ambiental/métodos , Poluentes Químicos da Água/análise , Poluição Ambiental/história , Poluição Ambiental/análise
3.
Biomaterials ; 180: 24-35, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30014964

RESUMO

Fibrous proteins found in the natural extracellular matrix (ECM) function as host substrates for migration and growth of endogenous cells during wound healing and tissue repair processes. Although various fibrous scaffolds have been developed to recapitulate the microstructures of the native ECM, facile synthesis of hydrogel microfibers that are mechanically robust and biologically active have been elusive. Described herein is the use of interfacial bioorthogonal polymerization to create hydrogel-based microfibrous scaffolds via tetrazine ligation. Combination of a trifunctional strained trans-cyclooctene monomer and a difunctional s-tetrazine monomer at the oil-water interface led to the formation of microfibers that were stable under cell culture conditions. The bioorthogonal nature of the synthesis allows for direct incorporation of tetrazine-conjugated peptides or proteins with site-selectively, genetically encoded tetrazines. The microfibers provide physical guidance and biochemical signals to promote the attachment, division and migration of fibroblasts. Mechanistic investigations revealed that fiber-guided cell migration was both F-actin and microtubule-dependent, confirming contact guidance by the microfibers. Prolonged culture of fibroblasts in the presence of an isolated microfiber resulted in the formation of a multilayered cell sheet wrapping around the fiber core. A fibrous mesh provided a 3D template to promote cell infiltration and tissue-like growth. Overall, the bioorthogonal approach led to the straightforward synthesis of crosslinked hydrogel microfibers that can potentially be used as instructive materials for tissue repair and regeneration.


Assuntos
Hidrogéis/química , Animais , Técnicas de Cultura de Células , Movimento Celular/fisiologia , Fibroblastos/citologia , Humanos , Peptídeos/química , Polimerização , Proteínas/química , Alicerces Teciduais/química , Cicatrização/fisiologia
4.
ACS Appl Mater Interfaces ; 10(31): 26016-26027, 2018 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-30015482

RESUMO

Chemical modification of engineered microenvironments surrounding living cells represents a means for directing cellular behaviors through cell-matrix interactions. Presented here is a temporally controlled method for modulating the properties of biomimetic, synthetic extracellular matrices (ECM) during live cell culture employing the rapid, bioorthogonal tetrazine ligation with trans-cyclooctene (TCO) dienophiles. This approach is diffusion-controlled, cytocompatible, and does not rely on light, catalysts, or other external triggers. Human bone-marrow-derived mesenchymal stem cells (hMSCs) were initially entrapped in a hydrogel prepared using hyaluronic acid carrying sulfhydryl groups (HA-SH) and a hydrophilic polymer bearing both acrylate and tetrazine groups (POM-AT). Inclusion of a matrix metalloprotease (MMP)-degradable peptidic cross-linker enabled hMSC-mediated remodeling of the synthetic environment. The resultant network displayed dangling tetrazine groups for subsequent conjugation with TCO derivatives. Two days later, the stiffness of the matrix was increased by adding chemically modified HA carrying multiple copies of TCO (HA-TCO) to the hMSC growth media surrounding the cell-laden gel construct. In response, cells developed small processes radially around the cell body without a significant alteration of the overall shape. By contrast, modification of the 3D matrix with a TCO-tagged cell-adhesive motif caused the resident cells to undergo significant actin polymerization, changing from a rounded shape to spindle morphology with long cellular processes. After additional 7 days of culture in the growth media, quantitative analysis showed that, at the mRNA level, RGD tagging upregulated cellular expression of MMP1, but downregulated the expression of collagen I/III and tenascin C. RGD tagging, however, was not sufficient to induce the classic osteoblastic, chondrogenic, adipogenic, or fibroblastic/myofibroblastic differentiation. The modular approach allows facile manipulation of synthetic ECM to modulate cell behavior, thus potentially applicable to the engineering of functional tissues or tissue models.


Assuntos
Células-Tronco , Técnicas de Cultura de Células , Diferenciação Celular , Condrogênese , Matriz Extracelular , Humanos , Hidrogéis , Células-Tronco Mesenquimais
5.
Biomacromolecules ; 17(11): 3750-3760, 2016 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-27723964

RESUMO

Toward the goal of establishing physiologically relevant in vitro tumor models, we synthesized and characterized a biomimetic hydrogel using thiolated hyaluronic acid (HA-SH) and an acrylated copolymer carrying multiple copies of cell adhesive peptide (PolyRGD-AC). PolyRGD-AC was derived from a random copolymer of tert-butyl methacrylate (tBMA) and oligomeric (ethylene glycol) methacrylate (OEGMA), synthesized via atom transfer radical polymerization (ATRP). Acid hydrolysis of tert-butyl moieties revealed the carboxylates, through which acrylate groups were installed. Partial modification of the acrylate groups with a cysteine-containing RGD peptide generated PolyRGD-AC. When PolyRGD-AC was mixed with HA-SH under physiological conditions, a macroscopic hydrogel with an average elastic modulus of 630 Pa was produced. LNCaP prostate cancer cells encapsulated in HA-PolyRGD gels as dispersed single cells formed multicellular tumoroids by day 4 and reached an average diameter of ∼95 µm by day 28. Cells in these structures were viable, formed cell-cell contacts through E-cadherin (E-CAD), and displayed cortical organization of F-actin. Compared with the control gels prepared using PolyRDG, multivalent presentation of the RGD signal in the HA matrix increased cellular metabolism, promoted the development of larger tumoroids, and enhanced the expression of E-CAD and integrins. Overall, hydrogels with multivalently immobilized RGD are a promising 3D culture platform for dissecting principles of tumorigenesis and for screening anticancer drugs.


Assuntos
Carcinogênese/efeitos dos fármacos , Hidrogéis/química , Peptídeos/química , Polímeros/química , Biomimética , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Hidrogéis/síntese química , Hidrogéis/farmacologia , Masculino , Metacrilatos/síntese química , Metacrilatos/química , Metacrilatos/farmacologia , Peptídeos/síntese química , Peptídeos/farmacologia , Polímeros/síntese química , Polímeros/farmacologia , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/patologia
6.
ACS Appl Mater Interfaces ; 8(28): 17915-26, 2016 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-27322677

RESUMO

Epithelial-to-mesenchymal transition (EMT) is a well-studied biological process that takes place during embryogenesis, carcinogenesis, and tissue fibrosis. During EMT, the polarized epithelial cells with a cuboidal architecture adopt an elongated fibroblast-like morphology. This process is accompanied by the expression of many EMT-specific molecular markers. Although the molecular mechanism leading to EMT has been well-established, the effects of matrix topography and microstructure have not been clearly elucidated. Synthetic scaffolds mimicking the meshlike structure of the basement membrane with an average fiber diameter of 0.5 and 5 µm were produced via the electrospinning of poly(ε-caprolactone) (PCL) and were used to test the significance of fiber diameter on EMT. Cell-adhesive peptide motifs were conjugated to the fiber surface to facilitate cell attachment. Madin-Darby Canine Kidney (MDCK) cells grown on these substrates showed distinct phenotypes. On 0.5 µm substrates, cells grew as compact colonies with an epithelial phenotype. On 5 µm scaffolds, cells were more individually dispersed and appeared more fibroblastic. Upon the addition of hepatocyte growth factor (HGF), an EMT inducer, cells grown on the 0.5 µm scaffold underwent pronounced scattering, as evidenced by the alteration of cell morphology, localization of focal adhesion complex, weakening of cell-cell adhesion, and up-regulation of mesenchymal markers. In contrast, HGF did not induce a pronounced scattering of MDCK cells cultured on the 5.0 µm scaffold. Collectively, our results show that the alteration of the fiber diameter of proteins found in the basement membrane may create enough disturbances in epithelial organization and scattering that might have important implications in disease progression.


Assuntos
Materiais Biomiméticos/química , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/fisiologia , Alicerces Teciduais , Animais , Adesão Celular/fisiologia , Moléculas de Adesão Celular/fisiologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Microambiente Celular/fisiologia , Cães , Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Células Madin Darby de Rim Canino , Poliésteres/química
7.
J Cell Sci ; 128(23): 4366-79, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26483386

RESUMO

In epithelial cancers, carcinoma cells coexist with normal cells. Although it is known that the tumor microenvironment (TME) plays a pivotal role in cancer progression, it is not completely understood how the tumor influences adjacent normal epithelial cells. In this study, a three-dimensional co-culture system comprising non-transformed epithelial cells (MDCK) and transformed carcinoma cells (MSV-MDCK) was used to demonstrate that carcinoma cells sequentially induce preneoplastic lumen filling and epithelial-mesenchymal transition (EMT) in epithelial cysts. MMP-9 secreted by carcinoma cells cleaves cellular E-cadherin (encoded by CDH1) from epithelial cells to generate soluble E-cadherin (sE-cad), a pro-oncogenic protein. We show that sE-cad induces EGFR activation, resulting in lumen filling in MDCK cysts. Long-term sE-cad treatment induced EMT. sE-cad caused lumen filling by induction of the ERK signaling pathway and triggered EMT through the sustained activation of the AKT pathway. Although it is known that sE-cad induces MMP-9 release and consequent EGFR activation in tumor cells, our results, for the first time, demonstrate that carcinoma cells can induce sE-cad shedding in adjacent epithelial cells, which leads to EGFR activation and the eventual transdifferentiation of the normal epithelial cells.


Assuntos
Caderinas/metabolismo , Carcinoma/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Animais , Caderinas/genética , Carcinoma/genética , Carcinoma/patologia , Cães , Células Epiteliais/patologia , Receptores ErbB/genética , Células Madin Darby de Rim Canino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Mol Pharm ; 12(6): 2101-11, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25898125

RESUMO

Nanomedicine has advanced to clinical trials for adult cancer therapy. However, the field is still in its infancy for treatment of childhood malignancies such as acute lymphoblastic leukemia (ALL). Nanotherapy offers multiple advantages over conventional therapy. It facilitates targeted delivery and enables controlled release of drugs to reduce treatment-related side effects. Here, we demonstrate that doxorubicin (DOX) encapsulated in polymeric nanoparticles (NPs) modified with targeting ligands against CD19 (CD19-DOX-NPs) can be delivered in a CD19-specific manner to leukemic cells. The CD19-DOX-NPs were internalized via receptor-mediated endocytosis and imparted cytotoxicity in a CD19-dependent manner in CD19-positive ALL cells. Leukemic mice treated with CD19-DOX-NPs survived significantly longer and manifested a higher degree of agility, indicating reduced apparent systemic toxicity during treatment compared to mice treated with free DOX. We suggest that targeted delivery of drugs used in childhood cancer treatment should improve therapeutic efficacy and reduce treatment-related side effects in children.


Assuntos
Antígenos CD19/metabolismo , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C
9.
PLoS One ; 10(4): e0122442, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25836370

RESUMO

Glucocorticoids are commonly used as palliative or chemotherapeutic clinical agents for treatment of a variety of cancers. Although steroid treatment is beneficial, the mechanisms by which steroids improve outcome in cancer patients are not well understood. Na,K-ATPase beta-subunit isoform 1 (NaK-ß1) is a cell-cell adhesion molecule, and its expression is down-regulated in cancer cells undergoing epithelial-to mesenchymal-transition (EMT), a key event associated with cancer progression to metastatic disease. In this study, we performed high-throughput screening to identify small molecules that could up-regulate NaK-ß1 expression in cancer cells. Compounds related to the glucocorticoids were identified as drug candidates enhancing NaK-ß1 expression. Of these compounds, triamcinolone, dexamethasone, and fluorometholone were validated to increase NaK-ß1 expression at the cell surface, enhance cell-cell adhesion, attenuate motility and invasiveness and induce mesenchymal to epithelial like transition of renal cell carcinoma (RCC) cells in vitro. Treatment of NaK-ß1 knockdown cells with these drug candidates confirmed that these compounds mediate their effects through up-regulating NaK-ß1. Furthermore, we demonstrated that these compounds attenuate tumor growth in subcutaneous RCC xenografts and reduce local invasiveness in orthotopically-implanted tumors. Our results strongly indicate that the addition of glucocorticoids in the treatment of RCC may improve outcome for RCC patients by augmenting NaK-ß1 cell-cell adhesion function.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/enzimologia , Glucocorticoides/farmacologia , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Carcinoma de Células Renais/patologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dexametasona/farmacologia , Progressão da Doença , Fluormetolona/farmacologia , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Pelados , Camundongos SCID , Invasividade Neoplásica/prevenção & controle , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , Triancinolona/farmacologia , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Kidney Cancer VHL ; 2(1): 15-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-28326255

RESUMO

Gramicidin A (GA) is a channel-forming ionophore that renders biological membranes permeable to specific cations which disrupts cellular ionic homeostasis. It is a well-known antibiotic, however it's potential as a therapeutic agent for cancer has not been widely evaluated. In two recently published studies, we showed that GA treatment is toxic to cell lines and tumor xenografts derived from renal cell carcinoma (RCC), a devastating disease that is highly resistant to conventional therapy. GA was found to possess the qualities of both a cytotoxic drug and a targeted angiogenesis inhibitor, and this combination significantly compromised RCC growth in vitro and in vivo. In this review, we summarize our recent research on GA, discuss the possible mechanisms whereby it exerts its anti-tumor effects, and share our perspectives on the future opportunities and challenges to the use of GA as a new anticancer agent.

11.
PLoS One ; 9(7): e102041, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25025131

RESUMO

The Wilms' tumor transcription factor (WT1) was originally classified as a tumor suppressor, but it is now known to also be associated with cancer progression and poor prognosis in several malignancies. WT1 plays an essential role in orchestrating a developmental process known as mesenchymal-to-epithelial transition (MET) during kidney development, but also induces the reverse process, epithelial-to-mesenchymal transition (EMT) during heart development. WT1 is not expressed in the adult kidney, but shows elevated expression in clear cell renal cell carcinoma (ccRCC). However, the role of WT1 in this disease has not been characterized. In this study, we demonstrate that WT1 is upregulated in ccRCC cells that are deficient in the expression of the von Hippel-Lindau tumor suppressor protein (VHL). We found that WT1 transcriptionally activated Snail, a master transcriptional repressor that is known to induce EMT. Although Snail represses E-cadherin and induces mesenchymal characteristics, we found partial maintenance of E-cadherin and associated epithelial characteristics in kidney cells and ccRCC cells that express WT1, since WT1 upregulates E-cadherin expression and competes with Snail repression. These findings support a novel paradigm in which WT1 induces an epithelial-mesenchymal hybrid transition (EMHT), characterized by Snail up-regulation with E-cadherin maintenance, a tumor cell differentiation state in which cancer cells keep both EMT and MET characteristics which may promote tumor cell plasticity and tumor progression.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Transição Epitelial-Mesenquimal/genética , Neoplasias Renais/genética , Neoplasias Renais/patologia , Proteínas Wnt/genética , Animais , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Proteínas WT1/genética
12.
Mol Cancer Ther ; 13(4): 788-99, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24493697

RESUMO

Ionophores are hydrophobic organic molecules that disrupt cellular transmembrane potential by permeabilizing membranes to specific ions. Gramicidin A is a channel-forming ionophore that forms a hydrophilic membrane pore that permits the rapid passage of monovalent cations. Previously, we found that gramicidin A induces cellular energy stress and cell death in renal cell carcinoma (RCC) cell lines. RCC is a therapy-resistant cancer that is characterized by constitutive activation of the transcription factor hypoxia-inducible factor (HIF). Here, we demonstrate that gramicidin A inhibits HIF in RCC cells. We found that gramicidin A destabilized HIF-1α and HIF-2α proteins in both normoxic and hypoxic conditions, which in turn diminished HIF transcriptional activity and the expression of various hypoxia-response genes. Mechanistic examination revealed that gramicidin A accelerates O(2)-dependent downregulation of HIF by upregulating the expression of the von Hippel-Lindau (VHL) tumor suppressor protein, which targets hydroxylated HIF for proteasomal degradation. Furthermore, gramicidin A reduced the growth of human RCC xenograft tumors without causing significant toxicity in mice. Gramicidin A-treated tumors also displayed physiologic and molecular features consistent with the inhibition of HIF-dependent angiogenesis. Taken together, these results demonstrate a new role for gramicidin A as a potent inhibitor of HIF that reduces tumor growth and angiogenesis in VHL-expressing RCC.


Assuntos
Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Carcinoma de Células Renais/tratamento farmacológico , Gramicidina/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Neoplasias Renais/tratamento farmacológico , Animais , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Neoplasias Renais/patologia , Camundongos , Neoplasias Experimentais , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Epigenetics ; 9(4): 579-86, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24452105

RESUMO

The Na,K-ATPase or sodium pump carries out the coupled extrusion of Na(+) and uptake of K(+) across the plasma membranes of cells of most higher eukaryotes. We have shown earlier that Na,K-ATPase-ß 1 (NaK-ß) protein levels are highly reduced in poorly differentiated kidney carcinoma cells in culture and in patients' tumor samples. The mechanism(s) regulating the expression of NaK-ß in tumor tissues has yet to be explored. We hypothesized that DNA methylation plays a role in silencing the NaK-ß gene (ATP1B1) expression in kidney cancers. In this study, to the best of our knowledge we provide the first evidence that ATP1B1 is epigenetically silenced by promoter methylation in both renal cell carcinoma (RCC) patients' tissues and cell lines. We also show that knockdown of the von Hippel-Lindau (VHL) tumor suppressor gene in RCC cell lines results in enhanced ATP1B1 promoter AT hypermethylation, which is accompanied by reduced expression of NaK-ß. Furthermore, treatment with 5-Aza-2'-deoxycytidine rescued the expression of ATP1B1 mRNA as well as NaK-ß protein in these cells. These data demonstrate that promoter hypermethylation is associated with reduced NaK-ß expression, which might contribute to RCC initiation and/or disease progression.


Assuntos
Carcinoma de Células Renais/metabolismo , Epigênese Genética , Neoplasias Renais/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Decitabina , Humanos , Neoplasias Renais/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
14.
Mol Cancer Ther ; 12(11): 2296-307, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24006494

RESUMO

Ionophores are lipid-soluble organic molecules that disrupt cellular transmembrane potential by rendering biologic membranes permeable to specific ions. They include mobile-carriers that complex with metal cations and channel-formers that insert into the membrane to form hydrophilic pores. Although mobile-carriers possess anticancer properties, investigations on channel-formers are limited. Here, we used the channel-forming ionophore gramicidin A to study its effects on the growth and survival of renal cell carcinoma (RCC) cells. RCC is a histologically heterogeneous malignancy that is highly resistant to conventional treatments. We found that gramicidin A reduced the in vitro viability of several RCC cell lines at submicromolar concentrations (all IC50 < 1.0 µmol/L). Gramicidin A exhibited similar toxicity in RCC cells regardless of histologic subtype or the expression of either the von Hippel-Lindau tumor suppressor gene or its downstream target, hypoxia-inducible factor-1α. Gramicidin A decreased cell viability equal to or greater than the mobile-carrier monensin depending on the cell line. Mechanistic examination revealed that gramicidin A blocks ATP generation by inhibiting oxidative phosphorylation and glycolysis, leading to cellular energy depletion and nonapoptotic cell death. Finally, gramicidin A effectively reduced the growth of RCC tumor xenografts in vivo. These results show a novel application of gramicidin A as a potential therapeutic agent for RCC therapy.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Renais/metabolismo , Morte Celular/efeitos dos fármacos , Gramicidina/farmacologia , Neoplasias Renais/metabolismo , Animais , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Cães , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glicólise/efeitos dos fármacos , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Nus , Fosforilação Oxidativa/efeitos dos fármacos , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Neurol India ; 61(2): 164-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23644317

RESUMO

Airplane travel headache is rare and has recently been described as a new form of headache associated with a specific situation. Of the 1,208 patients with primary headaches attending a tertiary care neurology hospital, two (0.16%) patients satisfied the criteria for headache related to airplane travel. Both the patients fulfilled the proposed diagnostic criteria for airplane travel headache. This unique headache had a mean duration of 24 minutes, localized to the medial supraorbital region described as having an intense jabbing or stabbing character that occurred exclusively and maximally during aircraft landing or take-off, following which pain intensity subsided . This rare headache felt on aircraft descent is probably due to the squeeze effect on the frontal sinus wall, when air trapped inside it contracts producing a negative pressure leading to mucosal edema, transudation and intense pain. Use of nasal decongestants either alone or in combination with naproxen sodium prior to ascent and descent abated the headache episodes. Awareness about this unique entity is essential to provide proper treatment and avoid patient suffering.


Assuntos
Aeronaves , Cefaleia/etiologia , Viagem , Adulto , Cefaleia/diagnóstico , Humanos , Masculino
16.
Mol Pharm ; 10(6): 2199-210, 2013 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-23194373

RESUMO

Nanotechnology approaches have tremendous potential for enhancing treatment efficacy with lower doses of chemotherapeutics. Nanoparticle (NP)-based drug delivery approaches are poorly developed for childhood leukemia. Dexamethasone (Dex) is one of the most common chemotherapeutic drugs used in the treatment of childhood leukemia. In this study, we encapsulated Dex in polymeric NPs and validated their antileukemic potential in vitro and in vivo. NPs with an average diameter of 110 nm were assembled from an amphiphilic block copolymer of poly(ethylene glycol) (PEG) and poly(ε-caprolactone) (PCL) bearing pendant cyclic ketals (ECT2). The blank NPs were nontoxic to cultured cells in vitro and to mice in vivo. Encapsulation of Dex into the NPs (Dex-NP) did not compromise the bioactivity of the drug. Dex-NPs induced glucocorticoid phosphorylation and showed cytotoxicity similar to the free Dex in leukemic cells. Studies using NPs labeled with fluorescent dyes revealed leukemic cell surface binding and internalization. In vivo biodistribution studies showed NP accumulation in the liver and spleen with subsequent clearance of the particles with time. In a preclinical model of leukemia, Dex-NPs significantly improved the quality of life and survival of mice as compared to the free drug. To our knowledge, this is the first report showing the efficacy of polymeric NPs to deliver Dex to potentially treat childhood leukemia and reveals that low doses of Dex should be sufficient for inducing cell death and improving survival.


Assuntos
Dexametasona/química , Dexametasona/uso terapêutico , Leucemia/tratamento farmacológico , Nanomedicina/métodos , Nanopartículas/química , Polímeros/química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação/efeitos dos fármacos , Poliésteres/química , Polietilenoglicóis/química , Baço/metabolismo
17.
J Cell Sci ; 125(Pt 23): 5711-20, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23077177

RESUMO

Na,K-ATPase is a hetero-oligomer of an α- and a ß-subunit. The α-subunit (Na,K-α) possesses the catalytic function, whereas the ß-subunit (Na,K-ß) has cell-cell adhesion function and is localized to the apical junctional complex in polarized epithelial cells. Earlier, we identified two distinct conserved motifs on the Na,K-ß(1) transmembrane domain that mediate protein-protein interactions: a glycine zipper motif involved in the cis homo-oligomerization of Na,K-ß(1) and a heptad repeat motif that is involved in the hetero-oligomeric interaction with Na,K-α(1). We now provide evidence that knockdown of Na,K-ß(1) prevents lumen formation and induces activation of extracellular regulated kinases 1 and 2 (ERK1/2) mediated by phosphatidylinositol 3-kinase in MDCK cells grown in three-dimensional collagen cultures. These cells sustained cell proliferation in an ERK1/2-dependent manner and did not show contact inhibition at high cell densities, as revealed by parental MDCK cells. This phenotype could be rescued by wild-type Na,K-ß(1) or heptad repeat motif mutant of Na,K-ß(1), but not by the glycine zipper motif mutant that abrogates Na,K-ß(1) cis homo-oligomerization. These studies suggest that Na,K-ß(1) cis homo-oligomerization rather than hetero-oligomerization with Na,K-α(1) is involved in epithelial lumen formation. The relevance of these findings to pre-neoplastic lumen filling in epithelial cancer is discussed.


Assuntos
ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Cães , Immunoblotting , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Multimerização Proteica/genética , Multimerização Proteica/fisiologia , ATPase Trocadora de Sódio-Potássio/química
19.
Cancer Res ; 72(12): 2917-23, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22659456

RESUMO

Strong cell-cell interactions represent a major barrier against cancer cell mobility, and loss of intercellular adhesion by E-cadherin is a fundamental change that occurs during the progression of cancer to invasive disease. However, some aggressive carcinomas retain characteristics of differentiated epithelial cells, including E-cadherin expression. Emerging evidence indicates that proteolysis of E-cadherin generates fragments that promote tumor growth, survival, and motility, suggesting that E-cadherin cleavage converts this tumor suppressor into an oncogenic factor. In this review we discuss the emerging roles of cleaved E-cadherin fragments as modulators of cancer progression, and explore the translational and clinical implications of this research.


Assuntos
Caderinas/metabolismo , Adesão Celular/fisiologia , Neoplasias/metabolismo , Comunicação Celular , Movimento Celular , Células Epiteliais , Genes Supressores de Tumor , Humanos , Neoplasias/patologia , Via de Sinalização Wnt
20.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1150-8, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22345575

RESUMO

Diminished Na,K-ATPase expression has been reported in several carcinomas and has been linked to tumor progression. However, few studies have determined whether Na,K-ATPase function and expression are altered in lung malignancies. Because cigarette smoke (CS) is a major factor underlying lung carcinogenesis and progression, we investigated whether CS affects Na,K-ATPase activity and expression in lung cell lines. Cells exposed to CS in vitro showed a reduction of Na,K-ATPase activity. We detected the presence of reactive oxygen species (ROS) in cells exposed to CS before Na,K-ATPase inhibition, and neutralization of ROS restored Na,K-ATPase activity. We further determined whether Na,K-ATPase expression correlated with increasing grades of lung adenocarcinoma and survival of patients with smoking history. Immunohistochemical analysis of lung adenocarcinoma tissues revealed reduced Na,K-ATPase expression with increasing tumor grade. Using tissue microarray containing lung adenocarcinomas of patients with known smoking status, we found that high expression of Na,K-ATPase correlated with better survival. For the first time, these data demonstrate that CS is associated with loss of Na,K-ATPase function and expression in lung carcinogenesis, which might contribute to disease progression.


Assuntos
Adenocarcinoma/enzimologia , Neoplasias Pulmonares/enzimologia , Nicotiana , Fumaça/efeitos adversos , ATPase Trocadora de Sódio-Potássio/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Linhagem Celular Tumoral , Progressão da Doença , Intervalo Livre de Doença , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Gradação de Tumores , Espécies Reativas de Oxigênio/metabolismo , Fumar/efeitos adversos , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...