Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Res ; 78(14): 4059-4072, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29764866

RESUMEN

Progress in understanding tumor stromal biology has been constrained in part because cancer-associated fibroblasts (CAF) are a heterogeneous population with limited cell-type-specific protein markers. Using RNA expression profiling, we identified the membrane protein leucine-rich repeat containing 15 (LRRC15) as highly expressed in multiple solid tumor indications with limited normal tissue expression. LRRC15 was expressed on stromal fibroblasts in many solid tumors (e.g., breast, head and neck, lung, pancreatic) as well as directly on a subset of cancer cells of mesenchymal origin (e.g., sarcoma, melanoma, glioblastoma). LRRC15 expression was induced by TGFß on activated fibroblasts (αSMA+) and on mesenchymal stem cells. These collective findings suggested LRRC15 as a novel CAF and mesenchymal marker with utility as a therapeutic target for the treatment of cancers with LRRC15-positive stromal desmoplasia or cancers of mesenchymal origin. ABBV-085 is a monomethyl auristatin E (MMAE)-containing antibody-drug conjugate (ADC) directed against LRRC15, and it demonstrated robust preclinical efficacy against LRRC15 stromal-positive/cancer-negative, and LRRC15 cancer-positive models as a monotherapy, or in combination with standard-of-care therapies. ABBV-085's unique mechanism of action relied upon the cell-permeable properties of MMAE to preferentially kill cancer cells over LRRC15-positive CAF while also increasing immune infiltrate (e.g., F4/80+ macrophages) in the tumor microenvironment. In summary, these findings validate LRRC15 as a novel therapeutic target in multiple solid tumor indications and support the ongoing clinical development of the LRRC15-targeted ADC ABBV-085.Significance: These findings identify LRRC15 as a new marker of cancer-associated fibroblasts and cancers of mesenchymal origin and provide preclinical evidence for the efficacy of an antibody-drug conjugate targeting the tumor stroma. Cancer Res; 78(14); 4059-72. ©2018 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Inmunoconjugados/farmacología , Proteínas de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Células del Estroma/efectos de los fármacos , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HCT116 , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias/metabolismo , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Sarcoma/tratamiento farmacológico , Sarcoma/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
Neoplasia ; 19(9): 695-706, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28787674

RESUMEN

Improving the congruity of preclinical models with cancer as it is manifested in humans is a potential way to mitigate the high attrition rate of new cancer therapies in the clinic. In this regard, three-dimensional (3D) tumor cultures in vitro have recently regained interest as they have been acclaimed to have higher similarity to tumors in vivo than to cells grown in monolayers (2D). To identify cancer functions that are active in 3D rather than in 2D cultures, we compared the transcriptional profiles (TPs) of two non-small cell lung carcinoma cell lines, NCI-H1650 and EBC-1 grown in both conditions to the TP of xenografted tumors. Because confluence, diameter or volume can hypothetically alter TPs, we made intra- and inter-culture comparisons using samples with defined dimensions. As projected by Ingenuity Pathway Analysis (IPA), a limited number of signal transduction pathways operational in vivo were better represented by 3D than by 2D cultures in vitro. Growth of 2D and 3D cultures as well as xenografts induced major changes in the TPs of these 3 modes of culturing. Alterations of transcriptional network activation that were predicted to evolve similarly during progression of 3D cultures and xenografts involved the following functions: hypoxia, proliferation, cell cycle progression, angiogenesis, cell adhesion, and interleukin activation. Direct comparison of TPs of 3D cultures and xenografts to monolayer cultures yielded up-regulation of networks involved in hypoxia, TGF and Wnt signaling as well as regulation of epithelial mesenchymal transition. Differences in TP of 2D and 3D cancer cell cultures are subject to progression of the cultures. The emulation of the predicted cell functions in vivo is therefore not only determined by the type of culture in vitro but also by the confluence or diameter of the 2D or 3D cultures, respectively. Consequently, the successful implementation of 3D models will require phenotypic characterization to verify the relevance of applying these models for drug development.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Transcriptoma , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Ratones , Esferoides Celulares
3.
Pharmacol Res Perspect ; 3(5): e00178, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26516589

RESUMEN

The Bcl-2 family inhibitors venetoclax and navitoclax demonstrated potent antitumor activity in chronic lymphocytic leukemia patients, notably in reducing marrow load and adenopathy. Subsequent trials with venetoclax have been initiated in non-Hodgkin's lymphoma and multiple myeloma patients. Traditional preclinical models fall short either in faithfully recapitulating disease progression within such compartments or in allowing the direct longitudinal analysis of systemic disease. We show that intravenous inoculation of engineered RS4;11 (acute lymphoblastic leukemia) and Granta 519 (mantle cell lymphoma) bioluminescent reporter cell lines result in tumor engraftment of bone marrow, with additional invasion of the central nervous system in the case of Granta 519. Importantly, apoptosis induction and response of these systemically engrafted tumors to Bcl-2 family inhibitors alone or in combination with standard-of-care agents could be monitored longitudinally with optical imaging, and was more accurately reflective of the observed clinical response.

4.
Mol Cancer Res ; 7(10): 1686-92, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19825992

RESUMEN

Many established cancer therapies involve DNA-damaging chemotherapy or radiotherapy. Gain of DNA repair capacity of the tumor represents a common mechanism used by cancer cells to survive DNA-damaging therapy. Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme that is activated by DNA damage and plays a critical role in base excision repair. Inhibition of PARP represents an attractive approach for the treatment of cancer. Previously, we have described the discovery and characterization of a potent PARP inhibitor, ABT-888. ABT-888 potentiates the activity of DNA-damaging agents such as temozolomide (TMZ) in a variety of preclinical models. We report here the generation of HCT116 cells resistant to treatment with TMZ and ABT-888 (HCT116R cells). HCT116R cells exhibit decreased H2AX phosphorylation in response to treatment with TMZ and ABT-888 relative to parental HCT116 cells. Microarray and Western blot studies indicate that HCT116R cells have decreased PARP-1 and elevated Rad51 expression levels. HCT116R cells are dependent on Rad51 for proliferation and survival, as shown by inhibition of proliferation and induction of apoptosis upon treatment with Rad51 small interfering RNA. In addition, HCT116R cells are more resistant to radiation than the parental HCT116 cells. Our study suggests that cancer cells upregulate the homologous recombination DNA repair pathway to compensate for the loss of base excision repair, which may account for the observed resistance to treatment with TMZ and ABT-888.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencimidazoles/farmacología , Reparación del ADN/efectos de los fármacos , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/genética , Recombinación Genética/efectos de los fármacos , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Reparación del ADN/genética , Dacarbazina/farmacología , Regulación hacia Abajo/genética , Histonas/efectos de los fármacos , Histonas/genética , Histonas/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño , Recombinasa Rad51/efectos de los fármacos , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Recombinación Genética/genética , Homología de Secuencia , Temozolomida
5.
Anticancer Res ; 28(2A): 957-63, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18507042

RESUMEN

Akt (PKB) is a serine/threonine protein kinase that plays an important role in the transduction of signals affecting apoptosis, cell proliferation and survival. The Akt gene is frequently hyperactivated in tumors and has been shown to be amplified in a number of types of human cancers. Furthermore, Akt activity is elevated in cell lines with the mutated PTEN tumor suppressor gene. These studies establish Akt as an attractive target for cancer therapy. To determine the roles of Akt1, Akt2 and Akt3 in signal transduction, constitutively active Akt1, Akt2 and Akt3 was ectopically overexpressed in human pancreatic MiaPaCa-2 cells. The three Akt stable clones were characterized to determine their effects on transformation and proliferation. Compared to a vector control, the three Akt clones were able to drive cellular proliferation even in reduced serum conditions. Furthermore, in soft-agar assays, the Akt clones showed an 25-38% increase in colony formation in 2% serum. Our results indicate that all three forms of Akt may have protective effects within the cell depending on the type of apoptotic stimuli. Using 2D-PAGE comparisons between parental and Akt overexpressing cells, we attempted to determine novel targets of Akt phosphorylation. In this study, we identified prohibitin as a substrate for Akt both in vitro and in vivo. These studies suggest that Akt may regulate the cellular function of prohibitin via its phosphorylation.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Humanos , Datos de Secuencia Molecular , Neoplasias Pancreáticas , Fosforilación , Prohibitinas , Isoformas de Proteínas/metabolismo , Transducción de Señal , Transfección
6.
Anticancer Agents Med Chem ; 7(6): 681-4, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18045062

RESUMEN

The focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that localizes to the points of cell contact with the extracellular matrix, called focal adhesions. FAK is involved in several cellular processes including invasion, motility, proliferation and apoptosis. In in vivo animal studies, FAK has been shown to contribute to tumor development and malignancy. Furthermore, FAK expression was shown to be elevated in a number of human cancers. Increased FAK expression and activity are correlated with malignant phenotype and poor prognosis in patients. Taken together, these studies suggest that FAK is a potentially good target for drug discovery. In this review, FAK and its relationship to cancer, as well as approaches to therapeutic intervention of FAK will be discussed.


Asunto(s)
Diseño de Fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Neoplasias , Animales , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Pirimidinas/química , Pirroles/química , ARN Interferente Pequeño/fisiología
7.
Clin Cancer Res ; 13(9): 2728-37, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17473206

RESUMEN

PURPOSE: To evaluate the preclinical pharmacokinetics and antitumor efficacy of a novel orally bioavailable poly(ADP-ribose) polymerase (PARP) inhibitor, ABT-888. EXPERIMENTAL DESIGN: In vitro potency was determined in a PARP-1 and PARP-2 enzyme assay. In vivo efficacy was evaluated in syngeneic and xenograft models in combination with temozolomide, platinums, cyclophosphamide, and ionizing radiation. RESULTS: ABT-888 is a potent inhibitor of both PARP-1 and PARP-2 with K(i)s of 5.2 and 2.9 nmol/L, respectively. The compound has good oral bioavailability and crosses the blood-brain barrier. ABT-888 strongly potentiated temozolomide in the B16F10 s.c. murine melanoma model. PARP inhibition dramatically increased the efficacy of temozolomide at ABT-888 doses as low as 3.1 mg/kg/d and a maximal efficacy achieved at 25 mg/kg/d. In the 9L orthotopic rat glioma model, temozolomide alone exhibited minimal efficacy, whereas ABT-888, when combined with temozolomide, significantly slowed tumor progression. In the MX-1 breast xenograft model (BRCA1 deletion and BRCA2 mutation), ABT-888 potentiated cisplatin, carboplatin, and cyclophosphamide, causing regression of established tumors, whereas with comparable doses of cytotoxic agents alone, only modest tumor inhibition was exhibited. Finally, ABT-888 potentiated radiation (2 Gy/d x 10) in an HCT-116 colon carcinoma model. In each model, ABT-888 did not display single-agent activity. CONCLUSIONS: ABT-888 is a potent inhibitor of PARP, has good oral bioavailability, can cross the blood-brain barrier, and potentiates temozolomide, platinums, cyclophosphamide, and radiation in syngeneic and xenograft tumor models. This broad spectrum of chemopotentiation and radiopotentiation makes this compound an attractive candidate for clinical evaluation.


Asunto(s)
Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Administración Oral , Animales , Antineoplásicos Alquilantes/uso terapéutico , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Daño del ADN , Modelos Animales de Enfermedad , Perros , Sinergismo Farmacológico , Femenino , Haplorrinos , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratas , Ratas Endogámicas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Neoplasia ; 7(11): 992-1000, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16331885

RESUMEN

Akt is a serine/threonine kinase that transduces survival signals from survival/growth factors. Deregulation and signal imbalance in cancer cells make them prone to apoptosis. Upregulation or activation of Akt to aid the survival of cancer cells is a common theme in human malignancies. We have developed small-molecule Akt inhibitors that are potent and specific. These Akt inhibitors can inhibit Akt activity and block phosphorylation by Akt on multiple downstream targets in cells. Synergy in apoptosis induction was observed when Akt inhibitors were combined with doxorubicin or camptothecin. Akt inhibitor-induced enhancement of topoisomerase inhibitor cytotoxicity was also evident in long-term cell survival assay. Synergy with paclitaxel in apoptosis induction was evident in cells pretreated with paclitaxel, and enhancement of tumor delay by paclitaxel was demonstrated through cotreatment with Akt inhibitor Compound A (A-443654). Combination with other classes of chemotherapeutic agents did not yield any enhancement of cytotoxicity. These findings provide important guidance in selecting appropriate classes of chemotherapeutic agents for combination with Akt inhibitors in cancer treatment.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/química , Humanos , Indazoles/farmacología , Indoles/farmacología , Cinética , Paclitaxel/farmacología
9.
Mol Cancer Ther ; 4(6): 977-86, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15956255

RESUMEN

The Akt kinases are central nodes in signal transduction pathways that are important for cellular transformation and tumor progression. We report the development of a series of potent and selective indazole-pyridine based Akt inhibitors. These compounds, exemplified by A-443654 (K(i) = 160 pmol/L versus Akt1), inhibit Akt-dependent signal transduction in cells and in vivo in a dose-responsive manner. In vivo, the Akt inhibitors slow the progression of tumors when used as monotherapy or in combination with paclitaxel or rapamycin. Tumor growth inhibition was observed during the dosing interval, and the tumors regrew when compound administration was ceased. The therapeutic window for these compounds is narrow. Efficacy is achieved at doses approximately 2-fold lower than the maximally tolerated doses. Consistent with data from knockout animals, the Akt inhibitors induce an increase in insulin secretion. They also induce a reactive increase in Akt phosphorylation. Other toxicities observed, including malaise and weight loss, are consistent with abnormalities in glucose metabolism. These data show that direct Akt inhibition may be useful in cancer therapy, but significant metabolic toxicities are likely dose limiting.


Asunto(s)
Indazoles/farmacología , Indoles/farmacología , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Indazoles/química , Indazoles/uso terapéutico , Indoles/química , Indoles/uso terapéutico , Ratones , Ratones SCID , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Piridinas/química , Piridinas/farmacología , Sensibilidad y Especificidad , Especificidad por Sustrato
10.
Anticancer Res ; 24(6): 3899-905, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15736429

RESUMEN

The focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that localizes to the points of cell contact with the extracellular matrix, called focal adhesions. Many factors induce tyrosine phosphorylation of FAK including growth factors, neuropeptides and integrin-dependent adhesion to the extracellular matrix. FAK has been implicated in several cellular processes such as invasion, motility, proliferation and apoptosis. In addition, FAK expression was shown to be elevated in a number of different human cancers, suggesting a role in the development of malignancy. We examined the biological functions of FAK using small inhibitory RNAs (siRNA) in cancer cells. Although FAK siRNA reduced the FAK protein levels by approximately 70% in several cancer cell lines, there was no clear evidence of apoptosis. However, in clonogenic and soft-agar assays in H1299, a lung cancer cell line, FAK siRNA treatment led to a 43% to 55% decrease in colony formation. Furthermore, FAK siRNA-treated cells displayed a decrease in migration when serum or EGF (epidermal growth factor) were used as chemo-attractants. Our results demonstrated that inhibition of FAK protein leads to alterations in cell growth and migration.


Asunto(s)
Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Factor de Crecimiento Epidérmico/farmacología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células Madre Neoplásicas , Fosforilación , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transfección
11.
J Biol Chem ; 278(1): 486-90, 2003 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-12419797

RESUMEN

Survivin, one of the most tumor-specific gene products, has been implicated in both anti-apoptosis and cytokinesis. However, the mechanism by which survivin regulates these two different processes is still elusive. Here, we show that survivin binds to the catalytic domain of Aurora-B. We demonstrate that in the presence of survivin, Aurora-B phosphorylates histone H3 much more efficiently than in the absence of survivin in a cell-free system. Furthermore, we confirm that cells lacking survivin due to survivin antisense oligonucleotide-treatment have lower Aurora-B kinase activity, whereas cells overexpressing survivin have higher Aurora-B kinase activity. We also provide evidence that depletion of survivin by survivin antisense oligonucleotide treatment causes significant reduction of endogenous phosphorylated histone H3 and mislocalization of Aurora-B. These results indicate that survivin stimulates Aurora-B kinase activity and helps correctly target Aurora-B to its substrates during the cell cycle, thus providing a mechanism as to how survivin exerts its function in human cells.


Asunto(s)
Inhibidores de Cisteína Proteinasa/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Dominio Catalítico , Ciclo Celular/fisiología , Línea Celular , Regulación hacia Abajo/fisiología , Histonas/metabolismo , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis , Sustancias Macromoleculares , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias , Oligonucleótidos Antisentido/metabolismo , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes de Fusión/metabolismo , Survivin
12.
Microbiology (Reading) ; 143 ( Pt 2): 367-376, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9043114

RESUMEN

The BGL2 gene encodes a unique 1,3-beta-glucosyltransferase (Bgl2p) present in the cell wall of Candida albicans and other fungi. Although believed to be involved in cell wall assembly, disruption of the gene in saccharomyces cerevisiae showed no apparent phenotype. We performed sequential disruptions of the BGL2 loci in a homozygous ura3 clinical isolate of C. albicans using the URA3 blaster method, in order to investigate the role of Bgl2p in this dimorphic, pathogenic fungus. Strain CACW-1 contained disruptions of both homologues of the BGL2 gene and lacked Bgl2p, as assessed by protein extraction, SDS-PAGE and Western blot analysis, and enzyme assay; however, residual non-Bgl2p transferase activity was detected. CACW-1 was attenuated in virulence for mice when compared to an isogenic parent strain, and fewer organisms were recovered from the kidneys of infected animals. Additional phenotypic changes included: (1) a dramatic increase in the sensitivity to the chitin synthesis inhibitor nikkomycin Z when CACW-1 cells were incubated at 37 or 42 degrees C; (2) an 8.7 +/- 1.6% slower growth rate at 37 degrees C for CACW-1 when compared to its isogenic parent; and (3) aggregation of CACW-1 cells during stationary phase and/or incubation of stationary phase cells in phosphate buffer. Characterization of SDS-extracted cell walls did not reveal any significant differences in the levels of 1,3-beta- or 1,6-beta-glucan. These data reveal that loss of Bgl2p does have a phenotype in C. albicans, and indicate that (1) loss of Bgl2p function renders cells more dependent on chitin for wall integrity, and attenuates virulence (probably due to subtle changes in wall structure), and (2) that additional 1,3-beta-glucosyltransferases are present in the C. albicans BGL2 disruptant.


Asunto(s)
Candida albicans/genética , Proteínas Fúngicas/genética , Genes Fúngicos , Glucano Endo-1,3-beta-D-Glucosidasa/genética , Glucosiltransferasas/genética , Animales , Antifúngicos/farmacología , Candida albicans/enzimología , Candida albicans/patogenicidad , Candidiasis , Pared Celular/química , Pared Celular/metabolismo , Pared Celular/ultraestructura , Glucanos/química , Glicosilación , Riñón/microbiología , Ratones , Pruebas de Sensibilidad Microbiana , Mutagénesis , Fenotipo , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA