Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 22(8): 936-946, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37294884

RESUMEN

Identifying novel, unique, and personalized molecular targets for patients with pancreatic ductal adenocarcinoma (PDAC) remains the greatest challenge in altering the biology of fatal tumors. Bromo- and extra-terminal domain (BET) proteins are activated in a noncanonical fashion by TGFß, a ubiquitous cytokine in the PDAC tumor microenvironment (TME). We hypothesized that BET inhibitors (BETi) represent a new class of drugs that attack PDAC tumors via a novel mechanism. Using a combination of patient and syngeneic murine models, we investigated the effects of the BETi drug BMS-986158 on cellular proliferation, organoid growth, cell-cycle progression, and mitochondrial metabolic disruption. These were investigated independently and in combination with standard cytotoxic chemotherapy (gemcitabine + paclitaxel [GemPTX]). BMS-986158 reduced cell viability and proliferation across multiple PDAC cell lines in a dose-dependent manner, even more so in combination with cytotoxic chemotherapy (P < 0.0001). We found that BMS-986158 reduced both human and murine PDAC organoid growth (P < 0.001), with associated perturbations in the cell cycle leading to cell-cycle arrest. BMS-986158 disrupts normal cancer-dependent mitochondrial function, leading to aberrant mitochondrial metabolism and stress via dysfunctional cellular respiration, proton leakage, and ATP production. We demonstrated mechanistic and functional data that BETi induces metabolic mitochondrial dysfunction, abrogating PDAC progression and proliferation, alone and in combination with systemic cytotoxic chemotherapies. This novel approach improves the therapeutic window in patients with PDAC and offers another treatment approach distinct from cytotoxic chemotherapy that targets cancer cell bioenergetics.


Asunto(s)
Antineoplásicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Gemcitabina , Línea Celular Tumoral , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Antineoplásicos/farmacología , Desoxicitidina/uso terapéutico , Proliferación Celular , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Mitocondrias , Microambiente Tumoral
2.
Cell Oncol (Dordr) ; 44(3): 673-687, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33694102

RESUMEN

PURPOSE: The transforming growth factor-beta (TGF-ß) pathway plays a paradoxical, context-dependent role in pancreatic ductal adenocarcinoma (PDAC): a tumor-suppressive role in non-metastatic PDAC and a tumor-promotive role in metastatic PDAC. We hypothesize that non-SMAD-TGF-ß signaling induces PDAC progression. METHODS: We investigated the expression of non-SMAD-TGF-ß signaling proteins (pMAPK14, PD-L1, pAkt and c-Myc) in patient-derived tissues, cell lines and an immunocompetent mouse model. Experimental models were complemented by comparing the signaling proteins in PDAC specimens from patients with various survival intervals. We manipulated models with TGF-ß, gemcitabine (DNA synthesis inhibitor), galunisertib (TGF-ß receptor inhibitor) and MK-2206 (Akt inhibitor) to investigate their effects on NF-κB, ß-catenin, c-Myc and PD-L1 expression. PD-L1 expression was also investigated in cancer cells and tumor associated macrophages (TAMs) in a mouse model. RESULTS: We found that tumors from patients with aggressive PDAC had higher levels of the non-SMAD-TGF-ß signaling proteins pMAPK14, PD-L1, pAkt and c-Myc. In PDAC cells with high baseline ß-catenin expression, TGF-ß increased ß-catenin expression while gemcitabine increased PD-L1 expression. Gemcitabine plus galunisertib decreased c-Myc and NF-κB expression, but induced PD-L1 expression in some cancer models. In mice, gemcitabine plus galunisertib treatment decreased metastases (p = 0.018), whereas galunisertib increased PD-L1 expression (p < 0.0001). In the mice, liver metastases contained more TAMs compared to the primary pancreatic tumors (p = 0.001), and TGF-ß increased TAM PD-L1 expression (p < 0.05). CONCLUSIONS: In PDAC, the non-SMAD-TGF-ß signaling pathway leads to more aggressive phenotypes, TAM-induced immunosuppression and PD-L1 expression. The divergent effects of TGF-ß ligand versus receptor inhibition in tumor cells versus TAMs may explain the TGF-ß paradox. Further evaluation of each mechanism is expected to lead to the development of targeted therapies.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Factor de Crecimiento Transformador beta/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Animales , Progresión de la Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
3.
J Med Chem ; 63(19): 10855-10878, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32886511

RESUMEN

Capuramycin displays a narrow spectrum of antibacterial activity by targeting bacterial translocase I (MraY). In our program of development of new N-acetylglucosaminephosphotransferase1 (DPAGT1) inhibitors, we have identified that a capuramycin phenoxypiperidinylbenzylamide analogue (CPPB) inhibits DPAGT1 enzyme with an IC50 value of 200 nM. Despite a strong DPAGT1 inhibitory activity, CPPB does not show cytotoxicity against normal cells and a series of cancer cell lines. However, CPPB inhibits migrations of several solid cancers including pancreatic cancers that require high DPAGT1 expression in order for tumor progression. DPAGT1 inhibition by CPPB leads to a reduced expression level of Snail but does not reduce E-cadherin expression level at the IC50 (DPAGT1) concentration. CPPB displays a strong synergistic effect with paclitaxel against growth-inhibitory action of a patient-derived pancreatic adenocarcinoma, PD002: paclitaxel (IC50: 1.25 µM) inhibits growth of PD002 at 0.0024-0.16 µM in combination with 0.10-2.0 µM CPPB (IC50: 35 µM).


Asunto(s)
Aminoglicósidos/farmacología , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , Neoplasias/patología , Aminoglicósidos/química , Antineoplásicos/química , Línea Celular Tumoral , Sinergismo Farmacológico , Inhibidores Enzimáticos/química , Humanos , Paclitaxel/farmacología , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Relación Estructura-Actividad
4.
Oncotarget ; 7(18): 26331-45, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27028866

RESUMEN

Melanoma risk is increased in patients with mutations of melanocortin 1 receptor (MC1R) yet the basis for the increased risk remains unknown. Here we report in vivo evidence supporting a critical role for MC1R in regulating melanoma tumor growth and determining overall survival time. Inhibition of MC1R by its physiologically relevant competitive inhibitor, agouti signaling protein (ASIP), reduced melanin synthesis and morphological heterogeneity in murine B16-F10 melanoma cells. In the lungs of syngeneic C57BL/6 mice, mCherry-marked, ASIP-secreting lung tumors inhibited MC1R on neighboring tumors lacking ASIP in a dose dependent manner as evidenced by a proportional loss of pigment in tumors from mice injected with 1:1, 3:1 and 4:1 mixtures of parental B16-F10 to ASIP-expressing tumor cells. ASIP-expressing B16-F10 cells formed poorly pigmented tumors in vivo that correlated with a 20% longer median survival than those bearing parental B16-F10 tumors (p=0.0005). Mice injected with 1:1 mixtures also showed survival benefit (p=0.0054), whereas injection of a 4:1 mixture showed no significant difference in survival. The longer survival time of mice bearing ASIP-expressing tumors correlated with a significantly slower growth rate than parental B16-F10 tumors as judged by quantification of numbers of tumors and total tumor load (p=0.0325), as well as a more homogeneous size and morphology of ASIP-expressing lung tumors. We conclude that MC1R plays an important role in regulating melanoma growth and morphology. Persistent inhibition of MC1R provided a significant survival advantage resulting in part from slower tumor growth, establishing MC1R as a compelling new molecular target for metastatic melanoma.


Asunto(s)
Proteína de Señalización Agouti/metabolismo , Melanoma Experimental/patología , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Proteína de Señalización Agouti/genética , Animales , Línea Celular Tumoral , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
J Innate Immun ; 2(6): 596-606, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20814186

RESUMEN

Group A Streptococcus (GAS) causes rare but life-threatening syndromes of necrotizing fasciitis and toxic shock-like syndrome in humans. The GAS serotype M1T1 clone has globally disseminated, and mutations in the control of virulence regulatory sensor kinase (covRS) operon correlate with severe invasive disease. Here, a cohort of non-M1 GAS was screened to determine whether mutation in covRS triggers systemic dissemination in divergent M serotypes. A GAS disease model defining parameters governing invasive propensity of differing M types is proposed. The vast majority of GAS infection is benign. Nonetheless, many divergent M types possess limited capacity to cause invasive infection. M1T1 GAS readily switch to a covRS mutant form that is neutrophil resistant and frequently associated with systemic infection. Whilst non-M1 GAS are shown in this study to less frequently accumulate covRS mutations in vivo, such mutants are isolated from invasive infections and exhibit neutrophil resistance and enhanced virulence. The reduced capacity of non-M1 GAS to switch to the hypervirulent covRS mutant form provides an explanation for the comparatively less frequent isolation of non-M1 serotypes from invasive human infections.


Asunto(s)
ADN Bacteriano/análisis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neutrófilos/metabolismo , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/fisiología , Animales , Células Cultivadas , Análisis Mutacional de ADN , Progresión de la Enfermedad , Prueba de Complementación Genética , Histidina Quinasa , Humanos , Evasión Inmune/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Mutación/genética , Neutrófilos/inmunología , Neutrófilos/microbiología , Neutrófilos/patología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/fisiopatología , Streptococcus pyogenes/patogenicidad , Virulencia/genética
6.
J Infect Dis ; 201(6): 855-65, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20151844

RESUMEN

Group A streptococci (GAS) may engage different sets of virulence strategies, depending on the site of infection and host context. We previously isolated 2 phenotypic variants of a globally disseminated M1T1 GAS clone: a virulent wild-type (WT) strain, characterized by a SpeB(+)/SpeA(-)/Sda1(low) phenotype, and a hypervirulent animal-passaged (AP) strain, better adapted to survive in vivo, with a SpeB(-)/SpeA(+)/Sda1(high) phenotype. This AP strain arises in vivo due to the selection of bacteria with mutations in covS, the sensor part of a key 2-component regulatory system, CovR/S. To determine whether covS mutations explain the hypervirulence of the AP strain, we deleted covS from WT bacteria (DeltaCovS) and were able to simulate the hypervirulence and gene expression phenotype of naturally selected AP bacteria. Correction of the covS mutation in AP bacteria reverted them back to the WT phenotype. Our data confirm that covS plays a direct role in regulating GAS virulence.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/biosíntesis , Cisteína Endopeptidasas/genética , Desoxirribonucleasa I/biosíntesis , Desoxirribonucleasa I/genética , Exotoxinas/biosíntesis , Exotoxinas/genética , Femenino , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa , Estimación de Kaplan-Meier , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos CBA , Mutación , Fenotipo , Reacción en Cadena de la Polimerasa , Infecciones Estreptocócicas/microbiología , Virulencia
7.
Nat Med ; 13(8): 981-5, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632528

RESUMEN

Most invasive bacterial infections are caused by species that more commonly colonize the human host with minimal symptoms. Although phenotypic or genetic correlates underlying a bacterium's shift to enhanced virulence have been studied, the in vivo selection pressures governing such shifts are poorly understood. The globally disseminated M1T1 clone of group A Streptococcus (GAS) is linked with the rare but life-threatening syndromes of necrotizing fasciitis and toxic shock syndrome. Mutations in the GAS control of virulence regulatory sensor kinase (covRS) operon are associated with severe invasive disease, abolishing expression of a broad-spectrum cysteine protease (SpeB) and allowing the recruitment and activation of host plasminogen on the bacterial surface. Here we describe how bacteriophage-encoded GAS DNase (Sda1), which facilitates the pathogen's escape from neutrophil extracellular traps, serves as a selective force for covRS mutation. The results provide a paradigm whereby natural selection exerted by the innate immune system generates hypervirulent bacterial variants with increased risk of systemic dissemination.


Asunto(s)
Desoxirribonucleasa I/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/patogenicidad , Animales , Supervivencia Celular , Desoxirribonucleasa I/genética , Humanos , Inmunidad Innata , Ratones , Neutrófilos/citología , Neutrófilos/microbiología , Fenotipo , Selección Genética , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/genética , Virulencia
8.
J Pediatr Hematol Oncol ; 28(9): 627-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17006272

RESUMEN

Group A Streptococcus pyogenes causes a distinctive clinical disorder, streptococcal toxic shock syndrome, mediated by superantigenic bacterial exotoxins. Oncology patients with viridans group streptococcal sepsis frequently present with a streptococcal toxic shocklike syndrome of unclear pathogenesis. Viridans group streptococci isolated from pediatric oncology patients with streptococcal toxic shocklike illnesses do not possess homologs of known superantigen genes. Supernatants from cultures of these bacteria also fail to stimulate T-cell proliferation, suggesting these bacteria do not commonly elaborate superantigens. Adjunctive treatment with intravenous immunoglobulin, which is advantageous in streptococcal toxic shock syndrome, may not benefit these patients.


Asunto(s)
Bacteriemia/inmunología , Neoplasias/microbiología , Infecciones Estreptocócicas/inmunología , Superantígenos/biosíntesis , Estreptococos Viridans/inmunología , Adolescente , Bacteriemia/metabolismo , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Infecciones Estreptocócicas/metabolismo , Estreptococos Viridans/metabolismo
9.
J Infect Dis ; 191(12): 2121-9, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15897999

RESUMEN

The role played by host-pathogen interactions in regulation of expression of streptococcal virulence factors in vivo is beginning to become clear. We have reported that the expression of 2 streptococcal virulence factors, the streptococcal pyrogenic exotoxin (Spe) A and the cysteine protease SpeB, was reciprocally modulated during infection with Streptococcus pyogenes. To identify host signals mediating this reciprocal regulation, we cocultured clonal M1T1 isolates with human peripheral blood mononuclear cells (PBMCs). In accordance with our in vivo findings, when bacteria were in direct contact with human PBMCs or were separated in transwells, expression of speA was induced, whereas expression of speB was down-regulated. This phenomenon was mediated by transferrin and lactoferrin and was influenced by the iron-saturation status of these proteins. Iron chelation from media induced expression of speA, but to a much lesser degree than did that with apotransferrin and lactoferrin, suggesting additional effects of these ferrins on modulation of expression of speA and speB. Thus, ferrins may play an important role in host-pathogen interactions in skin and mucosal tissues.


Asunto(s)
Apoproteínas/fisiología , Proteínas Bacterianas/biosíntesis , Exotoxinas/biosíntesis , Lactoferrina/fisiología , Proteínas de la Membrana/biosíntesis , Streptococcus pyogenes/inmunología , Superantígenos/biosíntesis , Transferrina/fisiología , Antígenos Bacterianos/biosíntesis , Regulación hacia Abajo , Regulación Bacteriana de la Expresión Génica , Humanos , Leucocitos Mononucleares/fisiología , Regulación hacia Arriba
10.
Mol Microbiol ; 56(3): 681-95, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15819624

RESUMEN

The pathogen group A Streptococcus (GAS) produces a wide spectrum of infections including necrotizing fasciitis (NF). Streptolysin S (SLS) produces the hallmark beta-haemolytic phenotype produced by GAS. The nine-gene GAS locus (sagA-sagI) resembling a bacteriocin biosynthetic operon is necessary and sufficient for SLS production. Using precise, in-frame allelic exchange mutagenesis and single-gene complementation, we show sagA, sagB, sagC, sagD, sagE, sagF and sagG are each individually required for SLS production, and that sagE may further serve an immunity function. Limited site-directed mutagenesis of specific amino acids in the SagA prepropeptide supports the designation of SLS as a bacteriocin-like toxin. No significant pleotrophic effects of sagA deletion were observed on M protein, capsule or cysteine protease production. In a murine model of NF, the SLS-negative M1T1 GAS mutant was markedly diminished in its ability to produce necrotic skin ulcers and spread to the systemic circulation. The SLS toxin impaired phagocytic clearance and promoted epithelial cell cytotoxicity, the latter phenotype being enhanced by the effects of M protein and streptolysin O. We conclude that all genetic components of the sag operon are required for expression of functional SLS, an important virulence factor in the pathogenesis of invasive M1T1 GAS infection.


Asunto(s)
Proteínas Bacterianas/fisiología , Operón , Infecciones Estreptocócicas/microbiología , Streptococcus/genética , Streptococcus/patogenicidad , Estreptolisinas/fisiología , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Prueba de Complementación Genética , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fagocitosis/genética , Enfermedades Cutáneas Bacterianas/microbiología , Estreptolisinas/genética , Estreptolisinas/metabolismo , Virulencia/genética
11.
J Infect Dis ; 187(3): 398-407, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12552423

RESUMEN

Streptococcal pyrogenic exotoxin (Spe) B, a streptococcal cysteine protease, is believed to be important in group A streptococcal (GAS) pathogenesis. The present study examined the effect of SpeB on the activity of superantigenic exotoxins secreted by M1T1 GAS isolates. The proliferative response of human lymphocytes to culture supernatant (SUP) from an SpeB(+) isolate increased significantly (P<.05) when the isolate was grown with N-[N-(L-3-trans-carboxyoxirane-2-carbonyl)-L-leucyl]-agmatine, a cysteine protease inhibitor. The lymphocyte-stimulating activity of SUP from a spontaneous SpeB(-) variant or SpeB(-) knockout (DeltaSpeB) mutant was also significantly higher than that of SUP from the SpeB(+) parent isolate (P<.001). The addition of recombinant SpeB to the DeltaSpeB mutant reduced the lymphocyte response to a level comparable to that with the SpeB(+) isolate. SpeB affected superantigens that stimulate cells expressing T cell receptor Vbeta (TCRBV)-4, TCRBV7, and TCRBV8 but not those that stimulate TCRBV2. SpeB has a selective proteolytic effect on GAS superantigens.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Leucina/análogos & derivados , Streptococcus/enzimología , Streptococcus/inmunología , Superantígenos/inmunología , Superantígenos/metabolismo , Proteínas Bacterianas , División Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Cisteína Endopeptidasas/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Leucina/farmacología , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA