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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 72(12): 4261-4278, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37926727

RESUMEN

A combination of chemotherapy with immunotherapy has been proposed to have better clinical outcomes in Pancreatic Ductal Adenocarcinoma (PDAC). On the other hand, chemotherapeutics is known to have certain unwanted effects on the tumor microenvironment that may mask the expected beneficial effects of immunotherapy. Here, we have investigated the effect of gemcitabine (GEM), on two immune checkpoint proteins (PD-L1 and PD-L2) expression in cancer associated fibroblasts (CAFs) and pancreatic cancer cells (PCCs). Findings of in vitro studies conducted by using in-culture activated mouse pancreatic stellate cells (mPSCs) and human PDAC patients derived CAFs demonstrated that GEM significantly induces PD-L1 and PD-L2 expression in these cells. Moreover, GEM induced phosphorylation of STAT1 and production of multiple known PD-L1-inducing secretory proteins including IFN-γ in CAFs. Upregulation of PD-L1 in PSCs/CAFs upon GEM treatment caused T cell inactivation and apoptosis in vitro. Importantly, Statins suppressed GEM-induced PD-L1 expression both in CAFs and PCCs while abrogating the inactivation of T-cells caused by GEM-treated PSCs/CAFs. Finally, in an immunocompetent syngeneic orthotopic mouse pancreatic tumor model, simvastatin and GEM combination therapy significantly reduced intra-tumor PD-L1 expression and noticeably reduced the overall tumor burden and metastasis incidence. Together, the findings of this study have provided experimental evidence that illustrates potential unwanted side effects of GEM that could hamper the effectiveness of this drug as mono and/or combination therapy. At the same time the findings also suggest use of statins along with GEM will help in overcoming these shortcomings and warrant further clinical investigation.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Antígeno B7-H1/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Gemcitabina , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Neoplasias Pancreáticas/patología , Resultado del Tratamiento , Microambiente Tumoral
2.
FASEB J ; 36(10): e22574, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36165227

RESUMEN

In this study, the caprine pancreas has been presented as an alternative to the porcine organ for pancreatic xenotransplantation with lesser risk factors. The obtained caprine pancreas underwent a systematic cycle of detergent perfusion for decellularization. It was perfused using anionic (0.5% w/v sodium dodecyl sulfate) as well as non-ionic (0.1% v/v triton X-100, t-octyl phenoxy polyethoxy ethanol) detergents and washed intermittently with 1XPBS supplemented with 0.1% v/v antibiotic and nucleases in a gravitation-driven set-up. After 48 h, a white decellularized pancreas was obtained, and its extracellular matrix (ECM) content was examined for scaffold-like properties. The ECM content was assessed for removal of cellular content, and nuclear material was evaluated with temporal H&E staining. Quantified DNA was found to be present in a negligible amount in the resultant decellularized pancreas tissue (DPT), thus prohibiting it from triggering any immunogenicity. Collagen and fibronectin were confirmed to be preserved upon trichrome and immunohistochemical staining, respectively. SEM and AFM images reveal interconnected collagen fibril networks in the DPT, confirming that collagen was unaffected. sGAG was visualized using Prussian blue staining and quantified with DMMB assay, where DPT has effectively retained this ECM component. Uniaxial tensile analysis revealed that DPT possesses better elasticity than NPT (native pancreatic tissue). Physical parameters like tensile strength, stiffness, biodegradation, and swelling index were retained in the DPT with negligible loss. The cytocompatibility analysis of DPT has shown no cytotoxic effect for up to 72 h on normal insulin-producing cells (MIN-6) and cancerous glioblastoma (LN229) cells in vitro. The scaffold was recellularized using isolated mouse islets, which have established in vitro cell proliferation for up to 9 days. The scaffold received at the end of the decellularization cycle was found to be non-toxic to the cells, retained biological and physical properties of the native ECM, suitable for recellularization, and can be used as a safer and better alternative as a transplantable organ from a xenogeneic source.


Asunto(s)
Detergentes , Insulinas , Animales , Antibacterianos/farmacología , Colágeno/metabolismo , ADN/metabolismo , Matriz Extracelular Descelularizada , Detergentes/química , Detergentes/metabolismo , Detergentes/farmacología , Etanol/farmacología , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Cabras , Insulinas/análisis , Insulinas/metabolismo , Insulinas/farmacología , Ratones , Octoxinol/análisis , Octoxinol/metabolismo , Octoxinol/farmacología , Páncreas , Estudios Prospectivos , Dodecil Sulfato de Sodio/análisis , Dodecil Sulfato de Sodio/metabolismo , Dodecil Sulfato de Sodio/farmacología , Porcinos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
3.
FASEB J ; 36(8): e22449, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35839070

RESUMEN

The presence of activated pancreatic stellate cells (PSCs) in the pancreatic ductal adenocarcinoma (PDAC) microenvironment plays a significant role in cancer progression. Macrophage migration inhibitory factor (MIF) is overexpressed in PDAC tissues and expressed by both cancer and stromal cells. The pathophysiological role of MIF in PDAC-associated fibroblasts or PSCs is yet to be elucidated. Here we report that the PSCs of mouse or cancer-associated fibroblast cells (CAFs) of human expresses MIF and its receptors, whose expression gets upregulated upon LPS or TNF-α stimulation. In vitro functional experiments showed that MIF significantly conferred a survival advantage to CAFs/PSCs upon growth factor deprivation. Genetic or pharmacological inhibition of MIF also corroborated these findings. Further, co-injection of mouse pancreatic cancer cells with PSCs isolated from Mif-/- or Mif+/+ mice confirmed the pro-survival effect of MIF in PSCs and also demonstrated the pro-tumorigenic role of MIF expressed by CAFs in vivo. Differential gene expression analysis and in vitro mechanistic studies indicated that MIF expressed by activated CAFs/PSCs confers a survival advantage to these cells by suppression of interferon pathway induced p53 dependent apoptosis.


Asunto(s)
Apoptosis , Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Factores Inhibidores de la Migración de Macrófagos , Neoplasias Pancreáticas , Animales , Apoptosis/genética , Apoptosis/fisiología , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral/metabolismo , Movimiento Celular , Proliferación Celular , Humanos , Interferones/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Pancreáticas
4.
World J Microbiol Biotechnol ; 39(7): 171, 2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-37101059

RESUMEN

Characterization of new potential probiotics is desirable in the field of research on probiotics for their extensive use in health and disease. Tribes could be an unusual source of probiotics due to their unique food habits and least dependence on medications and consumption of antibiotics. The aim of the present study is to isolate lactic acid bacteria from tribal fecal samples of Odisha, India, and characterize their genetic and probiotic attributes. In this context one of the catalase-negative and Gram-positive isolates, identified using 16S rRNA sequencing as Ligilactobacillus salivarius, was characterized in vitro for its acid and bile tolerance, cell adhesion and antimicrobial properties. The whole genome sequence was obtained and analyzed for strain level identification, presence of genomic determinants for probiotic-specific features, and safety. Genes responsible for its antimicrobial and immunomodulatory functions were detected. The secreted metabolites were analyzed using high resolution mass spectroscopy; the results indicated that the antimicrobial potential could be due to the presence of pyroglutamic acid, propionic acid, lactic acid, 2-hydroxyisocaproic acid, homoserine, and glutathione, and the immuno-modulating activity, contributed by the presence of short chain fatty acids such as acetate, propionate, and butyrate. So, to conclude we have successfully characterized a Ligilactobacillus salivarius species with potential antimicrobial and immunomodulatory ability. The health-promoting effects of this probiotic strain and/or its derivatives will be investigated in future.


Asunto(s)
Ligilactobacillus salivarius , Probióticos , ARN Ribosómico 16S/genética , Antibacterianos/farmacología , Genómica
5.
Lab Invest ; 102(3): 298-311, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34773069

RESUMEN

Pancreatic cancer (PC) is highly resistant to chemo and radiotherapy. Radiation-induced fibrosis (RIF) is a major cause of clinical concern for various malignancies, including PC. In this study, we aimed to evaluate the radiosensitizing and anti-RIF potential of fluvastatin in PC. Short-term viability and clonogenic survival assays were used to evaluate the radiosensitizing potential of fluvastatin in multiple human and murine PC cell lines. The expression of different proteins was analyzed to understand the mechanisms of fluvastatin-mediated radiosensitization of PC cells and its anti-RIF effects in both mouse and human pancreatic stellate cells (PSCs). Finally, these effects of fluvastatin and/or radiation were assessed in an immune-competent syngeneic murine model of PC. Fluvastatin radiosensitized multiple PC cell lines, as well as radioresistant cell lines in vitro, by inhibiting radiation-induced DNA damage repair response. Nonmalignant cells, such as PSCs and NIH3T3 cells, were less sensitive to fluvastatin-mediated radiosensitization than PC cells. Interestingly, fluvastatin suppressed radiation and/or TGF-ß-induced activation of PSCs, as well as the fibrogenic properties of these cells in vitro. Fluvastatin considerably augmented the antitumor effect of external radiation therapy and also suppressed intra-tumor RIF in vivo. These findings suggested that along with radiation, fluvastatin co-treatment may be a potential therapeutic approach against PC.


Asunto(s)
Fluvastatina/farmacología , Neoplasias Pancreáticas/patología , Tolerancia a Radiación/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Embrión no Mamífero/efectos de la radiación , Fibrosis/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Experimentales/radioterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Pez Cebra/embriología
6.
EMBO J ; 37(18)2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30143514

RESUMEN

Sequestration of protein aggregates in inclusion bodies and their subsequent degradation prevents proteostasis imbalance, cytotoxicity, and proteinopathies. The underlying molecular mechanisms controlling the turnover of protein aggregates are mostly uncharacterized. Herein, we show that a TRIM family protein, TRIM16, governs the process of stress-induced biogenesis and degradation of protein aggregates. TRIM16 facilitates protein aggregate formation by positively regulating the p62-NRF2 axis. We show that TRIM16 is an integral part of the p62-KEAP1-NRF2 complex and utilizes multiple mechanisms for stabilizing NRF2. Under oxidative and proteotoxic stress conditions, TRIM16 activates ubiquitin pathway genes and p62 via NRF2, leading to ubiquitination of misfolded proteins and formation of protein aggregates. We further show that TRIM16 acts as a scaffold protein and, by interacting with p62, ULK1, ATG16L1, and LC3B, facilitates autophagic degradation of protein aggregates. Thus, TRIM16 streamlines the process of stress-induced aggregate clearance and protects cells against oxidative/proteotoxic stress-induced toxicity in vitro and in vivo Taken together, this work identifies a new mechanism of protein aggregate turnover, which could be relevant in protein aggregation-associated diseases such as neurodegeneration.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Agregado de Proteínas , Proteolisis , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Complejos Multiproteicos/genética , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Ubiquitinación/genética
7.
Biochem Biophys Res Commun ; 635: 179-186, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36279679

RESUMEN

Amalgamation of target-based approach along with chemo and/or radiotherapy could be an effective strategy to combat pancreatic cancer (PC). ATAD2 (ATPase family AAA domain containing 2) is a potential oncoprotein and a poor prognostic factor in PC. ATAD2 inhibition sensitizes pancreatic cancer cells (PCCs) to gemcitabine (GEM). ATAD2 silencing also enhances radio-sensitivity in lung cancer cells. We therefore hypothesise that ATAD2 suppression along with application of GEM and radiation could show additive/synergistic response in reducing PC progression. At first, immunofluorescence and western blot analysis are carried out to investigate ATAD2 expression upon irradiation in PCCs (BxPC3, and PANC1). Colony forming assay is performed to analyse the radio-sensitization effect of ATAD2 suppressed PCCs in absence and presence of GEM. To evaluate the consequences of irradiation, expression of markers for DNA damage and repair as well as apoptosis and autophagy are also assessed. Results indicate that ATAD2 is elevated in irradiated PCCs. Silencing ATAD2 sensitizes PCCs to radiation. Survival fraction analysis of PCCs shows that combination treatment of GEM and radiation (with minimal doses) is more effective when ATAD2 is suppressed. Further, when GEM-radiation combination is applied to ATAD2 silenced PCCs, expression of DNA damage marker like γH2AX is induced, whereas DNA damage repair proteins (pChk1, and pChk2) are downregulated. Moreover, ATAD2 suppressed PCCs are more susceptible to apoptosis and autophagy in presence of radiation and GEM. Collectively, these findings support our hypothesis and demonstrate that targeting ATAD2 enhances efficacy of GEM-radiation treatment in PCCs.


Asunto(s)
Desoxicitidina , Neoplasias Pancreáticas , Humanos , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/metabolismo , Línea Celular Tumoral , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Gemcitabina , Neoplasias Pancreáticas
8.
FASEB J ; 35(7): e21713, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34105201

RESUMEN

Syrian golden hamsters (Mesocricetus auratus) infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) manifests lung pathology. In this study, efforts were made to check the infectivity of a local SARS-CoV-2 isolate in a self-limiting and non-lethal hamster model and evaluate the differential expression of lung proteins during acute infection and convalescence. The findings of this study confirm the infectivity of this isolate in vivo. Analysis of clinical parameters and tissue samples show the pathophysiological manifestation of SARS-CoV-2 infection similar to that reported earlier in COVID-19 patients and hamsters infected with other isolates. However, diffuse alveolar damage (DAD), a common histopathological feature of human COVID-19 was only occasionally noticed. The lung-associated pathological changes were very prominent on the 4th day post-infection (dpi), mostly resolved by 14 dpi. Here, we carried out the quantitative proteomic analysis of the lung tissues from SARS-CoV-2-infected hamsters on day 4 and day 14 post-infection. This resulted in the identification of 1585 proteins of which 68 proteins were significantly altered between both the infected groups. Pathway analysis revealed complement and coagulation cascade, platelet activation, ferroptosis, and focal adhesion as the top enriched pathways. In addition, we also identified altered expression of two pulmonary surfactant-associated proteins (Sftpd and Sftpb), known for their protective role in lung function. Together, these findings will aid in understanding the mechanism(s) involved in SARS-CoV-2 pathogenesis and progression of the disease.


Asunto(s)
COVID-19/metabolismo , COVID-19/patología , Interacciones Huésped-Patógeno , Pulmón/metabolismo , Pulmón/virología , Proteómica , SARS-CoV-2/patogenicidad , Animales , COVID-19/virología , Cricetinae , Modelos Animales de Enfermedad , Femenino , Pulmón/patología , Masculino , Proteoma/análisis , Proteoma/biosíntesis , Reproducibilidad de los Resultados , Carga Viral
9.
Cell Biol Toxicol ; 38(1): 111-127, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33651227

RESUMEN

The key to bacterial virulence relies on an exquisite balance of signals between microbe and hosts. Bacterial toxin-antitoxin (TA) system is known to play a vital role in response to stress adaptation, drug resistance, biofilm formation, intracellular survival, persistence as well as pathogenesis. In the present study, we investigated the role of Hha-TomB TA system in regulating virulence of Salmonella enterica serovar Typhimurium (S. Typhimurium) in a host model system, where we showed that deletion of hha and tomB genes displayed impaired cell adhesion, invasion, and uptake. The isogenic hha and tomB mutant strain was also found to be deficient in intracellular replication in vitro, with a highly repressed Salmonella Pathogenicity Island-2 (SPI-2) genes and downregulation of Salmonella Pathogenicity Island-1 (SPI-1) genes. In addition, the Δhha and ΔtomB did not show acute colitis in C57BL/6 mice and displayed less dissemination to systemic organs followed by their cecal pathology. The TA mutants also showed reduction in serum cytokine and nitric oxide levels both in vitro and in vivo. However, the inflammation phenotype was restored on complementing strain of TA gene to its mutant strain. In silico studies depicted firm interaction of Hha-TomB complex and the regulatory proteins, namely, SsrA, SsrB, PhoP, and PhoQ. Overall, we demonstrate that this study of Hha-TomB TA system is one of the prime regulating networks essential for S. Typhimurium pathogenesis. 1. Role of Hha-TomB toxin-antitoxin (TA) system in Salmonella pathogenesis was examined. 2. The TA mutants resulted in impaired invasion and intracellular replication in vitro. 3. The TA mutants displayed alteration in SPI-1 and SPI-2 regulatory genes inside host cells. 4. Mutation in TA genes also limited systemic colonization and inflammatory response in vivo.


Asunto(s)
Antitoxinas , Salmonella typhimurium , Animales , Antitoxinas/genética , Antitoxinas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Inmunidad , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Serogrupo
10.
Prostate ; 80(15): 1341-1352, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32835423

RESUMEN

BACKGROUND: The role of microbiota in the pathophysiology of benign prostate hyperplasia (BPH), especially in creating an inflammatory milieu may not be avoided. The major objectives of this study were to investigate the microbial composition of BPH tissues, its association with inflammation and check the effect of clinically isolated bacteria on prostate epithelial cells. METHODS: The study includes 36 patients with a pathological diagnosis of BPH. Following strict aseptic measures, tissues were collected after transurethral resection of prostate, multiple pieces of the resected tissues were subjected to histopathological analysis, bacterial culture and genomic DNA extraction. Microbial composition was analyzed by culture and/or next-generation sequencing methods. Annotation of operational taxonomy unit has been done with an in-house algorithm. The extent of inflammation was scored through histological evaluation of tissue sections. The effect of clinical isolates on nuclear factor-κB (NF-κB) activity and induction of DNA-damage in the prostate epithelial cells were evaluated. RESULTS: Histopathological analysis of the BPH tissues showed the presence of inflammation in almost all the tissues with a varied level at different regions of the same tissue section and the level of overall inflammation was different from patients to patients. Microbial culture of tissue samples showed the presence of live bacteria in 55.5% (20 out of 36) of the patient tissues. Majority of the isolates were coagulase-positive Staphylococcus, E. coli and Micrococcus spp. Further, V3 16S rRNA sequencing of the DNA isolated from BPH tissues showed the presence of multiple bacteria and the most common phylum in the BPH tissues were found to be Proteobacteria, Actinobacteria, Firmicutes, and Bacteroidetes. The E. coli, isolated from one of the tissue was able to activate NF-κB and induce DNA damage in prostate epithelial cells. Phospho-histone γH2A.X staining confirmed the presence of cells with damaged DNA lesion in BPH tissues and also correlated with the severity of inflammation. CONCLUSION: Our study has shown that the BPH tissues do have a divergent microbial composition including the commonly found E. coli (phylum Proteobacteria), and these bacteria might contribute to the BPH-associated inflammation and/or tissue damage. The BPH-associated E. coli induced NF-κB signaling and DNA damage in prostate epithelial cells in vitro.


Asunto(s)
Daño del ADN , Células Epiteliales/microbiología , Escherichia coli/aislamiento & purificación , Inflamación/microbiología , Próstata/microbiología , Hiperplasia Prostática/microbiología , Células Epiteliales/patología , Humanos , Inflamación/patología , Masculino , Persona de Mediana Edad , Próstata/patología , Próstata/cirugía , Hiperplasia Prostática/patología , Hiperplasia Prostática/cirugía , Resección Transuretral de la Próstata
11.
Prostate ; 79(2): 168-182, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30264470

RESUMEN

BACKGROUND: Previous studies have shown the effect of bacterial lipopolysaccharide (LPS) on enhanced cancer cells' growth and metastasis. However, the effect of LPS on prostate cancer (PCa) cells metastasis has not been investigated in details. This study aimed to investigate the functional role of LPS on PCa cells metastasis and determine the effect of dexamethasone (DEX) on this event. METHODS: Two different PCa reporter cells lines (DU145-NF-κB-Luc and MAT-LyLu- NF-κB-Luc) were used to assess the direct effect of LPS on NF-κB activation in PCa cells. Plasma collected from LPS-stimulated human and rodent blood were used to check the indirect effect of LPS on NF-κB activation in PCa cells. Trans-well migration assay and two different orthotopic PCa animal models were used to investigate the effect of LPS on DU145 and MAT-LyLu cells migration or metastasis in vitro and in vivo, respectively. In all the studies DEX was used with or without LPS stimulation. RESULTS: LPS and secretory factors present in plasma collected from LPS-stimulated blood, significantly activated NF-κB in DU145, and MAT-LyLu cells and enhanced their migration in vitro. DEX significantly suppressed LPS-mediated activation of cancer and blood cells and abrogated the direct and indirect pro-migratory effect of LPS on PCa cells. Systemic administration of LPS activated NF-κB in DU145 cells in vivo; however, failed to alter the metastatic properties of these cells. On the other hand, systemic administration of LPS to MAT-LyLu tumor bearing animals significantly enhanced the incidence of metastasis without altering the overall growth of primary tumors. Unexpectedly, though DEX significantly suppressed MAT-LyLu primary tumor weights, it aggravated metastasis of cancer cells in presence and absence of LPS. Moreover, consecutive DEX pre-treatment enhanced experimental peritoneal metastasis of MAT-LyLu cells. At the molecular level, LPS, and/or DEX induced overexpression of immunosuppressive molecules in MAT-LyLu tumors. CONCLUSIONS: Overall, our study has shown that LPS and/or LPS induced inflammation can increase PCa metastasis and immunosuppressive dose of DEX might further enhance cancer metastasis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Dexametasona/farmacología , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Xenoinjertos , Humanos , Lipopolisacáridos/sangre , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias de la Próstata/sangre , Distribución Aleatoria
12.
Lab Invest ; 98(1): 27-40, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29155423

RESUMEN

Desmoplasia is a fibro-inflammatory process and a well-established feature of pancreatic cancer. A key contributor to pancreatic cancer desmoplasia is the pancreatic stellate cell. Various in vitro and in vivo methods have emerged for the isolation, characterization, and use of pancreatic stellate cells in models of cancer-associated fibrosis. In addition to cell culture models, genetically engineered animal models have been established that spontaneously develop pancreatic cancer with desmoplasia. These animal models are currently being used for the study of pancreatic cancer pathogenesis and for evaluating therapeutics against pancreatic cancer. Here, we review various in vitro and in vivo models that are being used or have the potential to be used to study desmoplasia in pancreatic cancer.


Asunto(s)
Investigación Biomédica/métodos , Modelos Animales de Enfermedad , Fibroma/etiología , Neoplasias Pancreáticas/fisiopatología , Animales , Animales Modificados Genéticamente , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Investigación Biomédica/tendencias , Línea Celular Tumoral , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Femenino , Fibroma/tratamiento farmacológico , Fibroma/inmunología , Fibroma/patología , Fibrosis , Humanos , Masculino , Ratones , Trasplante de Neoplasias/métodos , Trasplante de Neoplasias/tendencias , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/inmunología , Células Estrelladas Pancreáticas/patología , Células Estrelladas Pancreáticas/trasplante , Ratas , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
13.
Mol Carcinog ; 57(9): 1102-1115, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29672923

RESUMEN

Epithelial-mesenchymal transition (EMT) is a critical event that occurs during the invasion and metastatic spread of cancer cells. Here, we conceive a dual mechanism of Par-4-mediated inhibition of EMT and induction of MET in metastatic pancreatic cancer cells. First, we demonstrate that 1,1'-ß-D-glucopyranosyl-3,3'-bis(5-bromoindolyl)-octyl methane (NGD16), an N-glycosylated derivative of medicinally important phytochemical 3,3'-diindolylmethane (DIM) abrogates EMT by inducing pro-apoptotic protein Par-4. Induction of Par-4 (by NGD16 or ectopic overexpression) strongly impedes invasion with inhibition of major mesenchymal markers viz. Vimentin and Twist-1 epithelial marker- E-cadherin. Further, NGD16 triggers MET phenotypes in pancreatic cancer cells by augmenting ALK2/Smad4 signaling in a Par-4-dependent manner. Conversely, siRNA-mediated silencing of endogenous Par-4 unveil reversal of MET with diminished E-cadherin expression and invasive phenotypes. Additionally, we demonstrate that intact Smad4 is essential for Par-4-mediated maintenance of E-cadherin level in MET induced cells. Notably, we imply that Par-4 induction regulates E-cadherin levels in the pancreatic cancer cells via modulating Twist-1 promoter activity. Finally, in vivo studies with syngenic mouse metastatic pancreatic cancer model reveal that NGD16 strongly suppresses metastatic burden, ascites formation, and prolongs the overall survival of animals effectively.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/genética , Animales , Cadherinas/genética , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Masculino , Ratones Endogámicos C57BL , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Interferencia de ARN , ARN Interferente Pequeño/genética
15.
Prostate ; 75(10): 1020-33, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25833062

RESUMEN

BACKGROUND: Prostate cancer (PCa) cells express Toll-like receptor-4 (TLR4), a known pro-tumorigenic molecule for different cancer cells. The cancer cells residing in the avascular region of the tumor confront various metabolic stresses and continuously adapt mechanisms to overcome them. We hypothesized that TLR4 activation might provide direct survival advantage to metabolically stressed PCa cells. METHODS: We first investigated the effect of LPS on survival of serum deprived PCa cells. To understand the molecular mechanisms involved in TLR4 mediated PCa survival, we next investigated change in expression of markers for apoptosis, senescence and autophagy. Ultimately, the effect of LPS on established prostate tumors was confirmed in vivo using a syngeneic rat model for PCa. RESULTS: Lipopolysaccharide (LPS)-mediated TLR4 activation significantly enhanced survival of serum deprived (SD) PC3, DU145 and MAT-LyLu PCa cells. TLR4 inhibition by a specific inhibitor resulted in rapid death of SD-PC3 cells, which was significantly suppressed by LPS. Interestingly, LPS treatment suppressed macroautophagy in SD-PC3 cells and increased expression of CCL2 (C-C motif ligand-2), a known autophagy inhibitor and pro-survival factor. Intra-tumor LPS injection resulted in increased tumor mass, induced TLR4 activation, suppressed autophagy, and increased the macrophage population in MAT-LyLu-tumors. CONCLUSIONS: Our study reveals that bacterial LPS enhance survival of PCa cells under conditions of nutrient stress through TLR4 activation. Moreover, LPS induces overexpression of CCL2 involved in the suppression of starvation-induced macroautophagy in PCa cells, and enhanced macrophage population in prostate tumors in vivo. Taken together, the current study suggests the importance of bacterial infection or TLR4-activation in prostate cancer pathogenesis.


Asunto(s)
Supervivencia Celular/fisiología , Lipopolisacáridos/farmacología , Neoplasias de la Próstata/patología , Receptor Toll-Like 4/fisiología , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Quimiocina CCL2/análisis , Medio de Cultivo Libre de Suero , Escherichia coli , Humanos , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Masculino , Trasplante de Neoplasias , Neoplasias Experimentales , Neoplasias de la Próstata/química , ARN Mensajero/análisis , ARN Interferente Pequeño/farmacología , Ratas , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética
16.
Trends Biochem Sci ; 35(4): 236-45, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19913432

RESUMEN

Mucins are high molecular weight glycoproteins that are involved in regulating diverse cellular activities both in normal and pathological conditions. Mucin activity and localization is mediated by several molecular mechanisms, including discrete interactions with other proteins. An understanding of the biochemistry behind the known interactions between mucins and other proteins, coupled with an appreciation of their pathophysiological significance, can lend insight into the development of novel therapeutic agents. Indeed, a recent study demonstrated that a cell permeable inhibitor, PMIP, that disrupts the MUC1-EGFR interaction, is effective in killing breast cancer cells in vitro and in tumor models.


Asunto(s)
Mucinas/antagonistas & inhibidores , Mucinas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Humanos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo
17.
Antimicrob Agents Chemother ; 58(11): 6896-903, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25199778

RESUMEN

We successfully produced two human ß-defensins (hBD-1 and hBD-2) in bacteria as functional peptides and tested their antibacterial activities against Salmonella enterica serovar Typhi, Escherichia coli, and Staphylococcus aureus employing both spectroscopic and viable CFU count methods. Purified peptides showed approximately 50% inhibition of the bacterial population when used individually and up to 90% when used in combination. The 50% lethal doses (LD50) of hBD-1 against S. Typhi, E. coli, and S. aureus were 0.36, 0.40, and 0.69 µg/µl, respectively, while those for hBD-2 against the same bacteria were 0.38, 0.36, and 0.66 µg/µl, respectively. Moreover, we observed that bacterium-derived antimicrobial peptides were also effective in increasing survival time and decreasing bacterial loads in the peritoneal fluid, liver, and spleen of a mouse intraperitoneally infected with S. Typhi. The 1:1 hBD-1/hBD-2 combination showed maximum effectiveness in challenging the Salmonella infection in vitro and in vivo. We also observed less tissue damage and sepsis formation in the livers of infected mice after treatment with hBD-1 and hBD-2 peptides individually or in combination. Based on these findings, we conclude that bacterium-derived recombinant ß-defensins (hBD-1 and hBD-2) are promising antimicrobial peptide (AMP)-based substances for the development of new therapeutics against typhoid fever.


Asunto(s)
Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Salmonella/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , beta-Defensinas/uso terapéutico , Animales , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Clonación Molecular , Quimioterapia Combinada , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/microbiología , Femenino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes/uso terapéutico , Infecciones por Salmonella/microbiología , Salmonella typhi/efectos de los fármacos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos
19.
Ther Deliv ; : 1-24, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38949622

RESUMEN

Aim: The current study aims to develop and optimize microemulsions (ME) through Quality-by-Design (QbD) approach to improve the aqueous solubility and dissolution of poorly water-soluble drug disulfiram (DSF) for repurposing in melanoma and breast cancer therapy. Materials & methods: The ME was formulated using Cinnamon oil & Tween® 80, statistically optimized using a D-optimal mixture design-based QbD approach to develop the best ME with low vesicular size (Zavg) and polydispersity index (PDI). Results: The DSF-loaded optimized stable ME showed enhanced dissolution, in-vitro cytotoxicity and improved cellular uptake in B16F10 and MCF-7 cell lines compared with their unformulated free DSF. Conclusion: Our investigations suggested the potential of the statistically designed DSF-loaded optimized ME for repurposing melanoma and breast cancer therapy.


Identifying new medicinal uses of an existing marketed drug can save both money and time in the process of drug development. From many of the recently reported literature, disulfiram (a drug used for alcoholism) has shown its activity against various cancers, including breast and skin cancer. However, it possesses poor water solubility and absorption, leading to low medicinal activity. The current study aims to develop a novel microemulsion dosage form through a statistical design approach to enhance the solubility, dissolution and anticancer activity for repurposing in melanoma and breast cancer treatment. The novel microemulsion was prepared, statistically analyzed and optimized. The optimized microemulsion was found to be stable and showed improved medicinal activity against breast and skin cancer compared with the pure drug. Our research showed the potential of the developed microemulsion of the disulfiram for its new therapeutic use in skin cancer and breast cancer.

20.
Artículo en Inglés | MEDLINE | ID: mdl-38345744

RESUMEN

Isolation and characterization of probiotics from traditional fermented food have contributed many beneficial strains to the field of health and nutritional sciences. Handia, a traditional fermented alcoholic beverage popular in different parts of Odisha, was our source of isolation. This study characterizes one such potential bacteria, Levilactobacillus brevis ILSH3 (H3) isolated from Handia. The investigation for the probiotic attributes as per ICMR-DBT guidelines qualified the checkpoint assays like acid and bile tolerance, bile salt hydrolase activity, antimicrobial properties, and pathogen exclusion ability. The whole genome sequence of H3 (2,460,966 bp in size with GC content of 45.62%) was subjected to comparative genome analysis for its taxonomic identification and validation of probiotic potential. Various genes pertaining to its probiotic potential were identified in the genome and it showed zero matches against any pathogenic families. Metabolite profiling of cell-free supernatant using liquid chromatography-mass spectrometry revealed the presence of essential amino acids, short-chain fatty acids, antimicrobial molecules, immunomodulatory molecules, and flavor/aroma-enhancing compounds. Immunomodulatory property investigation using Bioplex and qRT-PCR showed a reduction in the levels of pro-inflammatory cytokines in L. brevis ILSH3-treated Caco-2 cells. Collectively, the results demonstrate that this Handia-origin bacteria Levilactobacillus brevis ILSH3 possesses desirable attributes of a probiotic, which is now open for nutritional and health biologists to explore. This new probiotic strain may show promising results when utilized in healthcare or functional foods.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA