Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol In Vitro ; 88: 105540, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36563973

RESUMEN

Mass spectrometry based 'omics pairs well with organ-on-a-chip-based investigations, which often have limited cellular material for sampling. However, a common issue with these chip-based platforms is well-to-well or chip-to-chip variability in the proteome and metabolome due to factors such as plate edge effects, cellular asynchronization, effluent flow, and limited cell count. This causes high variability in the quantitative multi-omics analysis of samples, potentially masking true biological changes within the system. Solutions to this have been approached via data processing tools and post-acquisition normalization strategies such as constant median, constant sum, and overall signal normalization. Unfortunately, these methods do not adequately correct for the large variations, resulting in a need for increased biological replicates. The methods in this work utilize a dansylation based assay with a subset of labeled metabolites that allow for pre-acquisition normalization to better correlate the biological perturbations that truly occur in chip-based platforms. BCA protein assays were performed in tandem with a proteomics pipeline to achieve pre-acquisition normalization. The CN Bio PhysioMimix was seeded with primary hepatocytes and challenged with VX after six days of culture, and the metabolome and proteome were analyzed using the described normalization methods. A decreased coefficient of variation percentage is achieved, significant changes are observed through the proteome and metabolome, and better classification of biological replicates acquired because of these strategies.


Asunto(s)
Proteoma , Proteómica , Sistemas Microfisiológicos , Metabolómica/métodos , Espectrometría de Masas/métodos , Metaboloma
2.
Arch Toxicol ; 94(9): 3249-3264, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32720192

RESUMEN

The inhibition of acetylcholinesterase is regarded as the primary toxic mechanism of action for chemical warfare agents. Recently, there have been numerous reports suggesting that metabolic processes could significantly contribute to toxicity. As such, we applied a multi-omics pipeline to generate a detailed cascade of molecular events temporally occurring in guinea pigs exposed to VX. Proteomic and metabolomic profiling resulted in the identification of several enzymes and metabolic precursors involved in glycolysis and the TCA cycle. All lines of experimental evidence indicated that there was a blockade of the TCA cycle at isocitrate dehydrogenase 2, which converts isocitrate to α-ketoglutarate. Using a primary beating cardiomyocyte cell model, we were able to determine that the supplementation of α-ketoglutarate subsequently rescued cells from the acute effects of VX poisoning. This study highlights the broad impacts that VX has and how understanding these mechanisms could result in new therapeutics such as α-ketoglutarate.


Asunto(s)
Acetilcolinesterasa/metabolismo , Agentes Nerviosos/toxicidad , Intoxicación/tratamiento farmacológico , Proteoma/efectos de los fármacos , Animales , Sustancias para la Guerra Química/toxicidad , Cobayas , Redes y Vías Metabólicas , Metabolómica , Intoxicación/metabolismo , Proteómica
3.
PLoS One ; 11(4): e0152985, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27110941

RESUMEN

Integrated Discrete Multiple Organ Co-culture (IDMOC) is emerging as an in-vitro alternative to in-vivo animal models for pharmacology studies. IDMOC allows dose-response relationships to be investigated at the tissue and organoid levels, yet, these relationships often exhibit responses that are far more complex than the binary responses often measured in whole animals. To accommodate departure from binary endpoints, IDMOC requires an expansion of analytic techniques beyond simple linear probit and logistic models familiar in toxicology. IDMOC dose-responses may be measured at continuous scales, exhibit significant non-linearity such as local maxima or minima, and may include non-independent measures. Generalized additive mixed-modeling (GAMM) provides an alternative description of dose-response that relaxes assumptions of independence and linearity. We compared GAMMs to traditional linear models for describing dose-response in IDMOC pharmacology studies.


Asunto(s)
Modelos Biológicos , Células 3T3-L1 , Animales , Técnicas de Cocultivo , Humanos , Ratones
4.
Int J Toxicol ; 34(5): 433-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26173615

RESUMEN

Organophosphorus (OP) pesticides are known to induce pulmonary toxicity in both humans and experimental animals. To elucidate the mechanism of OP-induced cytotoxicity, we examined the effects of parathion and malathion and their respective metabolites, paraoxon and malaoxon, on primary cultured human large and small airway cells. Exposure to paraoxon and malaoxon produced a dose-dependent increase in cytotoxicity following a 24-hour exposure, while treatment with parathion or malathion produced no effects at clinically relevant concentrations. Exposure to paraoxon-induced caspase activation, but malaoxon failed to induce this response. Since caspases have a major role in the regulation of apoptosis and cell death, we evaluated OP-induced cell death in the presence of a caspase inhibitor. Pharmacological caspase inhibition protected against paraoxon-induced cell death but not malaoxon-induced cell death. These data suggest that caspase activation is a key signaling element in paraoxon-induced cell death, but not malaoxon-induced cellular death in the pulmonary epithelium.


Asunto(s)
Inhibidores de la Colinesterasa/toxicidad , Células Epiteliales/efectos de los fármacos , Insecticidas/toxicidad , Malatión/análogos & derivados , Paraoxon/toxicidad , Clorometilcetonas de Aminoácidos/farmacología , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/metabolismo , Humanos , Malatión/toxicidad , Paratión/toxicidad , Sistema Respiratorio/citología
5.
J Biomol Screen ; 19(10): 1402-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25239051

RESUMEN

In vitro models that accurately and rapidly assess hepatotoxicity and the effects of hepatic metabolism on nonliver cell types are needed by the U.S. Department of Defense and the pharmaceutical industry to screen compound libraries. Here, we report the first use of high content analysis on the Integrated Discrete Multiple Organ Co-Culture (IdMOC) system, a high-throughput method for such studies. We cultured 3T3-L1 cells in the presence and absence of primary human hepatocytes, and exposed the cultures to 4-aminophenol and cyclophosphamide, model toxicants that are respectively detoxified and activated by the liver. Following staining with calcein-AM, ethidium homodimer-1, and Hoechst 33342, high content analysis of the cultures revealed four cytotoxic endpoints: fluorescence intensities of calcein-AM and ethidium homodimer-1, nuclear area, and cell density. Using these endpoints, we observed that the cytotoxicity of 4-aminophenol in 3T3-L1 cells in co-culture was less than that observed for 3T3-L1 monocultures, consistent with the known detoxification of 4-aminophenol by hepatocytes. Conversely, cyclophosphamide cytotoxicity for 3T3-L1 cells was enhanced by co-culturing with hepatocytes, consistent with the known metabolic activation of this toxicant. The use of IdMOC plates combined with high content analysis is therefore a multi-endpoint, high-throughput capability for measuring the effects of metabolism on toxicity.


Asunto(s)
Aminofenoles/toxicidad , Ciclofosfamida/toxicidad , Pruebas de Toxicidad/métodos , Células 3T3-L1/efectos de los fármacos , Células 3T3-L1/metabolismo , Animales , Técnicas de Cocultivo , Fluorescencia , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Inactivación Metabólica , Ratones
6.
Inhal Toxicol ; 25(1): 37-62, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23293972

RESUMEN

Chemical warfare agents (CWAs) as well as biological toxins present a significant inhalation injury risk to both deployed warfighters and civilian targets of terrorist attacks. Inhalation of many CWAs and biological toxins can induce severe pulmonary toxicity leading to the development of acute lung injury (ALI) as well as acute respiratory distress syndrome (ARDS). The therapeutic options currently used to treat these conditions are very limited and mortality rates remain high. Recent evidence suggests that human stem cells may provide significant therapeutic options for ALI and ARDS in the near future. The threat posed by CWAs and biological toxins for both civilian populations and military personnel is growing, thus understanding the mechanisms of toxicity and potential therapies is critical. This review will outline the pulmonary toxic effects of some of the most common CWAs and biological toxins as well as the potential role of stem cells in treating these types of toxic lung injuries.


Asunto(s)
Armas Biológicas , Sustancias para la Guerra Química/toxicidad , Pulmón/efectos de los fármacos , Síndrome de Dificultad Respiratoria/terapia , Trasplante de Células Madre , Células Madre , Toxinas Biológicas/toxicidad , Animales , Humanos , Pulmón/metabolismo , Pulmón/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Células Madre/citología , Células Madre/fisiología
7.
PLoS One ; 6(5): e19716, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21611119

RESUMEN

Yersinia pestis is a gram negative zoonotic pathogen responsible for causing bubonic and pneumonic plague in humans. The pathogen uses a type III secretion system (T3SS) to deliver virulence factors directly from bacterium into host mammalian cells. The system contains a single ATPase, YscN, necessary for delivery of virulence factors. In this work, we show that deletion of the catalytic domain of the yscN gene in Y. pestis CO92 attenuated the strain over three million-fold in the Swiss-Webster mouse model of bubonic plague. The result validates the YscN protein as a therapeutic target for plague. The catalytic domain of the YscN protein was made using recombinant methods and its ATPase activity was characterized in vitro. To identify candidate therapeutics, we tested computationally selected small molecules for inhibition of YscN ATPase activity. The best inhibitors had measured IC(50) values below 20 µM in an in vitro ATPase assay and were also found to inhibit the homologous BsaS protein from Burkholderia mallei animal-like T3SS at similar concentrations. Moreover, the compounds fully inhibited YopE secretion by attenuated Y. pestis in a bacterial cell culture and mammalian cells at µM concentrations. The data demonstrate the feasibility of targeting and inhibiting a critical protein transport ATPase of a bacterial virulence system. It is likely the same strategy could be applied to many other common human pathogens using type III secretion system, including enteropathogenic E. coli, Shigella flexneri, Salmonella typhimurium, and Burkholderia mallei/pseudomallei species.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Portadoras/antagonistas & inhibidores , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Yersinia pestis/enzimología , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia mallei/efectos de los fármacos , Burkholderia mallei/enzimología , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Eliminación de Gen , Genes Bacterianos/genética , Células HeLa , Humanos , Hidrólisis/efectos de los fármacos , Concentración 50 Inhibidora , Cinética , Proteínas de Unión a Maltosa/metabolismo , Ratones , Modelos Moleculares , Peste/microbiología , Multimerización de Proteína/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Yersinia pestis/efectos de los fármacos , Yersinia pestis/genética , Yersinia pestis/patogenicidad
8.
J Immunother ; 30(5): 490-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17589289

RESUMEN

Current immunotherapies are limited by several factors, including the failure to recruit sufficient numbers of immune effector cells to tumors. The chemokine monokine induced by gamma-interferon (Mig; CXCL9) attracts activated T cells and natural killer (NK) cells bearing the chemokine receptor CXCR3. We investigated Mig as an immunotherapeutic agent in a syngeneic murine model of metastatic breast cancer. We transfected the highly malignant murine mammary tumor cell line 66.1 to stably express murine Mig cDNA. Immune-competent mice injected with Mig-expressing tumor cells developed smaller local tumors and fewer lung metastases, and they survived longer than mice injected with vector-control tumor cells. Mig-mediated inhibition of local tumor growth was lost in the absence of host T cells. Mig-transduced tumors had increased numbers of CD4 T cells compared with vector-control tumors, consistent with the T-cell chemoattractant property of Mig, and many tumor-infiltrating host cells expressed CXCR3. NK cells had not been examined previously as a possible effector cell in Mig-based therapies. Our studies now show that NK cells are critical to the mechanism by which Mig limits metastasis. Inhibition of angiogenesis was not implicated as a mechanism of Mig-mediated therapy in this model. These studies support the hypothesis that by manipulating the Mig-CXCR3 gradient, it is possible to direct host immune effector cells to tumors, curtailing both local tumor growth and metastasis. These studies also implicate host NK cells as an additional effector cell critical for Mig-mediated control of metastasis.


Asunto(s)
Adenocarcinoma/terapia , Quimiocinas CXC/uso terapéutico , Neoplasias Pulmonares/terapia , Neoplasias Mamarias Experimentales/terapia , Adenocarcinoma/inmunología , Adenocarcinoma/secundario , Animales , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Quimiocina CXCL9 , Quimiocinas CXC/inmunología , Femenino , Inmunoterapia , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Neovascularización Patológica
9.
Cancer Res ; 62(9): 2606-10, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11980656

RESUMEN

The cytokine interleukin (IL)-10 has potent antitumor activity in many model systems when expressed locally at very high levels from the time of tumor transplantation. We now demonstrate that systemic administration of recombinant human IL-10 to animals bearing established highly malignant mammary tumors also leads to significant growth inhibition. We had shown previously that expression of the CXC chemokines Mig (monokine induced by IFN-gamma) and IP-10 (inducible protein 10) is observed in IL-10 transduced but not neo-vector control tumors. We now demonstrate that treatment of IL-10-tumor-bearing mice with antibodies to either chemokine partially reverses the therapeutic effect of IL-10. Tumor growth in animals treated with both antibodies is comparable with that of vector control tumors. Direct transduction of Mig cDNA into the parental tumor cell line before transplantation also results in smaller tumors. This tumor growth inhibition is associated with increased numbers of CD4+ cells, consistent with a T-cell chemoattractant activity for Mig. No change in vascularization, as indicated by CD31+ cells, was observed in either Mig or IL-10-transfected tumors. Thus, an antiangiogenic activity for either cytokine could not be confirmed. Mig and IP-10 are critical to the therapeutic response resulting from high levels of IL-10, and, furthermore, Mig as a single agent also has tumor-inhibitory activity in a model of breast cancer.


Asunto(s)
Adenocarcinoma/inmunología , Quimiocinas CXC/inmunología , Péptidos y Proteínas de Señalización Intercelular , Interleucina-10/farmacología , Neoplasias Mamarias Experimentales/inmunología , Adenocarcinoma/genética , Adenocarcinoma/terapia , Secuencia de Aminoácidos , Animales , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Inmunoterapia , Interleucina-10/genética , Interleucina-10/inmunología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Transducción Genética , Transfección
10.
J Immunother (1991) ; 24(2): 138-143, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11449070

RESUMEN

SUMMARY: Several laboratories have reported marked tumor inhibition when the cytokine interleukin-10 (IL-10) is overexpressed as a transgene in a variety of tumor cells. To identify critical effector molecules, we compared the expression of the chemokine crg-2, the murine homolog of human inducible protein 10 (human IP-10) in murine mammary tumors derived from the transplantation of six IL-10 expressing clones of tumor cell line 66.1, parental 66.1, or 66-neo-cells. We observed increased levels of IP-10 mRNA in all IL-10-expressing tumors examined in comparison to 66-neo. IP-10 mRNA was not detected in parental 66.1 tumors. The closely related chemokine Mig (monokine induced by interferon-gamma [IFN-gamma]) was also induced in all IL-10-expressing tumors. Studies of cultured tumor cells in vitro show that mammary epithelial tumor cells, in the absence of host elements, can express IP-10 and Mig in response to induction with either lipopolysaccharide (LPS) or IFN-gamma alone. The combination of LPS plus IFN-gamma resulted in even greater induction of IP-10 RNA. The kinetics of induction differ somewhat for the two chemokines, with IP-10 showing slower induction and less rapid decline. Because both Mig and IP-10 are chemotactic for tumor-infiltrating lymphocytes, we examined the presence of CD4+ and CD8+ lymphocytes in these tumors. Consistent with the upregulation of Mig and IP-10, we saw significantly increased numbers of CD8+ cells and a lesser increase in CD4+ cells in tumors with elevated levels of both chemokines.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...