Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell ; 174(1): 172-186.e21, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29958106

RESUMEN

The fusion oncoprotein CBFß-SMMHC, expressed in leukemia cases with chromosome 16 inversion, drives leukemia development and maintenance by altering the activity of the transcription factor RUNX1. Here, we demonstrate that CBFß-SMMHC maintains cell viability by neutralizing RUNX1-mediated repression of MYC expression. Upon pharmacologic inhibition of the CBFß-SMMHC/RUNX1 interaction, RUNX1 shows increased binding at three MYC distal enhancers, where it represses MYC expression by mediating the replacement of the SWI/SNF complex component BRG1 with the polycomb-repressive complex component RING1B, leading to apoptosis. Combining the CBFß-SMMHC inhibitor with the BET inhibitor JQ1 eliminates inv(16) leukemia in human cells and a mouse model. Enhancer-interaction analysis indicated that the three enhancers are physically connected with the MYC promoter, and genome-editing analysis demonstrated that they are functionally implicated in deregulation of MYC expression. This study reveals a mechanism whereby CBFß-SMMHC drives leukemia maintenance and suggests that inhibitors targeting chromatin activity may prove effective in inv(16) leukemia therapy.


Asunto(s)
Apoptosis , Cromatina/metabolismo , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Apoptosis/efectos de los fármacos , Azepinas/farmacología , Azepinas/uso terapéutico , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Inversión Cromosómica/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , ADN/química , ADN/metabolismo , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Triazoles/farmacología , Triazoles/uso terapéutico
3.
Cell ; 151(7): 1474-87, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260137

RESUMEN

DNA Ligase IV is responsible for sealing of double-strand breaks (DSBs) during nonhomologous end-joining (NHEJ). Inhibiting Ligase IV could result in amassing of DSBs, thereby serving as a strategy toward treatment of cancer. Here, we identify a molecule, SCR7 that inhibits joining of DSBs in cell-free repair system. SCR7 blocks Ligase IV-mediated joining by interfering with its DNA binding but not that of T4 DNA Ligase or Ligase I. SCR7 inhibits NHEJ in a Ligase IV-dependent manner within cells, and activates the intrinsic apoptotic pathway. More importantly, SCR7 impedes tumor progression in mouse models and when coadministered with DSB-inducing therapeutic modalities enhances their sensitivity significantly. This inhibitor to target NHEJ offers a strategy toward the treatment of cancer and improvement of existing regimens.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , ADN Ligasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Pirimidinas/uso terapéutico , Bases de Schiff/uso terapéutico , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , ADN Ligasa (ATP) , ADN Ligasas/química , ADN Ligasas/genética , Modelos Animales de Enfermedad , Diseño de Fármacos , Resistencia a Antineoplásicos , Humanos , Linfocitos/efectos de los fármacos , Linfoma/tratamiento farmacológico , Linfoma/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Pirimidinas/síntesis química , Pirimidinas/química , Tolerancia a Radiación , Ratas , Bases de Schiff/síntesis química , Bases de Schiff/química , Alineación de Secuencia
4.
Mol Cell Biochem ; 426(1-2): 149-160, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27882441

RESUMEN

Pyrazole moiety represents an important category of heterocyclic compound in pharmaceutical and medicinal chemistry. The novel 1-aryl-3, 5-bis (het) aryl pyrazole derivatives were synthesized with complementary regioselectivity. The chemical structures were confirmed by IR, 1H NMR, 13C NMR, and mass spectral analysis. The chemical entities were screened in various cancer cell lines to assess their cell viability activity. Results showed that the compound 3-(1-(4-bromophenyl)-5-phenyl-1H-pyrazol-3-yl) pyridine (5d) possessed maximum cytotoxic effect against breast cancer and leukemic cells. The cytotoxicity was confirmed by live-dead cell assay and cell cycle analysis. Mitochondrial membrane potential, Annexin V-FITC staining, DNA fragmentation, Hoechst staining, and western blot assays revealed the ability of compound 5d to induce cell death by activating apoptosis in cancer cells. Thus, the present study demonstrates that compound 5d could be an attractive chemical entity for the development of small molecule inhibitors for treatment of leukemia and breast cancer.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Citotoxinas , Leucemia/tratamiento farmacológico , Pirazoles , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Muerte Celular/efectos de los fármacos , Citotoxinas/síntesis química , Citotoxinas/química , Citotoxinas/farmacología , Femenino , Humanos , Células K562 , Leucemia/metabolismo , Células MCF-7 , Espectrometría de Masas , Ratones , Pirazoles/síntesis química , Pirazoles/química , Pirazoles/farmacología
5.
Mol Cell Biochem ; 420(1-2): 141-50, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27473146

RESUMEN

Isoxazole derivatives are an important group of chemotherapeutic prototypes. In the current study, we have synthesized few isoxazole derivatives and tested them for their antiproliferative properties in cancer cell lines such as MCF7 and HeLa. The lead compound, 3-(3,4-dimethoxyphenyl)-5-(thiophen-2-yl)isoxazole (2b), showed considerable inhibition of proliferation of MCF7 and HeLa cells with the IC50 values of 19.5 and 39.2 µM, respectively. Cell cycle analyses and annexin-FITC staining in 2b-treated breast adenocarcinoma cells (MCF7) showed increased sub-G1 population and apoptosis. Furthermore, we tested the tumor inhibitory effect of 2b and estrogen receptor expression profile in DMBA-induced mammary tumors in Sprague-Dawley rats. The gross morphology of tumor studies was investigated by histopathology and ERα protein expression was evaluated by immunohistochemistry, which showed tumor regression and downregulation of ERα in tumor cells. The present results implicate that compound 2b could be used for the further derivatization for the treatment of breast cancer.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Androstenoles/farmacología , Apoptosis/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Receptor alfa de Estrógeno/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Mamarias Experimentales , Proteínas de Neoplasias/biosíntesis , Animales , Femenino , Humanos , Células MCF-7 , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratas , Ratas Sprague-Dawley
6.
Mol Cell Biochem ; 414(1-2): 137-51, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26899710

RESUMEN

Thrombocytopenia is a major hematological concern in oxidative stress-associated pathologies and chronic clinical disorders, where premature platelet destruction severely affects the normal functioning of thrombosis and hemostasis. In addition, frequent exposure of platelets to chemical entities and therapeutic drugs immensely contributes in the development of thrombocytopenia leading to huge platelet loss, which might be fatal sometimes. Till date, there are only few platelet protective molecules known to combat thrombocytopenia. Hence, small molecule therapeutics are extremely in need to relieve the burden on limited treatment strategies of thrombocytopenia. In this study, we have synthesized a series of novel 3,4,5 trisubstituted isoxazole derivatives, among which compound 4a [4-methoxy-N'-(5-methyl-3-phenylisoxazole-4-carbonyl) benzenesulfonohydrazide] was found to significantly ameliorate the oxidative stress-induced platelet apoptosis by restoring various apoptotic markers such as ROS content, cytosolic Ca(2+) levels, eIF2-α phosphorylation, mitochondrial membrane depolarization, cytochrome c release, caspase activation, PS externalization, and cytotoxicity markers. Additionally, compound 4a dose dependently inhibits collagen-induced platelet aggregation. Hence, compound 4a can be considered as a prospective molecule in the treatment regime of platelet activation and apoptosis and other clinical conditions of thrombocytopenia. Further studies might ensure the use of compound 4a as a supplementary therapeutic agent to treat, thrombosis and CVD-associated complications. Over all, the study reveals a platelet protective efficacy of novel isoxazole derivative 4a with a potential to combat oxidative stress-induced platelet apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Isoxazoles/farmacología , Agregación Plaquetaria/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Plaquetas/metabolismo , Calcio/metabolismo , Caspasas/efectos de los fármacos , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Isoxazoles/química , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , gamma-Glutamiltransferasa/antagonistas & inhibidores
7.
Bioorg Med Chem Lett ; 25(17): 3616-20, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26152430

RESUMEN

4-Thiazolidinone derivatives were synthesized using T3P®-DMSO media as a cyclodehydrating agent. All the molecules were tested for their cytotoxicity against leukemic cell lines. The compound 3-(4-bromophenyl)-2-(4-(dimethylamino)phenyl)thiazolidin-4-one (4e) with electron donating substituent at para position of phenyl ring displayed considerable cytotoxicity against Reh and Nalm6 cells with an IC50 value of 11.9 and 13.5 µM, respectively. Furthermore, the compound 4e tested for tumor regression studies induced by EAC in Swiss albino mouse. Both in vitro and in vivo results suggested significant antiproliferative activity of compound 4e in Reh cells and mouse tumor tissue treated with compound 4e showed multifocal areas of necrosis and numerous number of apoptotic cells.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Tiazolidinas/química , Tiazolidinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia/tratamiento farmacológico , Leucemia/patología , Ratones , Neoplasias/patología , Relación Estructura-Actividad , Tiazolidinas/síntesis química , Tiazolidinas/uso terapéutico
8.
Bioorg Med Chem Lett ; 24(19): 4682-4688, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25205189

RESUMEN

A series of 2,5,6-substituted imidazo[2,1-b][1,3,4]thiadiazole derivatives have been prepared and were tested for antiproliferative activity on cancer cells at the National Cancer Institute. Results showed that molecules with a benzyl group at position 2, exhibited an increase in activity for the introduction of a formyl group at the 5 position. The compound 2-benzyl-5-formyl-6-(4-bromophenyl)imidazo[2,1-b][1,3,4]thiadiazole 22 has been chosen for understanding the mechanism of action by various molecular and cellular biology studies. Results obtained from cell cycle evaluation analysis, analysis of mitochondrial membrane potential and Annexin V-FITC by flow cytometric analysis, ROS production and expression of apoptotic and DNA-repair proteins suggested that compound 22 induced cytotoxicity by activating extrinsic pathway of apoptosis, however, without affecting cell cycle progression.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Tiadiazoles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Imidazoles/síntesis química , Imidazoles/química , Estructura Molecular , Relación Estructura-Actividad , Tiadiazoles/síntesis química , Tiadiazoles/química
9.
Blood Adv ; 7(23): 7304-7318, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37756546

RESUMEN

Germ line mutations in the RUNX1 gene cause familial platelet disorder (FPD), an inherited disease associated with lifetime risk to hematopoietic malignancies (HM). Patients with FPD frequently show clonal expansion of premalignant cells preceding HM onset. Despite the extensive studies on the role of RUNX1 in hematopoiesis, its function in the premalignant bone marrow (BM) is not well-understood. Here, we characterized the hematopoietic progenitor compartments using a mouse strain carrying an FPD-associated mutation, Runx1R188Q. Immunophenotypic analysis showed an increase in the number of hematopoietic stem and progenitor cells (HSPCs) in the Runx1R188Q/+ mice. However, the comparison of Sca-1 and CD86 markers suggested that Sca-1 expression may result from systemic inflammation. Cytokine profiling confirmed the dysregulation of interferon-response cytokines in the BM. Furthermore, the expression of CD48, another inflammation-response protein, was also increased in Runx1R188Q/+ HSPCs. The DNA-damage response activity of Runx1R188Q/+ hematopoietic progenitor cells was defective in vitro, suggesting that Runx1R188Q may promote genomic instability. The differentiation of long-term repopulating HSCs was reduced in Runx1R188Q/+ recipient mice. Furthermore, we found that Runx1R188Q/+ HSPCs outcompete their wild-type counterparts in bidirectional repopulation assays, and that the genetic makeup of recipient mice did not significantly affect the clonal dynamics under this setting. Finally, we demonstrate that Runx1R188Q predisposes to HM in cooperation with somatic mutations found in FPDHM, using 3 mouse models. These studies establish a novel murine FPDHM model and demonstrate that germ line Runx1 mutations induce a premalignant phenotype marked by BM inflammation, selective expansion capacity, defective DNA-damage response, and predisposition to HM.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas , Neoplasias Hematológicas , Animales , Ratones , Humanos , Mutación de Línea Germinal , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Susceptibilidad a Enfermedades , Trastornos de las Plaquetas Sanguíneas/genética , Inflamación/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/complicaciones , ADN
10.
Otolaryngol Head Neck Surg ; 140(6): 889-93, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19467410

RESUMEN

OBJECTIVE: A clinically useful test for eustachian tube function (ETF) is still lacking. Here we plan to evaluate the mucociliary function of the ET by saccharin and methylene blue test, and compare the outcome of surgery with normal and abnormal ET functions. STUDY DESIGN: Case series with planned data collection. SETTING: Department of Otolaryngology-Head and Neck Surgery, Kasturba Medical College, Mangalore (Manipal University), a tertiary care center in South India. SUBJECTS AND METHODS: This study comprised 86 patients diagnosed with mucosal chronic otitis media in quiescent/inactive stage. All were subjected to a detailed clinical examination and investigations. Preoperative evaluation of ETF was compared with postoperative outcome of surgery, and the results were analyzed. RESULTS: The saccharin test and methylene blue dye test had a good correlation in evaluating ETF. The mean value for saccharin perception time and the clearance time for methylene blue were 17.5 and 8.1 minutes, respectively. ETF was best in anterior, worst among posterior, and intermediate in subtotal perforations. Type 1 tympanoplasty was successful in 94 percent with normal ETF and in 68 percent with partial dysfunction. CONCLUSION: The saccharin test is a simple, cost-effective, and valuable diagnostic tool to assess the mucociliary function of the ET. The outcome of middle ear surgery would be a success in normal ETF, whereas in partial dysfunction the outcome need not necessarily be a failure.


Asunto(s)
Trompa Auditiva/fisiología , Depuración Mucociliar/fisiología , Otitis Media/cirugía , Timpanoplastia/métodos , Adolescente , Adulto , Enfermedad Crónica , Endoscopía , Femenino , Humanos , Masculino , Azul de Metileno , Persona de Mediana Edad , Otitis Media/fisiopatología , Sacarina , Edulcorantes , Resultado del Tratamiento
11.
Sci Rep ; 7(1): 2533, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28566733

RESUMEN

Chemically synthesized small molecules play important role in anticancer therapy. Several chemical compounds have been reported to damage the DNA, either directly or indirectly slowing down the cancer cell progression by causing a cell cycle arrest. Direct or indirect reactive oxygen species formation causes DNA damage leading to cell cycle arrest and subsequent cell death. Therefore, identification of chemically synthesized compounds with anticancer potential is important. Here we investigate the effect of benzothiazole derivative (5g) for its ability to inhibit cell proliferation in different cancer models. Interestingly, 5g interfered with cell proliferation in both, cell lines and tumor cells leading to significant G2/M arrest. 5g treatment resulted in elevated levels of ROS and subsequently, DNA double-strand breaks (DSBs) explaining observed G2/M arrest. Consistently, we observed deregulation of many cell cycle associated proteins such as CDK1, BCL2 and their phosphorylated form, CyclinB1, CDC25c etc. Besides, 5g treatment led to decreased levels of mitochondrial membrane potential and activation of apoptosis. Interestingly, 5g administration inhibited tumor growth in mice without significant side effects. Thus, our study identifies 5g as a potent biochemical inhibitor to induce G2/M phase arrest of the cell cycle, and demonstrates its anticancer properties both ex vivo and in vivo.


Asunto(s)
Benzotiazoles/farmacología , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ratones , Neoplasias/genética , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Ear Nose Throat J ; 85(1): 44-6, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16509243

RESUMEN

Cartilaginous tumors are common in the long bones of the body and relatively rare in the head and neck. When they do occur in the head and neck, the most common site is the midface. Since the first case report by Morgan in 1842, approximately 150 cases of head and neck chondroma have been recorded in the English-language literature. In this article, the authors describe a new case in which a chondroma of the nasal bone caused an external nasal deformity in a 17-year-old boy. The lesion was excised via an external rhinoplasty approach. The authors believe that this is the first reported case of a chondroma arising from the nasal bone. The authors have made an attempt to comprehensively review the literature on this rare and controversial tumor and place special emphasis on its uncertain biologic nature. A detailed discussion of the diagnosis and management of this tumor is also included in this report.


Asunto(s)
Condroma/diagnóstico , Condroma/cirugía , Hueso Nasal/cirugía , Neoplasias Nasales/diagnóstico , Neoplasias Nasales/cirugía , Adolescente , Condroma/patología , Humanos , Cartílago Hialino , Masculino , Hueso Nasal/diagnóstico por imagen , Hueso Nasal/patología , Neoplasias Nasales/patología , Rinoplastia , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
14.
Sci Rep ; 6: 34653, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27748367

RESUMEN

Resveratrol is one of the most widely studied bioactive plant polyphenols which possesses anticancer properties. Previously we have reported synthesis, characterization and identification of a novel resveratrol analog, SS28. In the present study, we show that SS28 induced cytotoxicity in several cancer cell lines ex vivo with an IC50 value of 3-5 µM. Mechanistic evaluation of effect of SS28 in non-small cell lung cancer cell line (A549) and T-cell leukemic cell line (CEM) showed that it inhibited Tubulin polymerization during cell division to cause cell cycle arrest at G2/M phase of the cell cycle at 12-18 h time period. Immunofluorescence studies confirmed the mitotic arrest upon treatment with SS28. Besides, we show that SS28 binds to Tubulin with a dissociation constant of 0.414 ± 0.11 µM. Further, SS28 treatment resulted in loss of mitochondrial membrane potential, activation of Caspase 9 and Caspase 3, leading to PARP-1 cleavage and finally cell death via intrinsic pathway of apoptosis. Importantly, treatment with SS28 resulted in regression of tumor in mice. Hence, our study reveals the antiproliferative activity of SS28 by disrupting microtubule dynamics by binding to its cellular target Tubulin and its potential to be developed as an anticancer molecule.


Asunto(s)
Apoptosis/efectos de los fármacos , Derivados del Benceno/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Mitosis/efectos de los fármacos , Estilbenos/farmacología , Moduladores de Tubulina/farmacología , Células A549 , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/metabolismo , Antineoplásicos Fitogénicos/farmacología , Derivados del Benceno/química , Derivados del Benceno/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Polimerizacion/efectos de los fármacos , Resveratrol , Estilbenos/química , Estilbenos/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo
15.
Target Oncol ; 11(5): 655-665, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27188390

RESUMEN

BACKGROUND: Cancer is a multifactorial disease, which makes it difficult to cure. Since more than one defective cellular component is often involved during oncogenesis, combination therapy is gaining prominence in the field of cancer therapeutics. OBJECTIVE: The purpose of this study was to investigate the combinatorial effects of a novel PARP inhibitor, P10, and HDAC inhibitor, SAHA, in leukemic cells. METHODS: Combinatorial effects of P10 and SAHA were tested using propidium iodide staining in different leukemic cells. Further, flowcytometry-based assays such as calcein-AM/ethidium homodimer staining, annexin-FITC/PI staining, and JC-1 staining were carried out to elucidate the mechanism of cell death. In addition, cell-cycle analysis, immunocytochemistry studies, and western blotting analysis were conducted to check the combinatorial effect in Nalm6 cells. RESULTS: Propidium iodide staining showed that P10 in combination with SAHA induced cell death in Nalm6 cells, in which PARP expression and activity is high with a combination index of <0.2. Annexin-FITC/PI staining, JC-1 staining, and other biochemical assays revealed that P10 in combination with SAHA induced apoptosis by causing a change in mitochondrial membrane potential in >65 % cells. Importantly, combinatorial treatment induced S phase arrest in 40-45 % cells due to DNA damage and plausible replicative stress. Finally, we demonstrated that treatment with P10 led to DNA strand breaks, which were further potentiated by SAHA (p < 0.01), leading to activation of apoptosis and increased cell death in PARP-positive leukemic cells. CONCLUSIONS: Our study reveals that coadministration of PARP inhibitor with SAHA could be used as a combination therapy against leukemic cells that possess high levels of intrinsic PARP activity.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Leucemia/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Leucemia/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
16.
Sci Rep ; 6: 24049, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27068577

RESUMEN

Naturally occurring compounds are considered as attractive candidates for cancer treatment and prevention. Quercetin and ellagic acid are naturally occurring flavonoids abundantly seen in several fruits and vegetables. In the present study, we evaluate and compare antitumor efficacies of quercetin and ellagic acid in animal models and cancer cell lines in a comprehensive manner. We found that quercetin induced cytotoxicity in leukemic cells in a dose-dependent manner, while ellagic acid showed only limited toxicity. Besides leukemic cells, quercetin also induced cytotoxicity in breast cancer cells, however, its effect on normal cells was limited or none. Further, quercetin caused S phase arrest during cell cycle progression in tested cancer cells. Quercetin induced tumor regression in mice at a concentration 3-fold lower than ellagic acid. Importantly, administration of quercetin lead to ~5 fold increase in the life span in tumor bearing mice compared to that of untreated controls. Further, we found that quercetin interacts with DNA directly, and could be one of the mechanisms for inducing apoptosis in both, cancer cell lines and tumor tissues by activating the intrinsic pathway. Thus, our data suggests that quercetin can be further explored for its potential to be used in cancer therapeutics and combination therapy.


Asunto(s)
Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Quercetina/metabolismo , Animales , Antineoplásicos/administración & dosificación , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ácido Elágico/administración & dosificación , Ácido Elágico/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , Quercetina/administración & dosificación , Análisis de Supervivencia , Resultado del Tratamiento
17.
Biochem Pharmacol ; 122: 10-22, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27693384

RESUMEN

Antiapoptotic protein BCL2, serves as an excellent target for anticancer therapy owing to its increased level in cancers. Previously, we have described characterization of a novel BCL2 inhibitor, Disarib, which showed selective cytotoxicity in BCL2 'high' cancer cells and CLL patient cells. Here, we have investigated the mechanism of Disarib-induced cytotoxicity, and compared its efficacy with a well-established BCL2 inhibitor, ABT199. We show that Disarib administration caused tumor regression in mouse allograft and xenograft models, exhibited platelet sparing property and did not exhibit significant side effects. Importantly, comparison between Disarib and ABT199, revealed higher efficacy for Disarib in mouse tumor model and cancer cell lines. Disarib induced cell death by activating intrinsic apoptotic pathway. Interestingly, Disarib showed synergism with paclitaxel, suggesting its potential for combination therapy. Thus, we provide mechanistic insights into the cell death pathways induced by Disarib, report that Disarib exhibited better effect than currently used ABT199 and demonstrate its combinatorial potential with paclitaxel.


Asunto(s)
Antineoplásicos/farmacología , Indoles/farmacología , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Tiadiazoles/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Indoles/química , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Estructura Molecular , Especies Reactivas de Oxígeno , Tiadiazoles/química
18.
FEBS J ; 283(18): 3408-37, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27444341

RESUMEN

The antiapoptotic protein BCL2 is overexpressed in several cancers and contributes to prolonged cell survival and chemoresistance, lending itself as an excellent target for cancer therapy. Here, we report the design, synthesis, and characterization of Disarib, a novel BCL2 inhibitor. Disarib showed selective cytotoxicity in BCL2 high cancer cell lines, and CLL patient primary cells, as compared to BCL2 low cell lines. BCL2 knockdown in cells rendered remarkable resistance to Disarib, while sensitivity was regained upon its ectopic expression, establishing target specificity. In silico, biochemical and biophysical studies demonstrated strong affinity of Disarib to BCL2, but not to other antiapoptotic BCL2 family members viz., BCL-xL, BCL2A1 etc. Interestingly, biophysical studies showed that BH1 domain deletion mutant demonstrated ~ 67-fold reduction in BCL2-Disarib interaction, while it was only ~ 20-fold in the case of BH3 deletion mutant, suggesting predominant involvement of the BH1 domain for Disarib binding. Thus, we report identification of a novel BCL2 inhibitor with a unique mechanism of BCL2 inhibition, as opposed to the well-studied BH3 domain targeting.


Asunto(s)
Antineoplásicos/farmacología , Indoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Tiadiazoles/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Sitios de Unión , Fenómenos Biofísicos , Línea Celular Tumoral , Diseño de Fármacos , Resistencia a Antineoplásicos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Indoles/química , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Ratones , Modelos Moleculares , Estructura Molecular , Dominios Proteicos , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tiadiazoles/química , Células Tumorales Cultivadas
19.
J Clin Diagn Res ; 9(10): XC08-XC12, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26557601

RESUMEN

BACKGROUND: Considering the uprising number of Head and neck cancer in the state with limited options of medical and surgical treatment, the focus of this study involved on chemotherapy in advanced Head and neck cancers. The aim of this study was to evaluate the efficacy and toxicity of combination of Cisplatin and 5-Fluorouracil (PF) as induction chemotherapy in patients in locally advanced squamous cell cancer of head and neck. MATERIALS AND METHODS: Forty four patients with previously untreated stage III -IV advanced and inoperable cases were included in this prospective study. Induction chemotherapy consisted of 3 cycles of Cisplatin 100mg/mt(2) as infusion on day 1, 5-Fluorouracil of 750mg/mt(2) on day 2, 5-Fluorouracil of 1000mg/mt2 as infusion on day 3 in an inpatient basis. Cycles were repeated with an interval of 21 days. Patients were evaluated within a period of 3 weeks at the end of completion of third cycle of chemotherapy. Post chemotherapy local therapy was individualized based on the response, site and stage of the tumour. RESULTS: Out of 44 eligible and evaluable patients, major dominance was noted in male group constituting 68%. After induction chemotherapy 58.8% of stage III experienced stable response, & 44% had partial response. In stage IV, 44% showed a stable response and 33.3% had partial response. But in comparison to primary tumour response and nodal response, which had a significant clinical response, the overall response of malignancy with respect to stage and site specificity was clinically insignificant. Moderate adverse reaction was noted in 47.6% and 42.1% had mild reactions. Majority of patients experienced grade 3 adverse events, of which anaemia in females and leucopenia in males pre-dominated. CONCLUSION: With the use of cisplatin and 5-FU as induction chemotherapy agents in advanced and inoperable squamous cell carcinoma of head and neck, a distinct benefit was seen in stabilizing the tumour from progression. But achieving a significant complete response to the same is of faint possibility. An alternate multidrug regimen or multimodality treatment would be ideal to gain the optimum results from induction agents. Toxicity related to chemotherapy usually is transient at therapeutic doses, and can be controlled by adequate prophylactic measures.

20.
Indian J Otolaryngol Head Neck Surg ; 66(Suppl 1): 126-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24533371

RESUMEN

The study was designed to compare the oral microbiota in normal and HIV-infected individuals. The study tries to establish a significant shift in oral microflora in HIV-infected patients. Antibiotic sensitivity testing was performed to establish any rise in resistance against the antibiotics. It was a two and half year prospective study conducted in a tertiary care centre. The study group consisted of eighty subjects divided into two groups of control and HIV. The age range for this group was 9-75 years. The mean age in this group was 39.7 years. The male:female ratio was 2.75:1. Tuberculosis was the most common opportunistic infection in patients with HIV infection. The most common commensal micro organism isolated was the Viridans streptococci in 60% followed by Streptococcus pneumoniae in 23.33%. HIV Group: The most common commensal micro organism isolated was the Viridans streptococci in 42%; this was followed by the Micrococci spp. in 22% cases. S. pneumoniae was isolated in 6% of cases. The colony count for Viridans streptococci showed a heavy growth in 55.56% of cases in controls whereas the same in HIV group was 62.5%. Micrococcus spp. was isolated from 11 subjects in HIV group while it was not isolated from the controls. 50% subjects in the HIV group showed a heavy growth of Klebsiella spp. whereas controls showed only moderate and scanty growth. In patients with CD4+ T cell count less than 50 cells/µl we found a heavy colonization of the oral cavity with Micrococcus spp., Acinetobacter and Klebsiella spp. Viridans streptococcus was not isolated in any of the patients with CD4+ T cell count less than 50 cells/µl. As CD4+ T cells counts improved to 51-100 cells/µl Viridans streptococcus colonies returned and 37.5% patients showed a heavy growth. Micrococcus spp. colonies were isolated till the CD4+ T cells improved up to 300 cells/µl. At counts > 300 cells/µl the oral microbiota became comparable to that of the controls. Many of the opportunistic infections in HIV are caused by commensal bacteria which are otherwise harmless in a normal individual. Our study is unique in that such a study of the oral commensals in HIV patients has never been reported. We found an increased colonization of the oral cavity by Micrococcus spp. which is a normal commensal of the skin.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA