Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 125(2): 265-276, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33981016

RESUMEN

BACKGROUND: Anti-microtubule agents are widely used to treat ovarian cancers, but the efficacy is often compromised by drug resistance. We investigated co-targeting the actin/tropomyosin cytoskeleton and microtubules to increase treatment efficacy in ovarian cancers and potentially overcome resistance. METHODS: The presence of tropomyosin-3.1 (Tpm3.1) was examined in clinical specimens from ovarian cancer patients using immunohistochemistry. Combinatorial effects of an anti-Tpm3.1 compound, ATM-3507, with vinorelbine and paclitaxel were evaluated in ovarian cancer cells via MTS and apoptosis assays. The mechanisms of action were established using live- and fixed-cell imaging and protein analysis. RESULTS: Tpm3.1 is overexpressed in 97% of tumour tissues (558 of 577) representing all histotypes of epithelial ovarian cancer. ATM-3507 displayed synergy with both anti-microtubule agents to reduce cell viability. Only vinorelbine synergised with ATM-3507 in causing apoptosis. ATM-3507 significantly prolonged vinorelbine-induced mitotic arrest with elevated activity of the spindle assembly checkpoint and mitotic cell death; however, ATM-3507 showed minor impact on paclitaxel-induced mitotic defects. Both combinations substantially increased post-mitotic G1 arrest with cyclin D1 and E1 downregulation and an increase of p21Cip and p27Kip. CONCLUSION: Combined targeting of Tpm3.1/actin and microtubules is a promising treatment strategy for ovarian cancer that should be further tested in clinical settings.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Cloruros/farmacología , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Tropomiosina/metabolismo , Regulación hacia Arriba , Vinorelbina/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/tratamiento farmacológico , Tropomiosina/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
2.
Eur J Cancer ; 137: 183-192, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32784118

RESUMEN

Targeted neurotrophic tropomyosin receptor kinase (TRK) inhibitors offer a highly specific therapeutic option for patients with infantile fibrosarcoma (IFS) carrying the NTRK gene translocation. International recommendations are needed to define the role of TRK inhibitors (TRKI) for infants with IFS. We analysed retrospective data for all published patients with IFS in the European Paediatric Soft tissue sarcoma Study Group and Cooperative Weichteilsarkomstudiengruppe (CWS) experience and developed a consensus strategy with the Children's Oncology Group. Therapies consisted of tumour resection and/or perioperative chemotherapy for extensive tumours. Among the 172 European patients treated, 162 were alive at the end of the follow-up. Sixty-five patients (40% of all survivors) were treated with surgery alone and 64 patients (39%) with surgery combined with chemotherapy. Radiotherapy was delivered to 3% of survivors (five patients). In addition, 28 survivors (17%) exclusively received chemotherapy. Among the 129 patients treated with surgery, 91% had conservative surgery (118 cases). Overall, nine patients died of disease, one from toxicity (6%) and 20 patients (12%) survived with major functional deficits or had mutilating surgery. Overall, conventional conservative strategies before the era of targeted therapy, even in the case of extensive tumours, demonstrate efficacy in IFS, but are associated with acute and some chronic side effects. TRKI have demonstrated very rapid responses in the vast majority of children with IFS with limited acute toxicity. With the current state of our knowledge, both conventional chemotherapy and TRKI should be regarded as options for patients with localised disease at the physician's and parent's discretion. TRKI should be considered in patients with metastatic disease, and before mutilating surgery when conventional chemotherapy fails. Outside a clinical trial, additional data are needed to resolve the lack of consensus about front-line use of conventional chemotherapy versus TRKI in patients with localised disease.


Asunto(s)
Fibrosarcoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Tropomiosina/antagonistas & inhibidores , Tropomiosina/uso terapéutico , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Inhibidores de Proteínas Quinasas/farmacología , Tropomiosina/farmacología
3.
Mol Cancer Res ; 18(7): 1074-1087, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32269073

RESUMEN

Antimicrotubule vinca alkaloids are widely used in the clinic but their toxicity is often dose limiting. Strategies that enhance their effectiveness at lower doses are needed. We show that combining vinca alkaloids with compounds that target a specific population of actin filaments containing the cancer-associated tropomyosin Tpm3.1 result in synergy against a broad range of tumor cell types. We discovered that low concentrations of vincristine alone induce supernumerary microtubule asters that form transient multi-polar spindles in early mitosis. Over time these asters can be reconstructed into functional bipolar spindles resulting in cell division and survival. These microtubule asters are organized by the nuclear mitotic apparatus protein (NuMA)-dynein-dynactin complex without involvement of centrosomes. However, anti-Tpm3.1 compounds at nontoxic concentrations inhibit this rescue mechanism resulting in delayed onset of anaphase, formation of multi-polar spindles, and apoptosis during mitosis. These findings indicate that drug targeting actin filaments containing Tpm3.1 potentiates the anticancer activity of low-dose vincristine treatment. IMPLICATIONS: Simultaneously inhibiting Tpm3.1-containing actin filaments and microtubules is a promising strategy to potentiate the anticancer activity of low-dose vincristine.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Piperazinas/administración & dosificación , Tropomiosina/metabolismo , Vincristina/administración & dosificación , Células A549 , Citoesqueleto de Actina/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Células HeLa , Humanos , Neoplasias Pulmonares/metabolismo , Células MCF-7 , Ratones , Piperazinas/farmacología , Tropomiosina/antagonistas & inhibidores , Vincristina/farmacología
4.
Future Oncol ; 16(4): 61-74, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31942815

RESUMEN

Aim: To conduct a systematic review and meta-analysis feasibility of clinical, quality of life and economic evidence for neurotrophic tropomyosin-related receptor tyrosine kinases (NTRK) inhibitors in patients with NTRK gene fusion-positive tumors. Materials & methods: Databases were searched for studies on NTRK inhibitors in adult and pediatric patients. Results: 27 publications reported clinical data for seven interventions. Efficacy/safety data were available for two interventions only. Four trials each reported data for larotrectinib and entrectinib with pooled analyses reporting objective response rates of 75% (95% CI: 61-85) and 57.4% (43.2-70.8), respectively. No publications reported economic or quality of life evidence. Conclusion: Preliminary data demonstrate that NTRK inhibitors are well tolerated and show impressive clinical benefit; corroboration of existing studies and real-world data are required.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Tropomiosina/antagonistas & inhibidores , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Manejo de la Enfermedad , Humanos , Neoplasias/mortalidad , Neoplasias/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Calidad de Vida , Resultado del Tratamiento , Tropomiosina/genética , Tropomiosina/metabolismo
5.
Sci Rep ; 9(1): 11262, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31375704

RESUMEN

Tropomyosins (Tpm) determine the functional capacity of actin filaments in an isoform-specific manner. The primary isoform in cancer cells is Tpm3.1 and compounds that target Tpm3.1 show promising results as anti-cancer agents both in vivo and in vitro. We have determined the molecular mechanism of interaction of the lead compound ATM-3507 with Tpm3.1-containing actin filaments. When present during co-polymerization of Tpm3.1 with actin, 3H-ATM-3507 is incorporated into the filaments and saturates at approximately one molecule per Tpm3.1 dimer and with an apparent binding affinity of approximately 2 µM. In contrast, 3H-ATM-3507 is poorly incorporated into preformed Tpm3.1/actin co-polymers. CD spectroscopy and thermal melts using Tpm3.1 peptides containing the C-terminus, the N-terminus, and a combination of the two forming the overlap junction at the interface of adjacent Tpm3.1 dimers, show that ATM-3507 shifts the melting temperature of the C-terminus and the overlap junction, but not the N-terminus. Molecular dynamic simulation (MDS) analysis predicts that ATM-3507 integrates into the 4-helix coiled coil overlap junction and in doing so, likely changes the lateral movement of Tpm3.1 across the actin surface resulting in an alteration of filament interactions with actin binding proteins and myosin motors, consistent with the cellular impact of ATM-3507.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Antineoplásicos/farmacología , Tropomiosina/antagonistas & inhibidores , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Dicroismo Circular , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Conformación Proteica en Hélice alfa/efectos de los fármacos , Dominios Proteicos/genética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestructura , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Relación Estructura-Actividad , Termodinámica , Tropomiosina/metabolismo , Tropomiosina/ultraestructura
6.
Cytoskeleton (Hoboken) ; 75(7): 307-317, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29752871

RESUMEN

The migration and invasion of cells through tissues in the body is facilitated by a dynamic actin cytoskeleton. The actin-associating protein, tropomyosin Tpm3.1 has emerged to play important roles in cell migration and invasion. To date, investigations have focused on single cell migration and invasion where Tpm3.1 expression is inversely associated with Rac GTPase-mediated cell invasion. While single cell and collective cell invasion have many features in common, collective invasion is additionally impacted by cell-cell adhesion, and the role of Tpm3.1 in collective invasion has not been established. In the present study we have modelled multicellular invasion using neuroblastoma spheroids embedded in 3D collagen and analysed the function of Tpm3.1 using recently established compounds that target the Tpm3.1 C-terminus. The major findings from our study reveal that combined Rac inhibition and Tpm3.1 targeting result in greater inhibition of multicellular invasion than either treatment alone. Together, the data suggest that Tpm3.1 disruption sensitises neuroblastoma cells to inhibition of Rac-mediated multicellular invasion.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores Enzimáticos/farmacología , Neuroblastoma/tratamiento farmacológico , Tropomiosina/antagonistas & inhibidores , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Actinas/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Línea Celular Tumoral , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Humanos , Proteína Proto-Oncogénica N-Myc/genética , Invasividad Neoplásica , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Tropomiosina/metabolismo , Proteínas de Unión al GTP rac/metabolismo
7.
Sci Rep ; 8(1): 4604, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29545590

RESUMEN

The development of novel small molecule inhibitors of the cancer-associated tropomyosin 3.1 (Tpm3.1) provides the ability to examine the metabolic function of specific actin filament populations. We have determined the ability of these anti-Tpm (ATM) compounds to regulate glucose metabolism in mice. Acute treatment (1 h) of wild-type (WT) mice with the compounds (TR100 and ATM1001) led to a decrease in glucose clearance due mainly to suppression of glucose-stimulated insulin secretion (GSIS) from the pancreatic islets. The impact of the drugs on GSIS was significantly less in Tpm3.1 knock out (KO) mice indicating that the drug action is on-target. Experiments in MIN6 ß-cells indicated that the inhibition of GSIS by the drugs was due to disruption to the cortical actin cytoskeleton. The impact of the drugs on insulin-stimulated glucose uptake (ISGU) was also examined in skeletal muscle ex vivo. In the absence of drug, ISGU was decreased in KO compared to WT muscle, confirming a role of Tpm3.1 in glucose uptake. Both compounds suppressed ISGU in WT muscle, but in the KO muscle there was little impact of the drugs. Collectively, this data indicates that the ATM drugs affect glucose metabolism in vivo by inhibiting Tpm3.1's function with few off-target effects.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Tropomiosina/antagonistas & inhibidores , Citoesqueleto de Actina/efectos de los fármacos , Animales , Glucosa/administración & dosificación , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Tropomiosina/fisiología
8.
Cell Physiol Biochem ; 45(2): 692-705, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29414807

RESUMEN

BACKGROUND/AIMS: Tropomyosin-2 (TPM2) plays important roles in functions of the cytoskeleton, such as cytokinesis, vesicle transport, cell proliferation, migration and apoptosis,and these functions imply that TPM2 also plays a role in cancer development. Indeed, it has been shown that TPM2 plays a critical role in some cancers. However, the role of TPM2 in breast cancer is still poorly characterized. Thus, we explored the role of TPM2 in breast cancer. METHODS: We analysed TPM2 expression and its correlation with the clinicopathological features in breast cancer. Then, we examined the influence of hypoxia on TPM2 expression and methylation status using bisulfite sequencing PCR. Furthermore, we performed TPM2-mediated migration and invasion assays in the context of hypoxia and examined changes in matrix metalloproteinase-2 (MMP2) expression. Finally, we detected the influence of TPM2 on survival and chemotherapy drug sensitivity. RESULTS: We found that TPM2 expression is down-regulated in breast cancer cells compared to that in normal breast cells. The data from TCGA supported these results. Promoter methylation of TPM2, which could be induced by hypoxia, was responsible for its low expression. Hypoxia might regulate cell invasiveness partly by TPM2 down-regulation-mediated changes of MMP2 expression. Importantly, low TPM2 expression was correlated with lymph node metastasis (P=0.031), tumour node metastasis stage (P=0.01), histological grade (P=0.037), and shorter overall survival (P=0.028). Univariate and multivariate analyses indicated that TPM2 was an independent predictor in breast cancer patients. Paclitaxel chemotherapy did not benefit patients with low TPM2 expression (P<0.0001). TPM2 knockdown significantly reduced cell sensitivity to paclitaxel. CONCLUSION: TPM2 is a potential novel tumour suppressor gene in breast cancer. TPM2 is associated with poor survival and chemoresistance to paclitaxel in breast cancer, and TPM2 may represent a promising therapeutic gene target for breast cancer patients with chemoresistance.


Asunto(s)
Neoplasias de la Mama/patología , Hipoxia de la Célula , Tropomiosina/metabolismo , Antineoplásicos Fitogénicos/uso terapéutico , Antineoplásicos Fitogénicos/toxicidad , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , ADN/aislamiento & purificación , ADN/metabolismo , Metilación de ADN , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metástasis Linfática , Células MCF-7 , Metaloproteinasa 2 de la Matriz/metabolismo , Persona de Mediana Edad , Paclitaxel/uso terapéutico , Paclitaxel/toxicidad , Pronóstico , Regiones Promotoras Genéticas , Interferencia de ARN , Tropomiosina/antagonistas & inhibidores , Tropomiosina/genética
9.
Mol Cancer Ther ; 16(8): 1555-1565, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28522589

RESUMEN

Actin filaments, with their associated tropomyosin polymers, and microtubules are dynamic cytoskeletal systems regulating numerous cell functions. While antimicrotubule drugs are well-established, antiactin drugs have been more elusive. We previously targeted actin in cancer cells by inhibiting the function of a tropomyosin isoform enriched in cancer cells, Tpm3.1, using a first-in-class compound, TR100. Here, we screened over 200 other antitropomyosin analogues for anticancer and on-target activity using a series of in vitro cell-based and biochemical assays. ATM-3507 was selected as the new lead based on its ability to disable Tpm3.1-containing filaments, its cytotoxicity potency, and more favorable drug-like characteristics. We tested ATM-3507 and TR100 alone and in combination with antimicrotubule agents against neuroblastoma models in vitro and in vivo Both ATM-3507 and TR100 showed a high degree of synergy in vitro with vinca alkaloid and taxane antimicrotubule agents. In vivo, combination-treated animals bearing human neuroblastoma xenografts treated with antitropomyosin combined with vincristine showed minimal weight loss, a significant and profound regression of tumor growth and improved survival compared with control and either drug alone. Antitropomyosin combined with vincristine resulted in G2-M phase arrest, disruption of mitotic spindle formation, and cellular apoptosis. Our data suggest that small molecules targeting the actin cytoskeleton via tropomyosin sensitize cancer cells to antimicrotubule agents and are tolerated together in vivo This combination warrants further study. Mol Cancer Ther; 16(8); 1555-65. ©2017 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Microtúbulos/metabolismo , Neoplasias/tratamiento farmacológico , Tropomiosina/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Fase G2/efectos de los fármacos , Humanos , Ratones Desnudos , Microtúbulos/efectos de los fármacos , Mitosis/efectos de los fármacos , Neoplasias/patología , Tropomiosina/metabolismo , Vincristina/farmacología
10.
Sci Rep ; 6: 19816, 2016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26804624

RESUMEN

The tropomyosin family of proteins form end-to-end polymers along the actin filament. Tumour cells rely on specific tropomyosin-containing actin filament populations for growth and survival. To dissect out the role of tropomyosin in actin filament regulation we use the small molecule TR100 directed against the C terminus of the tropomyosin isoform Tpm3.1. TR100 nullifies the effect of Tpm3.1 on actin depolymerisation but surprisingly Tpm3.1 retains the capacity to bind F-actin in a cooperative manner. In vivo analysis also confirms that, in the presence of TR100, fluorescently tagged Tpm3.1 recovers normally into stress fibers. Assembling end-to-end along the actin filament is thereby not sufficient for tropomyosin to fulfil its function. Rather, regulation of F-actin stability by tropomyosin requires fidelity of information communicated at the barbed end of the actin filament. This distinction has significant implications for perturbing tropomyosin-dependent actin filament function in the context of anti-cancer drug development.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Isoformas de Proteínas/metabolismo , Tropomiosina/metabolismo , Citoesqueleto de Actina/química , Animales , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Multimerización de Proteína/efectos de los fármacos , Conejos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Tropomiosina/antagonistas & inhibidores , Tropomiosina/química
11.
Oncol Rep ; 33(6): 2807-14, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25873252

RESUMEN

The present study was designed to investigate the relationship between tropomyosin 1 (TPM1) and radioresistance in human U251 cells. Radioresistant U251 (RR-U251) cells were established by repeated small irradiating injury. TPM1 levels in the U251 and RR-U251 cells were inhibited by transfection with TPM1-short hairpin RNA (shRNA) while overexpression was induced by treatment with pcDNA3.1­TPM1. The radiosensitivity of the U251 and RR-U251 cells and the plasmid-transfected cells was evaluated by cell viability, migration and invasion assays. Cell apoptosis was also examined in vitro. The radiosensitivity of U251 xenografts was observed by tumor growth curve after radiotherapy in an in vivo experiment. Western blotting and immunohistochemistry were used to detect the level of TPM1 in vivo. The expression of TPM1 was significantly decreased in the RR-U251 cells, which may be correlated with the radioresistance of the glioma U251 cells. In the TPM1-silenced RR-U251 and TPM1-silenced U251 cells, cell viability, migration and invasion ability were significantly increased, and the rate of cell apoptosis was decreased. Consistent with these results, in the TPM1-overexpressing U251 and RR-U251 cells, cell viability, migration and invasion abilities were markedly decreased, and increased apoptosis was noted when compared to the control group. Tumor growth of the U251 xenografts was significantly inhibited following treatment with pcDNA3.1­TPM1 combined with radiotherapy. Taken together, these results indicate that TPM1 may be one mechanism underlying radiation resistance, and TPM1 may be a potential target for overcoming the radiation resistance in glioma.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Tolerancia a Radiación/genética , Tropomiosina/genética , Apoptosis/genética , Apoptosis/efectos de la radiación , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Silenciador del Gen , Glioma/patología , Glioma/radioterapia , Humanos , Invasividad Neoplásica/genética , Tropomiosina/antagonistas & inhibidores
12.
Parasit Vectors ; 7: 313, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24996670

RESUMEN

BACKGROUND: Trichinella spiralis expresses paramyosin (Ts-Pmy) not only as a structural protein but also as an immunomodulator that inhibits host complement as a survival strategy. Previous studies demonstrated that Ts-Pmy bound to complement components C8 and C9 and inhibited the polymerization of C9 during the formation of the membrane attack complex (MAC). The C9 binding domain of Ts-Pmy was identified within 14 amino acid residues at the C-terminus of Ts-Pmy. The production of a monoclonal antibody that specifically targets the C9 binding site is necessary for further studies of Ts-Pmy function and may be used as a therapeutic agent for T. spiralis infection. METHODS: In this study, a monoclonal antibody against the complement C9 binding domain of Ts-Pmy (mAb 9G3) was produced using hybridoma technology. The binding activity of the mAb produced for recombinant or native Ts-Pmy and the blockade of Ts-Pmy binding to C9 by the mAb were assessed by Western blot analysis. The effect of the mAb on the viability of T. spiralis was observed by co-incubation of T. spiralis with mAb 9G3 in the presence of complement in vitro and by passive transfer of the mAb into naive mice following T. spiralis larval challenge. RESULTS: mAb 9G3 was successfully produced against the C9 binding domain of Ts-Pmy and bound specifically not only to recombinant Ts-Pmy but also to native Ts-Pmy expressed in different stages of T. spiralis, including adult worms, newborn larvae and muscle larvae. The binding of mAb 9G3 to Ts-Pmy efficiently blocked the binding of Ts-Pmy to human complement C9, resulting in a significant increase in the complement-mediated killing of newborn larvae in vitro and reduced infectivity of T. spiralis larvae in mice passively transferred with the mAb. CONCLUSIONS: mAb 9G3 is a specific antibody that binds to the C9 binding domain of Ts-Pmy and interferes with Ts-Pmy's complement-binding activity. Therefore, this mAb is a protective antibody that has potential as a preventive and therapeutic agent for T. spiralis infection.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Complemento C9/metabolismo , Trichinella spiralis/efectos de los fármacos , Trichinella spiralis/metabolismo , Triquinelosis/prevención & control , Tropomiosina/antagonistas & inhibidores , Animales , Sitios de Unión , Femenino , Humanos , Larva , Ratones , Ratones Endogámicos ICR , Unión Proteica , Estructura Terciaria de Proteína , Tropomiosina/química , Tropomiosina/metabolismo
13.
Expert Opin Ther Pat ; 24(7): 731-44, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24809946

RESUMEN

INTRODUCTION: Tropomyosin receptor kinases (Trks) are a family of three similar tyrosine kinases activated by peptide hormones of the neurotrophin family. The nerve growth factor antibody tanezumab has provided clinical proof of concept for inhibition of the TrkA pathway in pain. As an alternative modality, small-molecule inhibitors of the Trks have been pursued in recent years to probe the role of these neurotrophin pathways in pain, cancer and other indications. AREAS COVERED: This paper reviews the patent literature between mid-2009 and 2013, claiming inhibitors of Trk family members as the primary biological targets. Additional patents have been reviewed where Trk is not the main kinase of interest but in which high Trk potency is observed and the chemical matter is particularly noteworthy. Patents pre-dating this period have been reviewed previously. Scifinder and Google were used to find relevant patents and clinical information using Trk or Tropomyosin as the search term. EXPERT OPINION: Considerable recent progress has been made in the identification of selective pan Trk inhibitors with pharmacodynamic and pharmacokinetic properties appropriate for clinical evaluation. Inhibitors of both active and inactive conformations of the Trks as well as peripherally restricted molecules have been identified. Furthermore, TrkA-selective allosteric inhibitors have recently been disclosed, which enables the biology of this isoform to be probed. The recent identification of a TrkA gene fusion in a subset of lung cancer patients will increase further the attraction of Trk inhibition to the pharmaceutical industry.


Asunto(s)
Glicoproteínas de Membrana/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Tropomiosina/antagonistas & inhibidores , Animales , Humanos , Patentes como Asunto , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor trkB
14.
Cancer Res ; 73(16): 5169-82, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23946473

RESUMEN

The actin cytoskeleton is a potentially vulnerable property of cancer cells, yet chemotherapeutic targeting attempts have been hampered by unacceptable toxicity. In this study, we have shown that it is possible to disrupt specific actin filament populations by targeting isoforms of tropomyosin, a core component of actin filaments, that are selectively upregulated in cancers. A novel class of anti-tropomyosin compounds has been developed that preferentially disrupts the actin cytoskeleton of tumor cells, impairing both tumor cell motility and viability. Our lead compound, TR100, is effective in vitro and in vivo in reducing tumor cell growth in neuroblastoma and melanoma models. Importantly, TR100 shows no adverse impact on cardiac structure and function, which is the major side effect of current anti-actin drugs. This proof-of-principle study shows that it is possible to target specific actin filament populations fundamental to tumor cell viability based on their tropomyosin isoform composition. This improvement in specificity provides a pathway to the development of a novel class of anti-actin compounds for the potential treatment of a wide variety of cancers.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Melanoma/tratamiento farmacológico , Ratones , Células 3T3 NIH , Neoplasias/patología , Neuroblastoma/tratamiento farmacológico , Tropomiosina/antagonistas & inhibidores , Tropomiosina/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
J Cell Sci ; 123(Pt 18): 3136-45, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20736303

RESUMEN

Regulation of actin filament assembly is essential for efficient contractile activity in striated muscle. Leiomodin is an actin-binding protein and homolog of the pointed-end capping protein, tropomodulin. These proteins are structurally similar, sharing a common domain organization that includes two actin-binding sites. Leiomodin also contains a unique C-terminal extension that has a third actin-binding WH2 domain. Recently, the striated-muscle-specific isoform of leiomodin (Lmod2) was reported to be an actin nucleator in cardiomyocytes. Here, we have identified a function of Lmod2 in the regulation of thin filament lengths. We show that Lmod2 localizes to the pointed ends of thin filaments, where it competes for binding with tropomodulin-1 (Tmod1). Overexpression of Lmod2 results in loss of Tmod1 assembly and elongation of the thin filaments from their pointed ends. The Lmod2 WH2 domain is required for lengthening because its removal results in a molecule that caps the pointed ends similarly to Tmod1. Furthermore, Lmod2 transcripts are first detected in the heart after it has begun to beat, suggesting that the primary function of Lmod2 is to maintain thin filament lengths in the mature heart. Thus, Lmod2 antagonizes the function of Tmod1, and together, these molecules might fine-tune thin filament lengths.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocardio/metabolismo , Tropomiosina/antagonistas & inhibidores , Tropomiosina/metabolismo , Citoesqueleto de Actina/química , Animales , Células Cultivadas , Embrión de Pollo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Células Musculares/metabolismo , Miocardio/química , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Tropomiosina/química , Tropomiosina/genética
16.
Mol Pharmacol ; 76(1): 183-91, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19369484

RESUMEN

Although a protective role for mesalamine against colon cancer in ulcerative colitis has been shown epidemiologically, its molecular mechanism is unknown. We cloned and sequenced a novel human tropomyosin (hTM) isoform, TC22, which is an alternatively spliced variant of normal epithelial hTM isoform 5 (hTM5), identical apart from 25 C-terminal amino acids. TC22 is expressed in 100% of colorectal carcinoma but is not expressed in normal colon epithelial cells. To explore a molecular mechanism of chemoprevention, we examined the effect of mesalamine on TC22 expression using LS180 colon cancer cells. Expression of hTM5 and TC22 was investigated at the protein and gene levels by fluorescence-activated cell sorting and real-time reverse transcription-polymerase chain reaction. Small interference RNA (siRNA) against the TC22 variant were transfected into LS180 colon cancer cells, reducing protein and transcript levels by 45 to 50%. Mesalamine or sulfasalazine (2 mM), but not sulfapyridine, significantly (p < 0.02-0.006) reduced the expression of the TC22 transcript and significantly (p < 0.05 to <0.0002) reduced the expression of TC22 protein in a dose-dependent and reversible manner. Rosiglitazone, a specific peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist, similarly and significantly (p < 0.002) reduced TC22 protein expression. A polymerase chain reaction array of 84 cancer-related genes performed on TC22 siRNA-transfected cells demonstrated a significant (more than two times) change in targets involved in apoptosis, adhesion, angiogenesis, and tissue remodeling. We conclude that mesalamine, sulfasalazine, and rosiglitazone significantly reduced the cellular expression of TC22, implicating PPARgamma in this modulation. Similar suppression of TC22 by siRNA produced gene level changes on several critical carcinogenic pathways. These findings suggest a novel antineoplastic molecular effect of mesalamine.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Mesalamina/farmacología , Tropomiosina/antagonistas & inhibidores , Anticuerpos Monoclonales/inmunología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/patología , Relación Dosis-Respuesta a Droga , Humanos , PPAR gamma/fisiología , Isoformas de Proteínas , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfasalazina/farmacología
17.
Cancer Biol Ther ; 6(8): 1318-23, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17660712

RESUMEN

Overexpression and activation of TPM3-ALK tyrosine kinase fusion protein is a causal oncogenic event in the development of Anaplastic Large Cell Lymphoma and Inflammatory Myofibroblastic ALK-positive tumors. Thus, the development of ALK specific tyrosine kinase inhibitors is a current therapeutic challenge. Animal models are essential to assess, in vivo, the efficiency of ALK-oncogene inhibitors and to identify new and/or additional therapeutic targets in the ALK tumorigenesis pathway. Using the tetracycline system to allow conditional and concomitant TPM3-ALK and luciferase expression, we have developed a unique transplant model for bioluminescent TPM3-ALK-induced fibroblastic tumors in athymic nude mice. The reversible TPM3-ALK expression allowed us to demonstrate that this oncogene is essential for the tumor growth and its maintenance. In addition, we showed that this model could be used to precisely assess tumor growth inhibition upon ALK chemical inactivation. As proof of principle, we used the general tyrosine kinase inhibitor herbimycin A to inhibit ALK oncoprotein activity. As expected, herbimycin A treatment reduced tumor growth as assessed both by tumor volume measurement and bioluminescent imaging. We conclude that this transplant model for TPM3-ALK-induced tumors represents a valuable tool not only to accurately and rapidly evaluate in vivo ALK-targeted therapies but also to gain insight into the mechanism of ALK-positive tumor development.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Modelos Animales de Enfermedad , Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Ratones , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Tropomiosina/antagonistas & inhibidores , Quinasa de Linfoma Anaplásico , Animales , Antibióticos Antineoplásicos/uso terapéutico , Benzoquinonas/farmacología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Genes Reporteros , Lactamas Macrocíclicas/farmacología , Luciferasas/análisis , Luciferasas/genética , Sustancias Luminiscentes/análisis , Linfoma Anaplásico de Células Grandes/enzimología , Ratones Desnudos , Trasplante de Neoplasias , Proteínas de Fusión Oncogénica/análisis , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/análisis , Proteínas Tirosina Quinasas Receptoras , Rifabutina/análogos & derivados , Tropomiosina/análisis
18.
J Biol Chem ; 282(19): 14328-36, 2007 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-17363372

RESUMEN

MicroRNAs are small noncoding RNA molecules that control expression of target genes. Our previous studies show that mir-21 is overexpressed in tumor tissues compared with the matched normal tissues. Moreover, suppression of mir-21 by antisense oligonucleotides inhibits tumor cell growth both in vitro and in vivo. However, it remains largely unclear as to how mir-21 affects tumor growth, because our understanding of mir-21 targets is limited. In this study, we performed two-dimensional differentiation in-gel electrophoresis of tumors treated with anti-mir-21 and identified the tumor suppressor tropomyosin 1 (TPM1) as a potential mir-21 target. In agreement with this, there is a putative mir-21 binding site at the 3'-untranslated region (3'-UTR) of TPM1 variants V1 and V5. Thus, we cloned the 3'-UTR of TPM1 into a luciferase reporter and found that although mir-21 down-regulated the luciferase activity, anti-mir-21 up-regulated it. Moreover, deletion of the mir-21 binding site abolished the effect of mir-21 on the luciferase activity, suggesting that this mir-21 binding site is critical. Western blot with the cloned TPM1-V1 plus the 3'-UTR indicated that TPM1 protein level was also regulated by mir-21, whereas real-time quantitative reverse transcription-PCR revealed no difference at the mRNA level, suggesting translational regulation. Finally, overexpression of TPM1 in breast cancer MCF-7 cells suppressed anchorage-independent growth. Thus, down-regulation of TPM1 by mir-21 may explain, at least in part, why suppression of mir-21 can inhibit tumor growth, further supporting the notion that mir-21 functions as an oncogene.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , MicroARNs/fisiología , Oligonucleótidos Antisentido/farmacología , Tropomiosina/metabolismo , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Luciferasas , Ratones , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tropomiosina/antagonistas & inhibidores , Tropomiosina/genética , Ensayo de Tumor de Célula Madre
19.
Curr Cancer Drug Targets ; 4(7): 543-53, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15578912

RESUMEN

Angiogenesis is characterized by the development of new vasculature from pre-existing vessels and plays a central role in physiological processes such as embryogenesis, wound healing and female reproductive function, as well as pathophysiologic events including cancer, rheumatoid arthritis and diabetic retinopathy. The growth and metastasis of tumors is critically dependent upon angiogenesis. Although targeting angiogenesis as a therapeutic strategy has to date met with limited success in the clinic, the recent FDA approval of the anti-VEGF antibody Avastin has validated the use of anti-angiogenic therapeutic strategies for cancer treatment. We have recently identified several plasma proteins having anti-angiogenic properties, including Histidine-Proline-Rich Glycoprotein (HPRG) and activated high-molecular-weight kininogen (HKa). Both of these proteins are able to induce apoptosis in endothelial cells in vitro and can inhibit angiogenesis in vivo. Recent studies from our laboratories have also identified a novel cell-surface binding protein for HKa that mediates its anti-angiogenic activity. This protein, tropomyosin, is normally found inside the cell and is associated with the actin cytoskeleton, where it plays a critical role in stabilizing actin filaments in a variety of cell types. However, in angiogenic endothelial cells, tropomyosin appears to have extracellular localization. Previous studies have also suggested the involvement of tropomyosin in the anti-angiogenic activity of endostatin, and our recent work indicates that tropomyosin may mediate the antiangiogenic activity of HPRG as well. In this review, we summarize data describing extracellular tropomyosin as a novel receptor for multiple anti-angiogenic proteins. Extracellular tropomyosin may therefore represent a previously undescribed central target for the development of anti-angiogenic therapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Líquido Extracelular/fisiología , Transducción de Señal/efectos de los fármacos , Tropomiosina/fisiología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/fisiología , Animales , Líquido Extracelular/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Transducción de Señal/fisiología , Tropomiosina/antagonistas & inhibidores
20.
Proc Natl Acad Sci U S A ; 99(19): 12224-9, 2002 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-12196635

RESUMEN

Conformationally altered proteins and protein fragments derived from the extracellular matrix and hemostatic system may function as naturally occurring angiogenesis inhibitors. One example of such a protein is cleaved high molecular weight kininogen (HKa). HKa inhibits angiogenesis by inducing apoptosis of proliferating endothelial cells, effects mediated largely by HKa domain 5. However, the mechanisms underlying the antiangiogenic activity of HKa have not been characterized, and its binding site on proliferating endothelial cells has not been defined. Here, we report that the induction of endothelial cell apoptosis by HKa, as well as the antiangiogenic activity of HKa in the chick chorioallantoic membrane, was inhibited completely by antitropomyosin monoclonal antibody TM-311. TM-311 also blocked the high-affinity Zn2+-dependent binding of HKa to both purified tropomyosin and proliferating endothelial cells. Confocal microscopic analysis of endothelial cells stained with monoclonal antibody TM-311, as well as biotin labeling of cell surface proteins on intact endothelial cells, revealed that tropomyosin exposure was enhanced on the surface of proliferating cells. These studies demonstrate that the antiangiogenic effects of HKa depend on high-affinity binding to endothelial cell tropomyosin.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Quininógeno de Alto Peso Molecular/metabolismo , Quininógeno de Alto Peso Molecular/farmacología , Tropomiosina/metabolismo , Alantoides/irrigación sanguínea , Alantoides/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Células Cultivadas , Embrión de Pollo , Corion/irrigación sanguínea , Corion/efectos de los fármacos , ADN Complementario/genética , Endotelio Vascular/citología , Humanos , Quininógeno de Alto Peso Molecular/genética , Neovascularización Fisiológica/efectos de los fármacos , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Tropomiosina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...