Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Hematol ; 96(5): E175-E179, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33617672
2.
Leukemia ; 35(6): 1586-1596, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33097838

RESUMEN

The low 5-year survival rate for patients with acute myeloid leukemia (AML), primarily caused due to disease relapse, emphasizes the need for better therapeutic strategies. Disease relapse is facilitated by leukemic stem cells (LSCs) that are resistant to standard chemotherapy and promote tumor growth. To target AML blasts and LSCs using natural killer (NK) cells, we have developed a trispecific killer engager (TriKETM) molecule containing a humanized anti-CD16 heavy chain camelid single-domain antibody (sdAb) that activates NK cells, an IL-15 molecule that drives NK-cell priming, expansion and survival, and a single-chain variable fragment (scFv) against human CLEC12A (CLEC12A TriKE). CLEC12A is a myeloid lineage antigen that is highly expressed by AML cells and LSCs, but not expressed by normal hematopoietic stem cells (HSCs), thus minimizing off-target toxicity. The CLEC12A TriKE induced robust NK-cell specific proliferation, enhanced NK-cell activation, and killing of both AML cell lines and primary patient-derived AML blasts in vitro while sparing healthy HSCs. Additionally, the CLEC12A TriKE was able to reduce tumor burden in preclinical mouse models. These findings highlight the clinical potential of the CLEC12A TriKE for the effective treatment of AML.


Asunto(s)
Inmunoterapia/métodos , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda/terapia , Receptores de IgG/inmunología , Receptores Mitogénicos/inmunología , Anticuerpos de Dominio Único/farmacología , Animales , Apoptosis , Proliferación Celular , Femenino , Proteínas Ligadas a GPI/inmunología , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , Receptores Mitogénicos/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Front Immunol ; 11: 2043, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973811

RESUMEN

Active co-delivery of tumor antigens (Ag) and α-galactosylceramide (α-GalCer), a potent agonist for invariant Natural Killer T (iNKT) cells, to cross-priming CD8α+ dendritic cells (DCs) was previously shown to promote strong anti-tumor responses in mice. Here, we designed a nanoparticle-based vaccine able to target human CD141+ (BDCA3+) DCs - the equivalent of murine CD8α+ DCs - and deliver both tumor Ag (Melan A) and α-GalCer. This nanovaccine was inoculated into humanized mice that mimic the human immune system (HIS) and possess functional iNKT cells and CD8+ T cells, called HIS-CD8/NKT mice. We found that multiple immunizations of HIS-CD8/NKT mice with the nanovaccine resulted in the activation and/or expansion of human CD141+ DCs and iNKT cells and ultimately elicited a potent Melan-A-specific CD8+ T cell response, as determined by tetramer staining and ELISpot assay. Single-cell proteomics further detailed the highly polyfunctional CD8+ T cells induced by the nanovaccine and revealed their predictive potential for vaccine potency. This finding demonstrates for the first time the unique ability of human iNKT cells to license cross-priming DCs in vivo and adds a new dimension to the current strategy of cancer vaccine development.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/inmunología , Galactosilceramidas/administración & dosificación , Trombomodulina/metabolismo , Animales , Antígenos de Neoplasias/administración & dosificación , Biomarcadores , Vacunas contra el Cáncer/inmunología , Humanos , Inmunofenotipificación , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Proteómica/métodos , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/inmunología , Análisis de la Célula Individual , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
4.
Eur J Med Chem ; 182: 111612, 2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31421631

RESUMEN

Sugar, as a carrier of human energy and an important bioinformatics molecule, plays a vital role in many important physiological processes, and can control the transmission of bioinformatics by specific recognition with biological molecules such as proteins. Herein, the mechanism of glycosylation-targeted drug delivery was described, various glycosylation methods were summarized, and the applications of glycosylation-targeted drug delivery in liver targeting, brain targeting and lung targeting were discussed. At the same time, the problems of glycosylation targeted drug delivery were suggested, and the application prospects of glycosylation targeting drug delivery were expected.


Asunto(s)
Sistemas de Liberación de Medicamentos , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores , Animales , Transportador de Glucosa de Tipo 1/metabolismo , Glicosilación , Humanos , Receptores Mitogénicos/metabolismo
5.
Cancer Immunol Res ; 7(6): 952-962, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31053598

RESUMEN

Invariant natural killer T (iNKT) cells are a subset of lymphocytes with immune regulatory activity. Their ability to bridge the innate and adaptive immune systems has been studied using the glycolipid ligand α-galactosylceramide (αGC). To better harness the immune adjuvant properties of iNKT cells to enhance priming of antigen-specific CD8+ T cells, we encapsulated both αGC and antigen in a Clec9a-targeted nanoemulsion (TNE) to deliver these molecules to cross-presenting CD8+ dendritic cells (DC). We demonstrate that, even in the absence of exogenous glycolipid, iNKT cells supported the maturation of CD8α+ DCs to drive efficient cross-priming of antigen-specific CD8+ T cells upon delivery of Clec9a/OVA-TNE. The addition of αGC to the TNE (Clec9a/OVA/αGC) further enhanced activation of iNKT cells, NK cells, CD8α+ DCs, and polyfunctional CD8+ T cells. When tested therapeutically against HPVE7-expressing TC-1 tumors, long-term tumor suppression was achieved with a single administration of Clec9a/E7 peptide/αGC TNE. Antitumor activity was correlated with the recruitment of mature DCs, NK cells, and tumor-specific effector CD8+ T cells to the tumor-draining lymph node and tumor tissue. Thus, Clec9a-TNE codelivery of CD8+ T-cell epitopes with αGC induces alternative helper signals from activated iNKT cells, elicits innate (iNKT, NK) immunity, and enhances antitumor CD8+ T-cell responses for control of solid tumors.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunidad/efectos de los fármacos , Lectinas Tipo C/antagonistas & inhibidores , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Receptores Mitogénicos/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citotoxicidad Inmunológica , Emulsiones , Galactosilceramidas/metabolismo , Inmunidad Innata , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Nanopartículas
6.
Blood Rev ; 34: 26-33, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30401586

RESUMEN

CLEC12A has recently been identified as an antigen, expressed on leukemic stem cells and leukemic blasts. Given the fact that this expression profile seems stable throughout diagnosis, treatment and relapse on leukemic blasts and leukemic stem cells, CLEC12A can be considered a highly potent and reliable marker for the detection of measurable residual disease and therefore applicable for risk stratification and prognostication in AML. Low CLEC12A expression on leukemic blasts seems to be independently associated with lower likelihood of achieving complete remission after 1 cycle of induction chemotherapy, shorter event free survival, as well as overall survival, indicating potential prognostic properties of CLEC12A expression itself. Lack of expression on the normal hematopoietic stem and progenitor cells, in contrast to CD123 and CD33, might result in less toxicity regarding cytopenias, making CLEC12A an interesting target for innovating immunotherapies, including monoclonal and bispecific antibodies, antibody-drug conjugates and CAR-T cells therapy.


Asunto(s)
Biomarcadores de Tumor , Lectinas Tipo C/genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Receptores Mitogénicos/genética , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Manejo de la Enfermedad , Regulación Leucémica de la Expresión Génica , Humanos , Inmunoterapia , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/química , Lectinas Tipo C/metabolismo , Leucemia Mieloide Aguda/mortalidad , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Pronóstico , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/química , Receptores Mitogénicos/metabolismo , Relación Estructura-Actividad
7.
Clin Cancer Res ; 25(4): 1358-1368, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29959143

RESUMEN

PURPOSE: The treatment of acute myeloid leukemia (AML) has not significantly changed in 40 years. Cytarabine- and anthracycline-based chemotherapy induction regimens (7 + 3) remain the standard of care, and most patients have poor long-term survival. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has demonstrated ADCs as a clinically validated option to enhance the effectiveness of induction therapy. We are interested in developing a next-generation ADC for AML to improve upon the initial success of Mylotarg. EXPERIMENTAL DESIGN: The expression pattern of CLL-1 and its hematopoietic potential were investigated. A novel anti-CLL-1-ADC, with a highly potent pyrrolobenzodiazepine (PBD) dimer conjugated through a self-immolative disulfide linker, was developed. The efficacy and safety profiles of this ADC were evaluated in mouse xenograft models and in cynomolgus monkeys. RESULTS: We demonstrate that CLL-1 shares similar prevalence and trafficking properties that make CD33 an excellent ADC target for AML, but lacks expression on hematopoietic stem cells that hampers current CD33-targeted ADCs. Our anti-CLL-1-ADC is highly effective at depleting tumor cells in AML xenograft models and lacks target independent toxicities at doses that depleted target monocytes and neutrophils in cynomolgus monkeys. CONCLUSIONS: Collectively, our data suggest that an anti-CLL-1-ADC has the potential to become an effective and safer treatment for AML in humans, by reducing and allowing for faster recovery from initial cytopenias than the current generation of ADCs for AML.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Inmunoconjugados/farmacología , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda/tratamiento farmacológico , Receptores Mitogénicos/inmunología , Animales , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Ratones , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Blood Adv ; 2(14): 1738-1749, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30037800

RESUMEN

The current standard of care for acute myeloid leukemia (AML) is largely ineffective with very high relapse rates and low survival rates, mostly due to the inability to eliminate a rare population of leukemic stem cells (LSCs) that initiate tumor growth and are resistant to standard chemotherapy. RNA-sequencing analysis on isolated LSCs confirmed C-type lectin domain family 12 member A (CLL1, also known as CLEC12A) to be highly expressed on LSCs but not on normal hematopoietic stem cells (HSCs) or other healthy organ tissues. Expression of CLL1 was consistent across different types of AML. We developed CLT030 (CLL1-ADC), an antibody-drug conjugate (ADC) based on a humanized anti-CLL1 antibody with 2 engineered cysteine residues linked covalently via a cleavable linker to a highly potent DNA-binding payload, thus resulting in a site-specific and homogenous ADC product. The ADC is designed to be stable in the bloodstream and to release its DNA-binding payload only after the ADC binds to CLL1-expressing tumor cells, is internalized, and the linker is cleaved in the lysosomal compartment. CLL1-ADC inhibits in vitro LSC colony formation and demonstrates robust in vivo efficacy in AML cell tumor models and tumor growth inhibition in the AML patient-derived xenograft model. CLL1-ADC demonstrated a reduced effect on differentiation of healthy normal human CD34+ cells to various lineages as observed in an in vitro colony formation assay and in an in vivo xenotransplantation model as compared with CD33-ADC. These results demonstrate that CLL1-ADC could be an effective ADC therapeutic for the treatment of AML.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunoconjugados/farmacología , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda , Proteínas de Neoplasias/inmunología , Células Madre Neoplásicas , Receptores Mitogénicos/inmunología , Animales , Femenino , Células HL-60 , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Masculino , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Células Madre Neoplásicas/patología , Receptores Mitogénicos/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Int J Mol Sci ; 18(11)2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-29077054

RESUMEN

The treatment of patients with acute myeloid leukemia (AML) with targeted immunotherapy is challenged by the heterogeneity of the disease and a lack of tumor-exclusive antigens. Conventional immunotherapy targets for AML such as CD33 and CD123 have been proposed as targets for chimeric antigen receptor (CAR)-engineered T-cells (CAR-T-cells), a therapy that has been highly successful in the treatment of B-cell leukemia and lymphoma. However, CD33 and CD123 are present on hematopoietic stem cells, and targeting with CAR-T-cells has the potential to elicit long-term myelosuppression. C-type lectin-like molecule-1 (CLL1 or CLEC12A) is a myeloid lineage antigen that is expressed by malignant cells in more than 90% of AML patients. CLL1 is not expressed by healthy Hematopoietic Stem Cells (HSCs), and is therefore a promising target for CAR-T-cell therapy. Here, we describe the development and optimization of an anti-CLL1 CAR-T-cell with potent activity on both AML cell lines and primary patient-derived AML blasts in vitro while sparing healthy HSCs. Furthermore, in a disseminated mouse xenograft model using the CLL1-positive HL60 cell line, these CAR-T-cells completely eradicated tumor, thus supporting CLL1 as a promising target for CAR-T-cells to treat AML while limiting myelosuppressive toxicity.


Asunto(s)
Lectinas Tipo C/antagonistas & inhibidores , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Mitogénicos/antagonistas & inhibidores , Proteínas Recombinantes de Fusión , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores Mitogénicos/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Oncotarget ; 8(27): 44605-44624, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28574834

RESUMEN

A key task in developing the field of personalized cancer therapy is the identification of novel molecular targets that enable treatment of cancers not susceptible to other means of specific therapy. The collagen receptor uPARAP/Endo180 is overexpressed by malignant cells in several non-epithelial cancers, notably including sarcomas, glioblastomas and subsets of acute myeloid leukemia. In contrast, in healthy adult individuals, expression is restricted to minor subsets of mesenchymal cells. Functionally, uPARAP/Endo180 is a rapidly recycling endocytic receptor that delivers its cargo directly into the endosomal-lysosomal system, thus opening a potential route of entry into receptor-positive cells. This combination of specific expression and endocytic function appears well suited for targeting of uPARAP/Endo180-positive cancers by antibody-drug conjugate (ADC) mediated drug delivery. Therefore, we utilized a specific monoclonal antibody against uPARAP/Endo180, raised through immunization of a uPARAP/Endo180 knock-out mouse, which reacts with both the human and the murine receptor, to construct a uPARAP-directed ADC. This antibody was coupled to the highly toxic dolastatin derivative, monomethyl auristatin E, via a cathepsin-labile valine-citrulline linker. With this ADC, we show strong and receptor-dependent cytotoxicity in vitro in uPARAP/Endo180-positive cancer cell lines of sarcoma, glioblastoma and leukemic origin. Furthermore, we demonstrate the potency of the ADC in vivo in a xenograft mouse model with human uPARAP/Endo180-positive leukemic cells, obtaining a complete cure of all tested mice following intravenous ADC treatment with no sign of adverse effects. Our study identifies uPARAP/Endo180 as a promising target for novel therapy against several highly malignant cancer types.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunoconjugados/farmacología , Lectinas de Unión a Manosa/antagonistas & inhibidores , Glicoproteínas de Membrana/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Supervivencia Celular , Modelos Animales de Enfermedad , Endocitosis , Expresión Génica , Humanos , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Leucemia/mortalidad , Leucemia/patología , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Terapia Molecular Dirigida , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Mitogénicos/genética , Receptores Mitogénicos/metabolismo , Sarcoma/tratamiento farmacológico , Sarcoma/metabolismo , Sarcoma/mortalidad , Sarcoma/patología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Sci Rep ; 7(1): 2707, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28578388

RESUMEN

The mechanism of dendritic cells (DCs) recruitment across the blood brain barrier (BBB) during neuroinflammation has been the least explored amongst all leukocytes. For cells of myeloid origin, while integrins function at the level of adhesion, the importance of lectins remains unknown. Here, we identified functions of one C-type lectin receptor, CLEC12A, in facilitating DC binding and transmigration across the BBB in response to CCL2 chemotaxis. To test function of CLEC12A in an animal model of multiple sclerosis (MS), we administered blocking antibody to CLEC12A that significantly ameliorated disease scores in MOG35-55-induced progressive, as well as PLP138-151-induced relapsing-remitting experimental autoimmune encephalomyelitis (EAE) mice. The decline in both progression and relapse of EAE occurred as a result of reduced demyelination and myeloid cell infiltration into the CNS tissue. DC numbers were restored in the spleen of C57BL/6 and peripheral blood of SJL/J mice along with a decreased TH17 phenotype within CD4+ T-cells. The effects of CLEC12A blocking were further validated using CLEC12A knockout (KO) animals wherein EAE disease induction was delayed and reduced disease severity was observed. These studies reveal the utility of a DC-specific mechanism in designing new therapeutics for MS.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunidad/efectos de los fármacos , Lectinas Tipo C/antagonistas & inhibidores , Células Mieloides/inmunología , Células Mieloides/metabolismo , Receptores Mitogénicos/antagonistas & inhibidores , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Quimiocina CCL2/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Endotelio Vascular/metabolismo , Inmunidad/genética , Lectinas/genética , Lectinas/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Células Mieloides/efectos de los fármacos , Fenotipo , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores Mitogénicos/genética , Recurrencia , Índice de Severidad de la Enfermedad , Transducción de Señal , Migración Transendotelial y Transepitelial
12.
J Mol Cell Cardiol ; 90: 59-69, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26674152

RESUMEN

Delineation of mechanisms underlying the regulation of fibrosis-related genes in the heart is an important clinical goal as cardiac fibrosis is a major cause of myocardial dysfunction. This study probed the regulation of Discoidin Domain Receptor 2 (DDR2) gene expression and the regulatory links between Angiotensin II, DDR2 and collagen in Angiotensin II-stimulated cardiac fibroblasts. Real-time PCR and western blot analyses showed that Angiotensin II enhances DDR2 mRNA and protein expression in rat cardiac fibroblasts via NADPH oxidase-dependent reactive oxygen species induction. NF-κB activation, demonstrated by gel shift assay, abolition of DDR2 expression upon NF-κB inhibition, and luciferase and chromatin immunoprecipitation assays confirmed transcriptional control of DDR2 by NF-κB in Angiotensin II-treated cells. Inhibitors of Phospholipase C and Protein kinase C prevented Angiotensin II-dependent p38 MAPK phosphorylation that in turn blocked NF-κB activation. Angiotensin II also enhanced collagen gene expression. Importantly, the stimulatory effects of Angiotensin II on DDR2 and collagen were inter-dependent as siRNA-mediated silencing of one abolished the other. Angiotensin II promoted ERK1/2 phosphorylation whose inhibition attenuated Angiotensin II-stimulation of collagen but not DDR2. Furthermore, DDR2 knockdown prevented Angiotensin II-induced ERK1/2 phosphorylation, indicating that DDR2-dependent ERK1/2 activation enhances collagen expression in cells exposed to Angiotensin II. DDR2 knockdown was also associated with compromised wound healing response to Angiotensin II. To conclude, Angiotensin II promotes NF-κB activation that up-regulates DDR2 transcription. A reciprocal regulatory relationship between DDR2 and collagen, involving cross-talk between the GPCR and RTK pathways, is central to Angiotensin II-induced increase in collagen expression in cardiac fibroblasts.


Asunto(s)
Angiotensina II/farmacología , Colágeno Tipo I/genética , Fibroblastos/efectos de los fármacos , Miocardio/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/genética , Angiotensina II/genética , Angiotensina II/metabolismo , Animales , Colágeno Tipo I/antagonistas & inhibidores , Colágeno Tipo I/metabolismo , Receptores con Dominio Discoidina , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocardio/citología , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Cultivo Primario de Células , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/metabolismo , Transducción de Señal , Transcripción Genética , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Biol Chem ; 291(9): 4343-55, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26702058

RESUMEN

A bacterial collagen-like protein Scl2 has been developed as a recombinant collagen model system to host human collagen ligand-binding sequences, with the goal of generating biomaterials with selective collagen bioactivities. Defined binding sites in human collagen for integrins, fibronectin, heparin, and MMP-1 have been introduced into the triple-helical domain of the bacterial collagen and led to the expected biological activities. The modular insertion of activities is extended here to the discoidin domain receptors (DDRs), which are collagen-activated receptor tyrosine kinases. Insertion of the DDR-binding sequence from human collagen III into bacterial collagen led to specific receptor binding. However, even at the highest testable concentrations, the construct was unable to stimulate DDR autophosphorylation. The recombinant collagen expressed in Escherichia coli does not contain hydroxyproline (Hyp), and complementary synthetic peptide studies showed that replacement of Hyp by Pro at the critical Gly-Val-Met-Gly-Phe-Hyp position decreased the DDR-binding affinity and consequently required a higher concentration for the induction of receptor activation. The ability of the recombinant bacterial collagen to bind the DDRs without inducing kinase activation suggested it could interfere with the interactions between animal collagen and the DDRs, and such an inhibitory role was confirmed in vitro and with a cell migration assay. This study illustrates that recombinant collagen can complement synthetic peptides in investigating structure-activity relationships, and this system has the potential for the introduction or inhibition of specific biological activities.


Asunto(s)
Proteínas Bacterianas/metabolismo , Colágeno Tipo III/metabolismo , Colágeno/metabolismo , Megacariocitos/metabolismo , Modelos Moleculares , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Movimiento Celular , Células Cultivadas , Colágeno/química , Colágeno/genética , Colágeno Tipo III/química , Colágeno Tipo III/genética , Receptores con Dominio Discoidina , Sangre Fetal/citología , Células HEK293 , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Ligandos , Megacariocitos/citología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/química , Receptores Mitogénicos/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Streptococcus pyogenes
14.
ACS Chem Biol ; 10(12): 2687-96, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26390252

RESUMEN

Despite advances in precision medicine approaches over the past decade, the majority of nonsmall cell lung cancers (NSCLCs) are refractory to treatment with targeted small molecule inhibitors. Previous work has identified mutations in the Discoidin Domain Receptor 2 (DDR2) kinase as potential therapeutic targets in NSCLCs. While DDR2 is potently targeted by several multitargeted kinase inhibitors, most notably dasatinib, toxicity has limited the clinical application of anti-DDR2 therapy. Here, we have characterized compound 1 and other tool compounds demonstrating selectivity for DDR2 and show that while these compounds inhibit DDR2 in lung cancer model systems, they display limited antiproliferative activity in DDR2 mutated cell lines as compared to dual DDR2/SRC inhibitors. We show that DDR2 and SRC are binding partners, that SRC activity is tied to DDR2 activation, and that dual inhibition of both DDR2 and SRC leads to enhanced suppression of DDR2 mutated lung cancer cell lines. These results support the further evaluation of dual SRC/DDR2 targeting in NSCLC, and we report a tool compound, compound 5, which potently inhibits both SRC and DDR2 with a distinct selectivity profile as compared to dasatinib.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores con Dominio Discoidina , Inhibidores Enzimáticos/química , Humanos , Immunoblotting , Concentración 50 Inhibidora , Estructura Molecular , Mutación , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/genética
15.
PLoS One ; 10(7): e0131947, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26158267

RESUMEN

BACKGROUND: Loss of functional allele for discoidin domain receptor 2 (Ddr2) results in impaired Leydig cell response to luteinizing hormone (LH), low testosterone production and arrested spermatogenesis in older male Ddr2slie/slie mice. However, the underlying mechanism responsible for this phenotype remains unknown. Herein, we reported for the first time that the deregulated expression of Ddr2 cognate ligand, namely collagen type I (COL1), may account for the disruption of the testicular steroidogenesis in Ddr2slie/slie mutant testes. METHODOLOGY/PRINCIPAL FINDINGS: Expression of Ddr2 increased gradually along postnatal development, whereas COL1 expression became negligible from adulthood onwards. In Ddr2slie/slie mutant testis, however, in contrast to the undetectable staining of Ddr2, COL1 expression was constantly detected, with the highest values detected during adulthood. In the experimental vasectomy model, Ddr2slie/slie mutant mice exhibited an early androgen deficiency than wild-type mice, along with the accumulation of fibrotic tissue in the interstitium. Functionally, ablation of endogenous Ddr2 resulted in a significant decrease of testosterone (T) level in TM3 cells in the presence of higher concentration of COL1 treatment. Conversely, overexpression of Ddr2 could help TM3 cells to maintain a normal testicular steroidogenesis even in the presence of high concentration of COL1. Additionally, attenuated expression of Ddr2 correlates to the deregulated level of serum T levels in human pathological testes. CONCLUSIONS: Abnormal accumulation of interstitial COL1 may be responsible for the steroidogenic dysfunction in Ddr2slie/slie mutant testes.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/metabolismo , Testículo/patología , Animales , Línea Celular , Colágeno Tipo I/metabolismo , Receptores con Dominio Discoidina , Modelos Animales de Enfermedad , Fibrosis , Humanos , Inmunohistoquímica , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Oligospermia/metabolismo , Oligospermia/patología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Radioinmunoensayo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/genética , Síndrome de Sólo Células de Sertoli/metabolismo , Síndrome de Sólo Células de Sertoli/patología , Testículo/metabolismo , Testosterona/sangre
16.
Biochem Biophys Res Commun ; 464(2): 392-5, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26067556

RESUMEN

The delivery of biologically functional peptides into mammalian cells can be a direct and effective method for cancer therapy and treatment of other diseases. Discoidin domain receptor 2 (DDR2) is a collagen-induced receptor tyrosine kinase recently identified as a novel therapeutic target in lung cancer. In this study, we report that peptides containing the functional domain of DDR2 can be efficiently delivered into lung malignant cancer cells via a gold nanoparticle-DNA aptamer conjugate (AuNP-Apt)-based system. Peptide delivery resulted in the abrogation of DDR2 activation triggered by collagen. Moreover, the peptide delivered by the AuNP-Apt system inhibited cancer cell proliferation and invasion mediated by DDR2 activation. Thus, these results suggest that peptide loaded onto AuNP-Apt conjugates can be used for the development of peptide-based biomedical applications for the treatment of DDR2-positive cancer.


Asunto(s)
Aptámeros de Nucleótidos , Carcinoma de Pulmón de Células no Pequeñas/patología , Oro/química , Neoplasias Pulmonares/patología , Nanopartículas del Metal , Péptidos/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores , Línea Celular Tumoral , Membrana Celular , Receptores con Dominio Discoidina , Humanos , Péptidos/química , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Mitogénicos/fisiología
17.
J Med Chem ; 58(8): 3287-301, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25569119

RESUMEN

Discoidin domain receptors (DDRs) are members of the transmembrane receptor tyrosine kinase (RTK) superfamily which are distinguished from others by the presence of a discoidin motif in the extracellular domain and their utilization of collagens as internal ligands. Two types of DDRs, DDR1 and DDR2, have been identified with distinct expression profiles and ligand specificities. These DDRs play important roles in the regulation of fundamental cellular process, such as proliferation, survival, differentiation, adhesion, and matrix remodeling. They have also been closely linked to a number of human diseases, including various fibrotic disorders, atherosclerosis, and cancer. As a consequence, DDRs have been considered as novel potential molecular targets for drug discovery and increasing efforts are being devoted to the identification of new small molecule inhibitors targeting the receptors. In this review, we offer a contemporary overview on the discovery of DDRs inhibitors and their potential medical application for the treatment of cancer and inflammation related disorders.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Secuencia de Aminoácidos , Animales , Receptores con Dominio Discoidina , Descubrimiento de Drogas , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Conformación Proteica , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/química , Receptores Mitogénicos/metabolismo
18.
Drug Discov Today ; 20(2): 255-61, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25284748

RESUMEN

Discoidin domain receptor (DDR) 1 and 2 are transmembrane receptors that belong to the family of receptor tyrosine kinases (RTK). Upon collagen binding, DDRs transduce cellular signaling involved in various cell functions, including cell adhesion, proliferation, differentiation, migration, and matrix homeostasis. Altered DDR function resulting from either mutations or overexpression has been implicated in several types of disease, including atherosclerosis, inflammation, cancer, and tissue fibrosis. Several established inhibitors, such as imatinib, dasatinib, and nilotinib, originally developed as Abelson murine leukemia (Abl) kinase inhibitors, have been found to inhibit DDR kinase activity. As we review here, recent discoveries of novel inhibitors and their co-crystal structure with the DDR1 kinase domain have made structure-based drug discovery for DDR1 amenable.


Asunto(s)
Descubrimiento de Drogas , Proteínas Tirosina Quinasas Receptoras/química , Receptores Mitogénicos/química , Animales , Receptores con Dominio Discoidina , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores
19.
Exp Dermatol ; 23(12): 909-15, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25346475

RESUMEN

Skin dendritic cells (DC) express C-type lectin receptors for the recognition of pathogens. Langerhans cells (LC) express the receptor Langerin/CD207, whereas DEC-205/CD205 is mainly expressed by dermal DC, but can also be detected at low levels on LC. In this study, we tested an ex vivo approach for targeting DC in situ with monoclonal antibodies (mAb) against Langerin and DEC-205. The targeting mAb was injected intradermally into human skin biopsies or added to the medium during skin explant culture. Corresponding to the expression patterns of these lectin receptors on skin DC, Langerin mAb was detected merely in LC in the epidermis and DEC-205 mainly in dermal DC in human skin explants, regardless of the application route. Migratory skin DC bound and carried targeting mAb from skin explants according to their lectin receptor expression profiles. In contrast to the very selective transport of Langerin mAb by LC, DEC-205 mAb was more widely distributed on all CD1a(+) skin DC subsets but almost absent in CD14(+) dermal DC. As effective vaccination requires the addition of adjuvant, we co-administered the toll-like receptor (TLR)-3 ligand poly I:C with the mAb. This adjuvant enhanced binding of DEC-205 mAb to all skin DC subsets, whereas Langerin targeting efficacy remained unchanged. Our findings demonstrate that LC can be preferentially targeted by Langerin mAb. In contrast, DEC-205 mAb can be bound by all CD1a(+) skin DC subsets. The efficacy of DEC-205 mAb targeting strategy can be boosted by addition of poly I:C underlining the potential of this combination for immunotherapeutical interventions.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Células de Langerhans/inmunología , Receptores Mitogénicos/antagonistas & inhibidores , Adyuvantes Inmunológicos/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Complejo Antígeno-Anticuerpo/metabolismo , Antígenos CD/inmunología , Células Cultivadas , Humanos , Inmunización , Inyecciones Intradérmicas , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/antagonistas & inhibidores , Lectinas de Unión a Manosa/inmunología , Antígenos de Histocompatibilidad Menor , Poli I-C/administración & dosificación , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/inmunología , Receptores Mitogénicos/inmunología , Receptor Toll-Like 3/metabolismo
20.
Biochim Biophys Acta ; 1842(9): 1350-63, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24787381

RESUMEN

Prostate cancer frequently metastasizes to the skeleton but the underlying mechanism remains largely undefined. Discoidin domain receptor 2 (DDR2) is a member of receptor tyrosine kinase (RTK) family and is activated by collagen binding. This study aimed to investigate the function and detailed mechanism of DDR2 in prostate cancer bone dissemination. Herein we found that DDR2 was strongly expressed in bone-metastatic prostate cancer cells and tissues compared to that in normal controls. Enhanced expression of constitutively activated DDR2 led to elevation in motility and invasiveness of prostate cancer cells, whereas knockdown of DDR2 through specific shRNA caused a dramatic repression. Knockdown of DDR2 in prostate cancer cells resulted in significant decrease in the proliferation, differentiation and function of osteoblast. Over-expression of DDR2 in prostate cancer cells resulted in notable acceleration of osteoclast differentiation and bone resorption, whereas knockdown of DDR2 exhibited the opposite effects. An intrabone injection bone metastasis animal model demonstrated that DDR2 promoted osteolytic metastasis in vivo. Molecular evidence demonstrated that DDR2 regulated the expression, secretion, and promoter activity of parathyroid hormone-related protein (PTHrP), via modulating Runx2 phosphorylation and transactivity. DDR2 was responsive to TGF-ß and involved in TGF-ß-mediated osteoclast activation and bone resorption. In addition, DDR2 facilitated prostate cancer cells adhere to type I collagen. This study reveals for the first time that DDR2 plays an essential role in prostate cancer bone metastasis. The mechanism disclosure may provide therapeutic targets for the treatment of prostate cancer.


Asunto(s)
Neoplasias Óseas/secundario , Regulación Neoplásica de la Expresión Génica , Proteína Relacionada con la Hormona Paratiroidea/genética , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/metabolismo , Animales , Western Blotting , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Adhesión Celular , Movimiento Celular , Proliferación Celular , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Receptores con Dominio Discoidina , Ensayo de Cambio de Movilidad Electroforética , Humanos , Luciferasas/metabolismo , Masculino , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...