Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Sci Transl Med ; 16(750): eadj7308, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38838131

RESUMEN

Progranulin (PGRN) haploinsufficiency is a major risk factor for frontotemporal lobar degeneration with TAR DNA-binding protein 43 (TDP-43) pathology (FTLD-GRN). Multiple therapeutic strategies are in clinical development to restore PGRN in the CNS, including gene therapy. However, a limitation of current gene therapy approaches aimed to alleviate FTLD-associated pathologies may be their inefficient brain exposure and biodistribution. We therefore developed an adeno-associated virus (AAV) targeting the liver (L) to achieve sustained peripheral expression of a transferrin receptor (TfR) binding, brain-penetrant (b) PGRN variant [AAV(L):bPGRN] in two mouse models of FTLD-GRN, namely, Grn knockout and GrnxTmem106b double knockout mice. This therapeutic strategy avoids potential safety and biodistribution issues of CNS-administered AAVs and maintains sustained concentrations of PGRN in the brain after a single dose. AAV(L):bPGRN treatment reduced several FTLD-GRN-associated pathologies including severe motor function deficits, aberrant TDP-43 phosphorylation, dysfunctional protein degradation, lipid metabolism, gliosis, and neurodegeneration in the brain. The potential translatability of our findings was tested in an in vitro model using cocultured human induced pluripotent stem cell (hiPSC)-derived microglia lacking PGRN and TMEM106B and wild-type hiPSC-derived neurons. As in mice, aberrant TDP-43, lysosomal dysfunction, and neuronal loss were ameliorated after treatment with exogenous TfR-binding protein transport vehicle fused to PGRN (PTV:PGRN). Together, our studies suggest that peripherally administered brain-penetrant PGRN replacement strategies ameliorate FTLD-GRN relevant phenotypes including TDP-43 pathology, neurodegeneration, and behavioral deficits. Our data provide preclinical proof of concept for the use of this AAV platform for treatment of FTLD-GRN and potentially other CNS disorders.


Asunto(s)
Encéfalo , Dependovirus , Modelos Animales de Enfermedad , Degeneración Lobar Frontotemporal , Ratones Noqueados , Progranulinas , Animales , Humanos , Ratones , Encéfalo/metabolismo , Encéfalo/patología , Dependovirus/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/patología , Terapia Genética , Fosforilación , Progranulinas/metabolismo , Progranulinas/genética , Receptores de Transferrina/metabolismo
2.
Cell ; 184(18): 4651-4668.e25, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34450028

RESUMEN

GRN mutations cause frontotemporal dementia (GRN-FTD) due to deficiency in progranulin (PGRN), a lysosomal and secreted protein with unclear function. Here, we found that Grn-/- mice exhibit a global deficiency in bis(monoacylglycero)phosphate (BMP), an endolysosomal phospholipid we identified as a pH-dependent PGRN interactor as well as a redox-sensitive enhancer of lysosomal proteolysis and lipolysis. Grn-/- brains also showed an age-dependent, secondary storage of glucocerebrosidase substrate glucosylsphingosine. We investigated a protein replacement strategy by engineering protein transport vehicle (PTV):PGRN-a recombinant protein linking PGRN to a modified Fc domain that binds human transferrin receptor for enhanced CNS biodistribution. PTV:PGRN rescued various Grn-/- phenotypes in primary murine macrophages and human iPSC-derived microglia, including oxidative stress, lysosomal dysfunction, and endomembrane damage. Peripherally delivered PTV:PGRN corrected levels of BMP, glucosylsphingosine, and disease pathology in Grn-/- CNS, including microgliosis, lipofuscinosis, and neuronal damage. PTV:PGRN thus represents a potential biotherapeutic for GRN-FTD.


Asunto(s)
Productos Biológicos/uso terapéutico , Encéfalo/metabolismo , Enfermedades por Almacenamiento Lisosomal/terapia , Progranulinas/uso terapéutico , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Endosomas/metabolismo , Femenino , Demencia Frontotemporal/sangre , Demencia Frontotemporal/líquido cefalorraquídeo , Gliosis/complicaciones , Gliosis/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/patología , Metabolismo de los Lípidos , Lipofuscina/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Degeneración Nerviosa/patología , Fenotipo , Progranulinas/deficiencia , Progranulinas/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Transferrina/metabolismo , Distribución Tisular
3.
Clin Cancer Res ; 27(15): 4435-4448, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34011561

RESUMEN

PURPOSE: Ovarian cancer represents a major clinical hurdle for immune checkpoint blockade (ICB), with reported low patient response rates. We found that the immune checkpoint ligand PD-L2 is robustly expressed in patient samples of ovarian cancers and other malignancies exhibiting suboptimal response to ICB but not in cancers that are ICB sensitive. Therefore, we hypothesize that PD-L2 can facilitate immune escape from ICB through incomplete blockade of the PD-1 signaling pathway. EXPERIMENTAL DESIGN: We engineered a soluble form of the PD-1 receptor (sPD-1) capable of binding and neutralizing both PD-L2 and PD-L1 with ×200 and ×10,000 folds improvement in binding affinity over wild-type PD-1 leading to superior inhibition of ligand-mediated PD-1 activities. RESULTS: Both in vitro and in vivo analyses performed in this study demonstrated that the high-affinity sPD-1 molecule is superior at blocking both PD-L1- and PD-L2-mediated immune evasion and reducing tumor growth in immune-competent murine models of ovarian cancer. CONCLUSIONS: The data presented in this study provide justification for using a dual targeting, high-affinity sPD-1 receptor as an alternative to PD-1 or PD-L1 therapeutic antibodies for achieving superior therapeutic efficacy in cancers expressing both PD-L2 and PD-L1.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Proteína 2 Ligando de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones
4.
J Clin Invest ; 127(1): 183-198, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27893463

RESUMEN

The AXL receptor and its activating ligand, growth arrest-specific 6 (GAS6), are important drivers of metastasis and therapeutic resistance in human cancers. Given the critical roles that GAS6 and AXL play in refractory disease, this signaling axis represents an attractive target for therapeutic intervention. However, the strong picomolar binding affinity between GAS6 and AXL and the promiscuity of small molecule inhibitors represent important challenges faced by current anti-AXL therapeutics. Here, we have addressed these obstacles by engineering a second-generation, high-affinity AXL decoy receptor with an apparent affinity of 93 femtomolar to GAS6. Our decoy receptor, MYD1-72, profoundly inhibited disease progression in aggressive preclinical models of human cancers and induced cell killing in leukemia cells. When directly compared with the most advanced anti-AXL small molecules in the clinic, MYD1-72 achieved superior antitumor efficacy while displaying no toxicity. Moreover, we uncovered a relationship between AXL and the cellular response to DNA damage whereby abrogation of AXL signaling leads to accumulation of the DNA-damage markers γH2AX, 53BP1, and RAD51. MYD1-72 exploited this relationship, leading to improvements upon the therapeutic index of current standard-of-care chemotherapies in preclinical models of advanced pancreatic and ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Leucemia/metabolismo , Ratones , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
5.
Nat Commun ; 7: 13898, 2016 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-28008921

RESUMEN

Increasing evidence suggests that ionizing radiation therapy (RT) in combination with checkpoint immunotherapy is highly effective in treating a subset of cancers. To better understand the limited responses to this combination we analysed the genetic, microenvironmental, and immune factors in tumours derived from a transgenic breast cancer model. We identified two tumours with similar growth characteristics but different RT responses primarily due to an antitumour immune response. The combination of RT and checkpoint immunotherapy resulted in cures in the responsive but not the unresponsive tumours. Profiling the tumours revealed that the Axl receptor tyrosine kinase is overexpressed in the unresponsive tumours, and Axl knockout resulted in slower growth and increased radiosensitivity. These changes were associated with a CD8+ T-cell response, which was improved in combination with checkpoint immunotherapy. These results suggest a novel role for Axl in suppressing antigen presentation through MHCI, and enhancing cytokine release, which promotes a suppressive myeloid microenvironment.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación , Animales , Línea Celular Tumoral , Proliferación Celular , Terapia Combinada , Citocinas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunidad , Terapia de Inmunosupresión , Inmunoterapia , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Tolerancia a Radiación/efectos de la radiación , Tirosina Quinasa del Receptor Axl
6.
Nat Chem Biol ; 10(11): 977-83, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25242553

RESUMEN

Aberrant signaling through the Axl receptor tyrosine kinase has been associated with a myriad of human diseases, most notably metastatic cancer, identifying Axl and its ligand Gas6 as important therapeutic targets. Using rational and combinatorial approaches, we engineered an Axl 'decoy receptor' that binds Gas6 with high affinity and inhibits its function, offering an alternative approach from drug discovery efforts that directly target Axl. Four mutations within this high-affinity Axl variant caused structural alterations in side chains across the Gas6-Axl binding interface, stabilizing a conformational change on Gas6. When reformatted as an Fc fusion, the engineered decoy receptor bound Gas6 with femtomolar affinity, an 80-fold improvement compared to binding of the wild-type Axl receptor, allowing effective sequestration of Gas6 and specific abrogation of Axl signaling. Moreover, increased Gas6 binding affinity was critical and correlative with the ability of decoy receptors to potently inhibit metastasis and disease progression in vivo.


Asunto(s)
Ingeniería Genética , Fragmentos Fc de Inmunoglobulinas/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/farmacología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Sitios de Unión , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/química , Ratones , Modelos Moleculares , Mutación/genética , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Relación Estructura-Actividad , Tirosina Quinasa del Receptor Axl
7.
Proc Natl Acad Sci U S A ; 111(37): 13373-8, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25187556

RESUMEN

Dysregulation of the von Hippel-Lindau/hypoxia-inducible transcription factor (HIF) signaling pathway promotes clear cell renal cell carcinoma (ccRCC) progression and metastasis. The protein kinase GAS6/AXL signaling pathway has recently been implicated as an essential mediator of metastasis and receptor tyrosine kinase crosstalk in cancer. Here we establish a molecular link between HIF stabilization and induction of AXL receptor expression in metastatic ccRCC. We found that HIF-1 and HIF-2 directly activate the expression of AXL by binding to the hypoxia-response element in the AXL proximal promoter. Importantly, genetic and therapeutic inactivation of AXL signaling in metastatic ccRCC cells reversed the invasive and metastatic phenotype in vivo. Furthermore, we define a pathway by which GAS6/AXL signaling uses lateral activation of the met proto-oncogene (MET) through SRC proto-oncogene nonreceptor tyrosine kinase to maximize cellular invasion. Clinically, AXL expression in primary tumors of ccRCC patients correlates with aggressive tumor behavior and patient lethality. These findings provide an alternative model for SRC and MET activation by growth arrest-specific 6 in ccRCC and identify AXL as a therapeutic target driving the aggressive phenotype in renal clear cell carcinoma.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Renales/secundario , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Familia-src Quinasas/metabolismo , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/secundario , Hipoxia de la Célula , Línea Celular Tumoral , Activación Enzimática , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Modelos Biológicos , Invasividad Neoplásica , Fenotipo , Proto-Oncogenes Mas , Transducción de Señal , Resultado del Tratamiento , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Tirosina Quinasa del Receptor Axl
8.
J Mol Recognit ; 24(1): 127-35, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21194123

RESUMEN

A truncated form of the Agouti-related protein (AgRP), a member of the cystine-knot family, has shown promise as a scaffold for engineering novel peptides with new molecular recognition properties. In this study, we replaced a constrained six amino acid loop in AgRP with a nine amino acid loop containing an Arg-Gly-Asp integrin recognition motif, and randomized the neighboring residues to create a library of approximately 20 million AgRP variants. We displayed the AgRP mutants as fusions on the surface of yeast and used high-throughput fluorescence-activated cell sorting (FACS) to isolate peptides that bound specifically to the platelet integrin α(IIb)ß(3), a clinically important target for the prevention and treatment of thrombosis. These AgRP peptides had equilibrium dissociation (K(D)) constants for α(IIb)ß(3) integrin ranging from 60 to 90 nM, and did not bind to α(v)ß(3), α(v)ß(5), or α(5)ß(1) integrins. Using an alternate library screening strategy, we identified AgRP peptides that bound to both α(IIb)ß(3) and α(v)ß(3) integrins with K(D) values ranging from 40 to 70 nM and 20 to 30 nM, respectively, and did not bind to α(v)ß(5) or α(5)ß(1) integrins. Unique consensus sequences were identified within both series of AgRP peptides suggesting alternative molecular recognition events that dictate different integrin binding specificities. In addition, the engineered AgRP peptides prevented platelet aggregation as well as or slightly better than the FDA-approved cyclic peptide eptifibatide. Collectively, these data demonstrate that cystine-knot peptides can be generated with high affinity and specificity to closely-related integrins, and provide insights into molecular interactions between small, structured peptide ligands and their receptors.


Asunto(s)
Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/farmacología , Plaquetas/efectos de los fármacos , Integrinas/metabolismo , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Proteína Relacionada con Agouti/química , Proteína Relacionada con Agouti/metabolismo , Secuencia de Aminoácidos , Humanos , Células K562 , Proteínas Mutantes , Biblioteca de Péptidos , Pichia/genética , Pichia/metabolismo , Inhibidores de Agregación Plaquetaria/química , Inhibidores de Agregación Plaquetaria/metabolismo , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA