Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Ther ; 27(8): 1436-1451, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31138510

RESUMEN

Acute graft-versus-host disease is a frequent complication associated with allogeneic hematopoietic stem cell transplantation. Patients that become refractory to initial steroid treatment have a poor prognosis. apceth-201 consists of human allogeneic mesenchymal stromal cells, engineered by lentiviral transduction to express the protease inhibitor alpha-1 antitrypsin, to augment the anti-inflammatory potential of the mesenchymal stromal cells. We show that apceth-201 mesenchymal stromal cells efficiently suppress T cell proliferation and polarize macrophages to an anti-inflammatory M2 type, in vitro. To assess the in vivo efficacy of apceth-201, it was tested in two different mouse models of acute graft-versus-host disease. Control animals in a humanized model succumbed quickly to disease, whereas median survival was doubled in apceth-201-treated animals. The product was also tested in a graft-versus-host disease model system that closely mimics haploidentical hematopoietic stem cell transplantation, an approach that is now being evaluated for use in the clinic. Control animals succumbed quickly to disease, whereas treatment with apceth-201 resulted in long-term survival of 57% of the animals. Within 25 days after the second injection, clinical scores returned to baseline in responding animals, indicating complete resolution of graft-versus-host disease. These promising data have led to planning of a phase I study using apceth-201.


Asunto(s)
Expresión Génica , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , alfa 1-Antitripsina/genética , Animales , Quimiotaxis de Leucocito/inmunología , Citocinas/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Orden Génico , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/terapia , Xenoinjertos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ratones , Especificidad de Órganos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo , Resultado del Tratamiento , alfa 1-Antitripsina/metabolismo
2.
Proc Natl Acad Sci U S A ; 112(45): 13982-7, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26508638

RESUMEN

Here, we show CRISPR/Cas9-based targeted somatic multiplex-mutagenesis and its application for high-throughput analysis of gene function in mice. Using hepatic single guide RNA (sgRNA) delivery, we targeted large gene sets to induce hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). We observed Darwinian selection of target genes, which suppress tumorigenesis in the respective cellular/tissue context, such as Pten or Cdkn2a, and conversely found low frequency of Brca1/2 alterations, explaining mutational spectra in human ICC/HCC. Our studies show that multiplexed CRISPR/Cas9 can be used for recessive genetic screening or high-throughput cancer gene validation in mice. The analysis of CRISPR/Cas9-induced tumors provided support for a major role of chromatin modifiers in hepatobiliary tumorigenesis, including that of ARID family proteins, which have recently been reported to be mutated in ICC/HCC. We have also comprehensively characterized the frequency and size of chromosomal alterations induced by combinatorial sgRNA delivery and describe related limitations of CRISPR/Cas9 multiplexing, as well as opportunities for chromosome engineering in the context of hepatobiliary tumorigenesis. Our study describes novel approaches to model and study cancer in a high-throughput multiplexed format that will facilitate the functional annotation of cancer genomes.


Asunto(s)
Sistemas CRISPR-Cas/genética , Carcinoma Hepatocelular/genética , Modelos Animales de Enfermedad , Genómica/métodos , Ensayos Analíticos de Alto Rendimiento , Neoplasias Hepáticas/genética , Mutagénesis/genética , Animales , Secuencia de Bases , Marcación de Gen , Técnicas Histológicas , Hígado/metabolismo , Ratones , Datos de Secuencia Molecular , Selección Genética/genética
3.
J Cell Sci ; 123(Pt 21): 3817-27, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20971706

RESUMEN

CD70 is a TNF-related transmembrane molecule expressed by mature dendritic cells (DCs), which present antigens to T cells via major histocompatibility complex (MHC) molecules. In DCs, CD70 localizes with MHC class II molecules in late endosomal vesicles, known as MHC class II compartments (MIICs). MIICs are transported to the immune synapse when a DC contacts an antigen-specific CD4(+) T cell. Consequently, MHC class II and CD70 are simultaneously exposed to the T cell. Thereby, T-cell activation via the antigen receptor and CD70-mediated co-stimulation are synchronized, apparently to optimize the proliferative response. We report here that the invariant chain (Ii), a chaperone known to transport MHC class II to MIICs, performs a similar function for CD70. CD70 was found to travel by default to the plasma membrane, whereas Ii coexpression directed it to late endosomes and/or lysosomes. In cells containing the MHC class II presentation pathway, CD70 localized to MIICs. This localization relied on Ii, since transport of CD70 from the Golgi to MIICs was impeded in Ii-deficient DCs. Biophysical and biochemical studies revealed that CD70 and Ii participate in an MHC-class-II-independent complex. Thus, Ii supports transport of both MHC class II and CD70 to MIICs and thereby coordinates their delivery to CD4(+) T cells.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/metabolismo , Ligando CD27/metabolismo , Células Dendríticas/metabolismo , Endosomas/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Animales , Presentación de Antígeno , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/inmunología , Ligando CD27/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/patología , Aparato de Golgi/metabolismo , Células HeLa , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Melanoma Experimental , Ratones , Ratones Noqueados , Transporte de Proteínas/genética
4.
Traffic ; 10(10): 1543-59, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19624487

RESUMEN

SNARE proteins mediate membrane fusion in the secretory pathway of eukaryotic cells. Genetic deletion and siRNA-based knockdown have been instrumental in assigning given SNAREs to defined intracellular transport steps. However, SNARE depletion occasionally results in barely detectable phenotypes. To understand how cells cope with SNARE loss, we have knocked down several SNAREs functioning in early endosome fusion. Surprisingly, knockdown of syntaxin 13, syntaxin 6 and vti1a, alone or in combinations, did not result in measurable changes of endosomal trafficking or fusion. We found that the residual SNARE levels (typically approximately 10%) were sufficient for a substantial amount of SNARE-SNARE interactions. Conversely, in wild-type cells, most SNARE molecules were concentrated in clusters, constituting a spare pool not readily available for interactions. Additionally, the knockdown organelles exhibited enhanced docking. We conclude that SNAREs are expressed at much higher levels than needed for maintenance of organelle fusion, and that loss of SNAREs is compensated for by the co-regulation of the docking machinery.


Asunto(s)
Endosomas/metabolismo , Membranas Intracelulares/metabolismo , Fusión de Membrana , Proteínas SNARE/genética , Animales , Western Blotting , Toxina del Cólera/metabolismo , Regulación hacia Abajo , Silenciador del Gen , Inmunoprecipitación , Fusión de Membrana/genética , Microscopía Fluorescente , Células PC12 , Transporte de Proteínas , ARN Interferente Pequeño/genética , Ratas , Transfección
5.
Cells ; 9(4)2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32260097

RESUMEN

Chimeric antigen receptor (CAR) redirected T cells are efficacious in the treatment of leukemia/lymphoma, however, showed less capacities in eliminating solid tumors which is thought to be partly due to the lack of cytokine support in the tumor lesion. In order to deliver supportive cytokines, we took advantage of the inherent ability of mesenchymal stem cells (MSCs) to actively migrate to tumor sites and engineered MSCs to release both IL7 and IL12 to promote homeostatic expansion and Th1 polarization. There is a mutual interaction between engineered MSCs and CAR T cells; in presence of CAR T cell released IFN-γ and TNF-α, chronic inflammatory Th2 MSCs shifted towards a Th17/Th1 pattern with IL2 and IL15 release that mutually activated CAR T cells with extended persistence, amplification, killing and protection from activation induced cell death. MSCs releasing IL7 and IL12 were superior over non-modified MSCs in supporting the CAR T cell response and improved the anti-tumor attack in a transplant tumor model. Data demonstrate the first use of genetically modified MSCs as vehicles to deliver immuno-modulatory proteins to the tumor tissue in order to improve the efficacy of CAR T cells in the treatment of solid malignancies.


Asunto(s)
Neoplasias Colorrectales/inmunología , Interleucina-12/metabolismo , Interleucina-7/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ingeniería de Proteínas , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Animales , Antígenos/inmunología , Proliferación Celular , Supervivencia Celular , Citocinas/metabolismo , Células HEK293 , Humanos , Ratones , Trasplante de Neoplasias
6.
Clin Cancer Res ; 26(11): 2626-2639, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31988196

RESUMEN

PURPOSE: Mesenchymal stem cells (MSCs) show an inherent brain tumor tropism that can be exploited for targeted delivery of therapeutic genes to invasive glioma. We assessed whether a motile MSC-based local immunomodulation is able to overcome the immunosuppressive glioblastoma microenvironment and to induce an antitumor immune response. EXPERIMENTAL DESIGN: We genetically modified MSCs to coexpress high levels of IL12 and IL7 (MSCIL7/12, Apceth-301). Therapeutic efficacy was assessed in two immunocompetent orthotopic C57BL/6 glioma models using GL261 and CT2A. Immunomodulatory effects were assessed by multicolor flow cytometry to profile immune activation and exhaustion of tumor-infiltrating immune cells. Diversity of the tumor-specific immune response as analyzed using T-cell receptor sequencing. RESULTS: Intratumoral administration of MSCIL7/12 induced significant tumor growth inhibition and remission of established intracranial tumors, as demonstrated by MR imaging. Notably, up to 50% of treated mice survived long-term. Rechallenging of survivors confirmed long-lasting tumor immunity. Local treatment with MSCIL7/12 was well tolerated and led to a significant inversion of the CD4+/CD8+ T-cell ratio with an intricate, predominantly CD8+ effector T-cell-mediated antitumor response. T-cell receptor sequencing demonstrated an increased diversity of TILs in MSCIL7/12-treated mice, indicating a broader tumor-specific immune response with subsequent oligoclonal specification during generation of long-term immunity. CONCLUSIONS: Local MSC-based immunomodulation is able to efficiently alter the immunosuppressive microenvironment in glioblastoma. The long-lasting therapeutic effects warrant a rapid clinical translation of this concept and have led to planning of a phase I/II study of apceth-301 in recurrent glioblastoma.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunomodulación , Interleucinas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Microambiente Tumoral/inmunología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
7.
Nat Commun ; 7: 10770, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26916719

RESUMEN

Mouse transgenesis has provided fundamental insights into pancreatic cancer, but is limited by the long duration of allele/model generation. Here we show transfection-based multiplexed delivery of CRISPR/Cas9 to the pancreas of adult mice, allowing simultaneous editing of multiple gene sets in individual cells. We use the method to induce pancreatic cancer and exploit CRISPR/Cas9 mutational signatures for phylogenetic tracking of metastatic disease. Our results demonstrate that CRISPR/Cas9-multiplexing enables key applications, such as combinatorial gene-network analysis, in vivo synthetic lethality screening and chromosome engineering. Negative-selection screening in the pancreas using multiplexed-CRISPR/Cas9 confirms the vulnerability of pancreatic cells to Brca2-inactivation in a Kras-mutant context. We also demonstrate modelling of chromosomal deletions and targeted somatic engineering of inter-chromosomal translocations, offering multifaceted opportunities to study complex structural variation, a hallmark of pancreatic cancer. The low-frequency mosaic pattern of transfection-based CRISPR/Cas9 delivery faithfully recapitulates the stochastic nature of human tumorigenesis, supporting wide applicability for biological/preclinical research.


Asunto(s)
Carcinogénesis/genética , Páncreas/metabolismo , Neoplasias Pancreáticas/genética , Animales , Proteína BRCA2/genética , Sistemas CRISPR-Cas , Deleción Cromosómica , Electroporación , Ingeniería Genética/métodos , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunohistoquímica , Imagen por Resonancia Magnética , Ratones , Mutación , Neoplasias Experimentales/genética , Filogenia , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas p21(ras)/genética , Análisis de Secuencia de ADN , Transfección/métodos , Translocación Genética/genética
8.
Nat Genet ; 47(1): 47-56, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25485836

RESUMEN

Here we describe a conditional piggyBac transposition system in mice and report the discovery of large sets of new cancer genes through a pancreatic insertional mutagenesis screen. We identify Foxp1 as an oncogenic transcription factor that drives pancreatic cancer invasion and spread in a mouse model and correlates with lymph node metastasis in human patients with pancreatic cancer. The propensity of piggyBac for open chromatin also enabled genome-wide screening for cancer-relevant noncoding DNA, which pinpointed a Cdkn2a cis-regulatory region. Histologically, we observed different tumor subentities and discovered associated genetic events, including Fign insertions in hepatoid pancreatic cancer. Our studies demonstrate the power of genetic screening to discover cancer drivers that are difficult to identify by other approaches to cancer genome analysis, such as downstream targets of commonly mutated human cancer genes. These piggyBac resources are universally applicable in any tissue context and provide unique experimental access to the genetic complexity of cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Elementos Transponibles de ADN/genética , Redes Reguladoras de Genes , Mutagénesis Insercional , Neoplasias Pancreáticas/genética , Secuencia de Aminoácidos , Animales , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Genes Sintéticos , Genes p16 , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mariposas Nocturnas/genética , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/patología , ATPasas de Translocación de Protón/genética , ARN Interferente Pequeño/farmacología , Proteínas Represoras/análisis , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transgenes , Transposasas/genética , Transposasas/fisiología
9.
Microsc Res Tech ; 73(6): 606-17, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19937745

RESUMEN

In the plasma membrane, membrane proteins are frequently organized in microdomains that are stabilized both by protein-protein and protein-lipid interactions, with the membrane lipid cholesterol being instrumental for microdomain stability. However, it is unclear whether such microdomains persist during endocytotic membrane trafficking. We used stimulated emission-depletion microscopy to investigate the domain structure of the endosomes. We developed a semiautomatic method for counting the individual domains, an approach that we have validated by immunoelectron microscopy. We found that in endosomes derived from neuroendocrine PC12 cells synaptophysin and several SNARE proteins are organized in microdomains. Cholesterol depletion by methyl-beta-cyclodextrin disintegrates most of the domains. Interestingly, no change in the frequency of microdomains was observed when endosomes were fused with protein-free liposomes of similar size (in what constitutes a novel approach in modifying acutely the lipid composition of organelles), regardless of whether the membrane lipid composition of the liposomes was similar or very different from that of the endosomes. Similarly, Rab depletion from the endosome membranes left the domain structure unaffected. Furthermore, labeled exogenous protein, introduced into endosomes by liposome fusion, equilibrated with the corresponding microdomains. We conclude that synaptic membrane proteins are organized in stable but dynamic clusters within endosomes, which are likely to persist during membrane recycling.


Asunto(s)
Membrana Celular/metabolismo , Endosomas/química , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/análisis , Membranas Sinápticas/química , Animales , Química Encefálica , Microscopía/métodos , Ratas , Proteínas SNARE/análisis , Sinaptofisina/análisis
10.
Mol Biol Cell ; 19(12): 5327-37, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18843044

RESUMEN

Docking and fusion of transport vesicles constitute elementary steps in intracellular membrane traffic. While docking is thought to be initiated by Rab-effector complexes, fusion is mediated by SNARE (N-ethylmaleimide-sensitive factor [NSF] attachment receptor) proteins. However, it has been recently debated whether SNAREs also play a role in the establishment or maintenance of a stably docked state. To address this question, we have investigated the SNARE dependence of docking and fusion of early endosomes, one of the central sorting compartments in the endocytic pathway. A new, fluorescence-based in vitro assay was developed, which allowed us to investigate fusion and docking in parallel. Similar to homotypic fusion, docking of early endosomes is dependent on the presence of ATP and requires physiological temperatures. Unlike fusion, docking is insensitive to the perturbation of SNARE function by means of soluble SNARE motifs, SNARE-specific F(ab) fragments, or by a block of NSF activity. In contrast, as expected, docking is strongly reduced by interfering with the synthesis of phosphatidyl inositol (PI)-3 phosphate, with the function of Rab-GTPases, as well as with early endosomal autoantigen 1 (EEA1), an essential tethering factor. We conclude that docking of early endosomes is independent of SNARE function.


Asunto(s)
Endocitosis/fisiología , Endosomas/metabolismo , Fusión de Membrana/fisiología , Proteínas SNARE/metabolismo , Vesículas Transportadoras/metabolismo , Animales , Biomarcadores/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células PC12 , Ratas , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
11.
EMBO J ; 26(17): 3981-92, 2007 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-17717530

RESUMEN

Soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins mediate organelle fusion in the secretory pathway. Different fusion steps are catalyzed by specific sets of SNARE proteins. Here we have used the SNAREs mediating the fusion of early endosomes and exocytosis, respectively, to investigate how pairing specificity is achieved. Although both sets of SNAREs promiscuously assemble in vitro, there is no functional crosstalk. We now show that they not only colocalize to overlapping microdomains in the membrane of early endosomes of neuroendocrine cells, but also form cis-complexes promiscuously, with the proportion of the different complexes being primarily dependent on mass action. Addition of soluble SNARE molecules onto native membranes revealed preference for cognate SNAREs. Furthermore, we found that SNAREs are laterally segregated at endosome contact sites, with the exocytotic synaptobrevin being depleted. We conclude that specificity in endosome fusion is mediated by the following two synergistically operating mechanisms: (i) preference for the cognate SNARE in 'trans' interactions and (ii) lateral segregation of SNAREs, leading to relative enrichment of the cognate ones at the prospective fusion sites.


Asunto(s)
Membrana Celular/metabolismo , Proteínas SNARE/metabolismo , Animales , Endosomas/metabolismo , Exocitosis , Fusión de Membrana , Células PC12 , Proteínas Q-SNARE/metabolismo , Ratas
12.
Mol Pharmacol ; 70(6): 1892-901, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16971553

RESUMEN

The extracellular part of transmembrane segment V (TM-V) is expected to be involved in the activation process of 7TM receptors, but its role is far from clear. Here, we study the highly constitutively active CXC-chemokine receptor encoded by human herpesvirus 8 (ORF74-HHV8), in which a metal ion site was introduced at the extracellular end of TM-V by substitution of two arginines at positions V:01 and V:05 with histidines [R208H; R212H]. The metal ion site conferred high-potency inverse agonist properties (EC(50), 1.7 microM) to Zn(II) in addition to agonist and allosteric enhancing properties at concentrations >10 microM. The chemokine interaction with [R208H;R212H]-ORF74 was altered compared with wild-type ORF74-HHV8 with decreased agonist (CXCL1/GROalpha) potency (84-fold), affinity (5.8- and 136-fold in competition against agonist and inverse agonist, respectively), and binding capacity (B(max); 25-fold). Zn(II) in activating concentrations (100 microM) acted as an allosteric enhancer as it increased the B(max) (7.1-fold), the potency (9.9-fold), the affinity (1.7- and 6.1-fold in competition against agonist and inverse agonist, respectively), and the efficacy (2.5-fold) of CXCL1/GROalpha. The activating properties of Zn(II) were not due to a metal ion site between the ligand and the receptor because CXCL1/GROalpha analogs in which the putative metal-ion binding residues had been substituted-[H19A] and [H34A]-acted like wild-type CXCL1/GROalpha. Based on the complex action of Zn(II) and on the chemokine interaction for [R208H;R212H]-ORF74, we conclude that the extracellular end of TM-V is important for the activation of this CXC-chemokine receptor.


Asunto(s)
Ingeniería de Proteínas , Receptores de Superficie Celular/química , Secuencia de Aminoácidos , Secuencia de Bases , Unión Competitiva , Dicroismo Circular , Clonación Molecular , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Humanos , Datos de Secuencia Molecular , Conformación Proteica , Receptores de Superficie Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA