Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 5346, 2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37660083

RESUMEN

Chimeric antigen receptor (CAR) T cells have transformed the treatment landscape for hematological malignancies. However, CAR T cells are less efficient against solid tumors, largely due to poor infiltration resulting from the immunosuppressive nature of the tumor microenvironment (TME). Here, we assessed the efficacy of Lewis Y antigen (LeY)-specific CAR T cells in patient-derived xenograft (PDX) models of prostate cancer. In vitro, LeY CAR T cells directly killed organoids derived from androgen receptor (AR)-positive or AR-null PDXs. In vivo, although LeY CAR T cells alone did not reduce tumor growth, a single prior dose of carboplatin reduced tumor burden. Carboplatin had a pro-inflammatory effect on the TME that facilitated early and durable CAR T cell infiltration, including an altered cancer-associated fibroblast phenotype, enhanced extracellular matrix degradation and re-oriented M1 macrophage differentiation. In a PDX less sensitive to carboplatin, CAR T cell infiltration was dampened; however, a reduction in tumor burden was still observed with increased T cell activation. These findings indicate that carboplatin improves the efficacy of CAR T cell treatment, with the extent of the response dependent on changes induced within the TME.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias de la Próstata , Masculino , Animales , Humanos , Carboplatino/farmacología , Carboplatino/uso terapéutico , Microambiente Tumoral , Linfocitos T , Neoplasias de la Próstata/tratamiento farmacológico , Modelos Animales de Enfermedad
2.
Ann Oncol ; 32(11): 1366-1380, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34375680

RESUMEN

Chimeric antigen receptor (CAR)-T-cell therapy is a promising approach for the treatment of childhood cancers, particularly high-risk tumors that fail to respond to standard therapies. CAR-T cells have been highly successful in treating some types of hematological malignancies. However, CAR-T cells targeting solid cancers have had limited success so far for multiple reasons, including their poor long-term persistence and proliferation. Evidence is emerging to show that maintaining CAR-T cells in an early, less-differentiated state in vitro results in superior persistence, proliferation, and antitumor effects in vivo. Children are ideal candidates for receiving less-differentiated CAR-T cells, because their peripheral T-cell pool primarily comprises naïve cells that could readily be harvested in large numbers to generate early-phenotype CAR-T cells. Although several studies have reported different approaches to successfully generate early CAR-T cells, there are only a few clinical trials testing these in adult patients. No trials are currently testing early CAR-T cells in children. Here, we summarize the different strategies used to maintain CAR-T cells in an early phenotypic stage and present evidence suggesting that this approach may be particularly relevant to treating childhood cancers.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia Adoptiva , Neoplasias/terapia , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T
3.
Nat Commun ; 9(1): 4805, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30442932

RESUMEN

CD8 T cells protect the liver against viral infection, but can also cause severe liver damage that may even lead to organ failure. Given the lack of mechanistic insights and specific treatment options in patients with acute fulminant hepatitis, we develop a mouse model reflecting a severe acute virus-induced CD8 T cell-mediated hepatitis. Here we show that antigen-specific CD8 T cells induce liver damage in a perforin-dependent manner, yet liver failure is not caused by effector responses targeting virus-infected hepatocytes alone. Additionally, CD8 T cell mediated elimination of cross-presenting liver sinusoidal endothelial cells causes endothelial damage that leads to a dramatically impaired sinusoidal perfusion and indirectly to hepatocyte death. With the identification of perforin-mediated killing as a critical pathophysiologic mechanism of liver failure and the protective function of a new class of perforin inhibitor, our study opens new potential therapeutic angles for fulminant viral hepatitis.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Hepatitis Viral Animal/tratamiento farmacológico , Hígado/efectos de los fármacos , Proteínas Citotóxicas Formadoras de Poros/antagonistas & inhibidores , Sustancias Protectoras/farmacología , Sulfonamidas/farmacología , Adenoviridae/genética , Adenoviridae/inmunología , Adenoviridae/patogenicidad , Animales , Anticuerpos/administración & dosificación , Antígenos CD40/antagonistas & inhibidores , Antígenos CD40/genética , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Capilares/efectos de los fármacos , Capilares/virología , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/virología , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/virología , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/virología , Humanos , Hígado/irrigación sanguínea , Hígado/patología , Hígado/virología , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/administración & dosificación , Poli I-C/administración & dosificación , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/inmunología
4.
Proc Natl Acad Sci U S A ; 115(9): E2068-E2076, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29440406

RESUMEN

Chimeric antigen receptor T (CAR-T) cells are effective serial killers with a faster off-rate from dying tumor cells than CAR-T cells binding target cells through their T cell receptor (TCR). Here we explored the functional consequences of CAR-mediated signaling using a dual-specific CAR-T cell, where the same cell was triggered via TCR (tcrCTL) or CAR (carCTL). The carCTL immune synapse lacked distinct LFA-1 adhesion rings and was less reliant on LFA to form stable conjugates with target cells. carCTL receptors associated with the synapse were found to be disrupted and formed a convoluted multifocal pattern of Lck microclusters. Both proximal and distal receptor signaling pathways were induced more rapidly and subsequently decreased more rapidly in carCTL than in tcrCTL. The functional consequence of this rapid signaling in carCTL cells included faster lytic granule recruitment to the immune synapse, correlating with faster detachment of the CTL from the target cell. This study provides a mechanism for how CAR-T cells can debulk large tumor burden quickly and may contribute to further refinement of CAR design for enhancing the quality of signaling and programming of the T cell.


Asunto(s)
Sinapsis Inmunológicas/inmunología , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Complejo CD3 , Adhesión Celular , Muerte Celular , Línea Celular Tumoral , Biología Computacional , Citocinas/metabolismo , Dineínas/química , Ligandos , Activación de Linfocitos , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Microtúbulos/metabolismo , Transducción de Señal
5.
Cell Death Differ ; 23(7): 1209-18, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26868908

RESUMEN

Vorinostat is an FDA-approved histone deacetylase inhibitor (HDACi) that has proven clinical success in some patients; however, it remains unclear why certain patients remain unresponsive to this agent and other HDACis. Constitutive STAT (signal transducer and activator of transcription) activation, overexpression of prosurvival Bcl-2 proteins and loss of HR23B have been identified as potential biomarkers of HDACi resistance; however, none have yet been used to aid the clinical utility of HDACi. Herein, we aimed to further elucidate vorinostat-resistance mechanisms through a functional genomics screen to identify novel genes that when knocked down by RNA interference (RNAi) sensitized cells to vorinostat-induced apoptosis. A synthetic lethal functional screen using a whole-genome protein-coding RNAi library was used to identify genes that when knocked down cooperated with vorinostat to induce tumor cell apoptosis in otherwise resistant cells. Through iterative screening, we identified 10 vorinostat-resistance candidate genes that sensitized specifically to vorinostat. One of these vorinostat-resistance genes was GLI1, an oncogene not previously known to regulate the activity of HDACi. Treatment of vorinostat-resistant cells with the GLI1 small-molecule inhibitor, GANT61, phenocopied the effect of GLI1 knockdown. The mechanism by which GLI1 loss of function sensitized tumor cells to vorinostat-induced apoptosis is at least in part through interactions with vorinostat to alter gene expression in a manner that favored apoptosis. Upon GLI1 knockdown and vorinostat treatment, BCL2L1 expression was repressed and overexpression of BCL2L1 inhibited GLI1-knockdown-mediated vorinostat sensitization. Taken together, we present the identification and characterization of GLI1 as a new HDACi resistance gene, providing a strong rationale for development of GLI1 inhibitors for clinical use in combination with HDACi therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/farmacología , Proteína con Dedos de Zinc GLI1/metabolismo , Acetilación/efectos de los fármacos , Antineoplásicos/farmacología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Genoma Humano , Células HCT116 , Histonas/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Interferencia de ARN , Regulación hacia Arriba/efectos de los fármacos , Vorinostat , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Proteína con Dedos de Zinc GLI1/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
7.
Eur J Drug Metab Pharmacokinet ; 40(4): 417-25, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25155444

RESUMEN

The cytolytic protein perforin is a key component of the immune response and is implicated in a number of human pathologies and therapy-induced conditions. A novel series of small molecule inhibitors of perforin function have been developed as potential immunosuppressive agents. The pharmacokinetics and metabolic stability of a series of 16 inhibitors of perforin was evaluated in male CD1 mice following intravenous administration. The compounds were well tolerated 6 h after dosing. After intravenous administration at 5 mg/kg, maximum plasma concentrations ranged from 532 ± 200 to 10,061 ± 12 ng/mL across the series. Plasma concentrations were greater than the concentrations required for in vitro inhibitory activity for 11 of the compounds. Following an initial rapid distribution phase, the elimination half-life values for the series ranged from 0.82 ± 0.25 to 4.38 ± 4.48 h. All compounds in the series were susceptible to oxidative biotransformation. Following incubations with microsomal preparations, a tenfold range in in vitro half-life was observed across the series. The data suggests that oxidative biotransformation was not singularly responsible for clearance of the compounds and no direct relationship between microsomal clearance and plasma clearance was observed. Structural modifications however, do provide some information as to the relative microsomal stability of the compounds, which may be useful for further drug development.


Asunto(s)
Inmunosupresores/farmacocinética , Perforina/antagonistas & inhibidores , Perforina/metabolismo , Animales , Evaluación Preclínica de Medicamentos/métodos , Masculino , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo
9.
Cell Death Dis ; 5: e1115, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24625974

RESUMEN

Granzymes are generally recognized for their capacity to induce various pathways of perforin-dependent target cell death. Within this serine protease family, Granzyme M (GrzM) is unique owing to its preferential expression in innate effectors such as natural killer (NK) cells. During Listeria monocytogenes infection, we observed markedly reduced secretion of macrophage inflammatory protein-1 alpha (MIP-1α) in livers of GrzM-deficient mice, which resulted in significantly impaired NK cell recruitment. Direct stimulation with IL-12 and IL-15 demonstrated that GrzM was required for maximal secretion of active MIP-1α. This effect was not due to reduced protein induction but resulted from heightened intracellular accumulation of MIP-1α, with reduced release. These results demonstrate that GrzM is a critical mediator of innate immunity that can regulate chemotactic networks and has an important role in the initiation of immune responses and pathogen control.


Asunto(s)
Quimiocina CCL3/metabolismo , Granzimas/metabolismo , Inmunidad Innata , Células Asesinas Naturales/enzimología , Listeriosis/enzimología , Animales , Células Cultivadas , Quimiotaxis de Leucocito , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Granzimas/deficiencia , Granzimas/genética , Humanos , Interleucinas/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Listeria monocytogenes/inmunología , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/inmunología , Listeriosis/microbiología , Ratones , Ratones Noqueados , Factores de Tiempo
10.
Cell Death Differ ; 21(5): 748-60, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24464226

RESUMEN

The human lymphocyte toxins granzyme B (hGrzB) and perforin cooperatively induce apoptosis of virus-infected or transformed cells: perforin pores enable entry of the serine protease hGrzB into the cytosol, where it processes Bid to selectively activate the intrinsic apoptosis pathway. Truncated Bid (tBid) induces Bax/Bak-dependent mitochondrial outer membrane permeability and the release of cytochrome c and Smac/Diablo. To identify cellular proteins that regulate perforin/hGrzB-mediated Bid cleavage and subsequent apoptosis, we performed a gene-knockdown (KD) screen using a lentiviral pool of short hairpin RNAs embedded within a miR30 backbone (shRNAmiR). We transduced HeLa cells with a lentiviral pool expressing shRNAmiRs that target 1213 genes known to be involved in cell death signaling and selected cells with acquired resistance to perforin/hGrzB-mediated apoptosis. Twenty-two shRNAmiRs were identified in the positive-selection screen including two, PCAF and ADA3, whose gene products are known to reside in the same epigenetic regulatory complexes. Small interfering (si)RNA-mediated gene-KD of PCAF or ADA3 also conferred resistance to perforin/hGrzB-mediated apoptosis providing independent validation of the screen results. Mechanistically, PCAF and ADA3 exerted their pro-apoptotic effect upstream of mitochondrial membrane permeabilization, as indicated by reduced cytochrome c release in PCAF-KD cells exposed to perforin/hGrzB. While overall levels of Bid were unaltered, perforin/hGrzB-mediated cleavage of Bid was reduced in PCAF-KD or ADA3-KD cells. We discovered that PCAF-KD or ADA3-KD resulted in reduced expression of PACS2, a protein implicated in Bid trafficking to mitochondria and importantly, targeted PACS2-KD phenocopied the effect of PCAF-KD or ADA3-KD. We conclude that PCAF and ADA3 regulate Bid processing via PACS2, to modulate the mitochondrial cell death pathway in response to hGrzB.


Asunto(s)
Granzimas/metabolismo , Mitocondrias/genética , Factores de Transcripción/genética , Factores de Transcripción p300-CBP/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/fisiología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Genómica/métodos , Granzimas/farmacología , Células HCT116 , Células HeLa , Humanos , Ratones , Mitocondrias/enzimología , Mitocondrias/metabolismo , Perforina/metabolismo , Perforina/farmacología , Transducción de Señal , Factores de Transcripción/metabolismo , Transfección , Factores de Transcripción p300-CBP/metabolismo
11.
Cell Death Differ ; 20(9): 1183-93, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744295

RESUMEN

Human and mouse granzyme (Gzm)B both induce target cell apoptosis in concert with pore-forming perforin (Pfp); however the mechanisms by which other Gzms induce non-apoptotic death remain controversial and poorly characterised. We used timelapse microscopy to document, quantitatively and in real time, the death of target cells exposed to primary natural killer (NK) cells from mice deficient in key Gzms. We found that in the vast majority of cases, NK cells from wild-type mice induced classic apoptosis. However, NK cells from syngeneic Gzm B-deficient mice induced a novel form of cell death characterised by slower kinetics and a pronounced, writhing, 'worm-like' morphology. Dying cells initially contracted but did not undergo membrane blebbing, and annexin-V staining was delayed until the onset of secondary necrosis. As it is different from any cell death process previously reported, we tentatively termed this cell death 'athetosis'. Two independent lines of evidence showed this alternate form of death was due to Gzm A: first, cell death was revealed in the absence of Gzm B, but was completely lost when the NK cells were deficient in both Gzm A and B; second, the athetotic morphology was precisely reproduced when recombinant mouse Gzm A was delivered by an otherwise innocuous dose of recombinant Pfp. Gzm A-mediated athetosis did not require caspase activation, early mitochondrial disruption or generation of reactive oxygen species, but did require an intact actin cytoskeleton and was abolished by latrunculin B and mycalolide B. This work defines an authentic role for mouse Gzm A in granule-induced cell death by cytotoxic lymphocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Granzimas/metabolismo , Células Asesinas Naturales/inmunología , Perforina/metabolismo , Citoesqueleto de Actina , Animales , Apoptosis/inmunología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Granzimas/deficiencia , Granzimas/genética , Células HeLa , Humanos , Células Asesinas Naturales/citología , Toxinas Marinas , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Oxazoles/farmacología , Tiazolidinas/farmacología , Imagen de Lapso de Tiempo
12.
Tissue Antigens ; 80(6): 477-87, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23137319

RESUMEN

Granzymes (Grz) are a family of serine proteases found in the granules of cytotoxic lymphocytes and are emerging as an important group of proteins involved in immune function and surveillance. Grz have both cytotoxic and more recently reported non-cytotoxic roles, however these functions are still subject to thorough investigation. The significance of the cytotoxic and importantly the non-cytotoxic roles of Grz will be discussed in this review, detailing accepted and controversial functions.


Asunto(s)
Granzimas/inmunología , Animales , Adhesión Celular/fisiología , Muerte Celular/fisiología , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Matriz Extracelular/fisiología , Granzimas/genética , Granzimas/fisiología , Humanos , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , Ratones , Modelos Biológicos , Perforina/fisiología , Polimorfismo Genético , Ratas , Especificidad de la Especie , Especificidad por Sustrato , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/inmunología
13.
Cell Death Dis ; 3: e344, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22764103

RESUMEN

Overexpression of Bcl-2 contributes to resistance of cancer cells to human cytotoxic lymphocytes (CL) by blocking granzyme B (GraB)-induced mitochondrial outer membrane permeabilization (MOMP). Drugs that neutralise Bcl-2 (e.g., ABT-737) may therefore be effective adjuvants for immunotherapeutic strategies that use CL to kill cancer cells. Consistent with this we found that ABT-737 effectively restored MOMP in Bcl-2 overexpressing cells treated with GraB or natural killer cells. This effect was observed even if ABT-737 was added up to 16 h after GraB, after which the cells reset their resistant phenotype. Sensitivity to ABT-737 required initial cleavage of Bid by GraB (gctBid) but did not require ongoing GraB activity once Bid had been cleaved. This gctBid remained detectable in cells that were sensitive to ABT-737, but Bax and Bak were only activated if ABT-737 was added to the cells. These studies demonstrate that GraB generates a prolonged pro-apoptotic signal that must remain active for ABT-737 to be effective. The duration of this signal is determined by the longevity of gctBid but not activation of Bax or Bak. This defines a therapeutic window in which ABT-737 and CL synergise to cause maximum death of cancer cells that are resistant to either treatment alone, which will be essential in defining optimum treatment regimens.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Granzimas/farmacología , Nitrofenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Citocromos c/metabolismo , Células HeLa , Humanos , Células Asesinas Naturales/inmunología , Mitocondrias/metabolismo , Piperazinas/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
14.
Cell Death Differ ; 19(1): 21-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22095283

RESUMEN

Within the powerful legacy left by Jurg Tschopp, we should not forget his early work that helped to elucidate the molecular pathways responsible for the clearance of virus-infected and transformed cells by cytotoxic T lymphocytes (CTL) and natural killer (NK) cells. Jurg's skilful biochemical approach formed a firm platform upon which the work of so many other biochemists, cell biologists and immunologists would come to rely. Jurg coined the shorthand term 'granzyme' to denote the individual members of a family of serine proteases sequestered in and secreted from the cytotoxic granules of CTL/NK cells. He was also one of the first to describe the lytic properties of purified perforin and to postulate the synergy of perforin and granzymes, which we now know to underpin target cell apoptosis. Jurg was a major protagonist in the debate that raged throughout the 1980's and early 1990's on the physiological relevance of the 'granule exocytosis' pathway. Ultimately, resolving this issue led Jurg and his colleagues to even greater and impactful discoveries in the broader field of apoptosis research. Jurg Tschopp ranks with other pioneers, particularly Gideon Berke, Chris Bleackley, Pierre Golstein, Pierre Henkart and Eckhard Podack for making seminal discoveries on our understanding of how the immune system eliminates dangerous cells.


Asunto(s)
Muerte Celular/genética , Granzimas/genética , Granzimas/historia , Perforina/historia , Apoptosis/inmunología , Gránulos Citoplasmáticos/metabolismo , Citotoxicidad Inmunológica/fisiología , Exocitosis/genética , Historia del Siglo XX , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Perforina/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
15.
Tissue Antigens ; 76(5): 341-51, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20860583

RESUMEN

The membrane-attack complex (MAC) of complement pathway and perforin (PF) are important tools deployed by the immune system to target pathogens. Both perforin and the C9 component of the MAC contain a common 'MACPF' domain and form pores in the cell membrane as part of their function. The MAC targets gram-negative bacteria and certain pathogenic parasites, while perforin, released by natural killer cells or cytotoxic T lymphocytes (CTLs), targets virus-infected and transformed host cells (1). Remarkably, recent structural studies show that the MACPF domain is homologous to the pore-forming portion of bacterial cholesterol-dependent cytolysins; these data have provided important insight into the mechanism of pore-forming MACPF proteins. In addition to their role in immunity, MACPF family members have been identified as animal venoms, factors required for pathogen migration across host cell membranes and factors that govern developmental processes such as embryonic patterning and neuronal guidance (2). While most MACPF proteins characterized to date either form pores or span lipid membranes, some do not (e.g. the C6 component of the MAC). A current challenge is thus to understand the role, pore forming or otherwise, of MACPF proteins in developmental biology. This review discusses structural and functional diversity of the mammalian MACPF proteins.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Perforina/química , Perforina/inmunología , Animales , Proteínas de Ciclo Celular , Complejo de Ataque a Membrana del Sistema Complemento/genética , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/inmunología , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/inmunología , Humanos , Inmunidad Celular , Inmunidad Humoral , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Perforina/genética , Proteínas Citotóxicas Formadoras de Poros , Multimerización de Proteína , Estructura Terciaria de Proteína
16.
Gene Ther ; 17(9): 1105-16, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20428216

RESUMEN

The anti-tumor efficacy of adoptively transferred T cells requires their in vivo persistence and memory polarization. It is unknown if human chimeric antigen receptor (CAR)-expressing T cells can also undergo memory polarization. We examined the functional status of CAR CD8(+) T cells, re-directed to Lewis Y antigen (LeY-T), throughout a period of ex vivo expansion. Immediately before culture CD8(+) T cells comprised a mixture of phenotypes including naive (CD45RA(+)/CCR7(+)/CD27(+)/CD28(+)/perforin-), central memory (CM, CD45RA(-)/CCR7(lo)/CD27(+)/CD28(+)/perforin(lo)), effector memory (EM, CD45RA(-)/CCR7(-)/CD27(+)/CD28(+)/perforin(mod)) and effector (Eff, CD45RA(+)/CCR7(-)/CD27(-)/CD28(-)/perforin(hi)) cells. After transduction and expansion culture of peripheral blood mononuclear cells from normal donors or multiple myeloma patients, CD8(+) LeY-T cells polarized to EM- and CM-like phenotype. CD8(+) LeY-T cells differed from starting CD8(+) CM and EM T cells in that CD27, but not CD28, was downregulated. In addition, CD8(+) LeY-T cells expressed high levels of perforin, similar to starting CD8(+) Eff. CD8(+) LeY-T cells also showed hallmarks of both memory and Eff function, underwent homeostatic proliferation in response to interleukin (IL)-15, and showed interferon (IFN)-γ production and cytotoxicity in response to Le-Y antigen on OVCAR-3 (human ovarian adenocarcinoma) cells. This study confirms CD8(+) LeY-T cells have a CM- and EM-like phenotype and heterogeneous function consistent with potential to persist in vivo after adoptive transfer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Memoria Inmunológica , Receptores de Antígenos/genética , Antígenos CD28/inmunología , Proliferación Celular , Humanos , Interferón gamma/metabolismo , Antígenos Comunes de Leucocito/inmunología , Fenotipo , Receptores de Antígenos/inmunología , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
17.
Gene Ther ; 17(5): 678-86, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20200563

RESUMEN

We have evaluated the carbohydrate antigen Lewis(Y) (Le(Y)) as a potential target for T-cell immunotherapy of hematological neoplasias. Analysis of 81 primary bone marrow samples revealed moderate Le(Y) expression on plasma cells of myeloma patients and myeloblasts of patients with acute myeloid leukemia (AML) (52 and 46% of cases, respectively). We developed a retroviral vector construct encoding a chimeric T-cell receptor that recognizes the Le(Y) antigen in a major histocompatibility complex-independent manner and delivers co-stimulatory signals to achieve T-cell activation. We have shown efficient transduction of peripheral blood-derived T cells with this construct, resulting in antigen-restricted interferon-gamma secretion and cell lysis of Le(Y)-expressing tumor cells. In vivo activity of gene-modified T cells was demonstrated in the delayed growth of myeloma xenografts in NOD/SCID mice, which prolonged survival. Therefore, targeting Le(Y)-positive malignant cells with T cells expressing a chimeric receptor recognizing Le(Y) was effective both in vitro and in a myeloma mouse model. Consequently, we plan to use T cells manufactured under Good Manufacturing Practice conditions in a phase I immunotherapy study for patients with Le(Y)-positive myeloma or AML.


Asunto(s)
Terapia Genética/métodos , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/terapia , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Mieloma Múltiple/terapia , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/trasplante , Animales , Femenino , Vectores Genéticos , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores de Antígenos de Linfocitos T/inmunología , Retroviridae/genética , Linfocitos T/inmunología , Transducción Genética
18.
Cell Death Differ ; 17(4): 607-15, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20075937

RESUMEN

Cytotoxic lymphocytes (CLs) are the killer cells that destroy intracellular pathogen-infected and transformed cells, predominantly through the cytotoxic granule-mediated death pathway. Soluble cytotoxic granule components, including pore-forming perforin and pro-apoptotic serine proteases, granzymes, synergize to induce unscheduled apoptosis of the target cell. A complete loss of CL function results in an aggressive immunoregulatory disorder, familial hemophagocytic lymphohistiocytosis, whereas a partial loss of function seems to be a factor strongly predisposing to hematological malignancies. This review discusses the pathological manifestations of CL deficiencies due to impaired perforin function and describes novel aspects of perforin biology.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Vigilancia Inmunológica/fisiología , Neoplasias/enzimología , Neoplasias/inmunología , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Linfocitos T Citotóxicos/enzimología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Predisposición Genética a la Enfermedad/genética , Granzimas/metabolismo , Humanos , Sistema Inmunológico/enzimología , Sistema Inmunológico/fisiopatología , Linfohistiocitosis Hemofagocítica/enzimología , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Ratones , Neoplasias/genética , Perforina , Proteínas Citotóxicas Formadoras de Poros/genética , Linfocitos T Citotóxicos/inmunología
19.
Cell Death Differ ; 17(4): 577-85, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19927156

RESUMEN

Type 1 diabetes results from autoimmune destruction of pancreatic beta-cells by CD8(+) T cells. The requirement for CD8(+) T cells implicates perforin and granzymes as effectors of tissue destruction. Diabetogenic cytotoxic T cells kill beta-cells by the perforin/granzyme pathway in vitro. In the non-obese diabetic mouse model of type I diabetes, perforin deficiency results in a highly significant reduction in disease, indicating a direct role for perforin in beta-cell death in vivo, although other cell death pathways must account for the residual diabetes in perforin-deficient mice. Perforin and granzyme B are also important in allogeneic destruction of islets. The dominant role of the perforin/granzyme pathway in beta-cell destruction in type I diabetes and allogeneic islet graft rejection make this pathway an important target for blockade in future therapies for type I diabetes. In addition, granzymes have a newly recognized role in inflammation, a feature of both type I and II diabetes, suggesting their role should be further explored in both the common forms of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Granzimas/fisiología , Perforina/fisiología , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/inmunología , Animales , Muerte Celular/inmunología , Diabetes Mellitus Tipo 1/fisiopatología , Rechazo de Injerto/inmunología , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/fisiopatología , Células Secretoras de Insulina/inmunología , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA