Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cancers (Basel) ; 16(6)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38539479

RESUMEN

Immune checkpoint inhibitors have been proposed as the standard treatment for different stages of non-small-cell lung cancer in multiple indications. Not all patients benefit from these treatments, however, and certain patients develop immune-related adverse events. Although the search for predictors of response to these drugs is a major field of research, these issues have yet to be resolved. It has been postulated that microbiota could play a relevant role in conditioning the response to cancer treatments; however, the human factor of intestinal permeability also needs to be considered as it is closely related to the regulation of host-microbiota interaction. In this article, we analyzed the possible relationship between the response to immune checkpoint inhibitors and the onset of immune-related adverse events, gut microbiota status, and intestinal membrane permeability. In a pioneering step, we also measured short-chain fatty acid content in feces. Although the correlation analyses failed to identify predictive biomarkers, even when all variables were integrated, our patients' microbial gut ecosystems were rich and diverse, and the intestinal barrier's integrity was preserved. These results add new knowledge on the composition of microbiota and its correlation with barrier permeability and short-chain fatty acids and suggest that more studies are required before these potential biomarkers can be incorporated into the clinical management of patients via immune checkpoint inhibitor treatment.

2.
Sci Rep ; 14(1): 4395, 2024 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-38388659

RESUMEN

Self-assembling peptides (SAPs) have gained significant attention in biomedicine because of their unique properties and ability to undergo molecular self-assembly driven by non-covalent interactions. By manipulating their composition and structure, SAPs can form well-ordered nanostructures with enhanced selectivity, stability and biocompatibility. SAPs offer advantages such as high chemical and biological diversity and the potential for functionalization. However, studies concerning its potentially toxic effects are very scarce, a limitation that compromises its potential translation to humans. This study investigates the potentially toxic effects of six different SAP formulations composed of natural amino acids designed for nervous tissue engineering and amenable to ready cross-linking boosting their biomechanical properties. All methods were performed in accordance with the relevant guidelines and regulations. A wound-healing assay was performed to evaluate how SAPs modify cell migration. The results in vitro demonstrated that SAPs did not induce genotoxicity neither skin sensitization. In vivo, SAPs were well-tolerated without any signs of acute systemic toxicity. Interestingly, SAPs were found to promote the migration of endothelial, macrophage, fibroblast, and neuronal-like cells in vitro, supporting a high potential for tissue regeneration. These findings contribute to the development and translation of SAP-based biomaterials for biomedical applications.


Asunto(s)
Nanoestructuras , Péptidos , Humanos , Péptidos/química , Ingeniería de Tejidos/métodos , Neuronas , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/química , Nanoestructuras/química
3.
Front Immunol ; 15: 1289303, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38352878

RESUMEN

Immunotherapy treatments aim to modulate the host's immune response to either mitigate it in inflammatory/autoimmune disease or enhance it against infection or cancer. Among different immunotherapies reaching clinical application during the last years, chimeric antigen receptor (CAR) immunotherapy has emerged as an effective treatment for cancer where different CAR T cells have already been approved. Yet their use against infectious diseases is an area still relatively poorly explored, albeit with tremendous potential for research and clinical application. Infectious diseases represent a global health challenge, with the escalating threat of antimicrobial resistance underscoring the need for alternative therapeutic approaches. This review aims to systematically evaluate the current applications of CAR immunotherapy in infectious diseases and discuss its potential for future applications. Notably, CAR cell therapies, initially developed for cancer treatment, are gaining recognition as potential remedies for infectious diseases. The review sheds light on significant progress in CAR T cell therapy directed at viral and opportunistic fungal infections.


Asunto(s)
Enfermedades Transmisibles , Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia , Inmunoterapia Adoptiva , Neoplasias/terapia , Enfermedades Transmisibles/terapia
4.
Sci Rep ; 13(1): 23061, 2023 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-38155275

RESUMEN

Suboptimal vaccine response is a significant concern in patients with Inflammatory Bowel Disease (IBD) receiving biologic drugs. This single-center observational study involved 754 patients with IBD. In Phase I (October 2020-April 2021), 754 IBD participants who had not previously received the SARS-CoV-2 vaccine, underwent blood extraction to assess the seroprevalence of SARS-CoV-2 infection and IBD-related factors. Phase II (May 2021-October 2021) included a subgroup of 52 IBD participants with confirmed previous SARS-CoV-2 infection, who were studied for humoral and cellular response to the SARS-CoV-2 vaccine. In Phase I, treatment with anti-TNF was associated with lower rates of seroconversion (aOR 0.25 95% CI [0.10-0.61]). In Phase II, a significant increase in post-vaccination IgG levels was observed regardless of biologic treatment. However, patients treated with anti-TNF exhibited significantly lower IgG levels compared to those without IBD therapy (5.32 ± 2.47 vs. 7.99 ± 2.59 U/ml, p = 0.042). Following vaccination, a lymphocyte, monocyte, and NK cell activation pattern was observed, with no significant differences between patients receiving biologic drugs and those without IBD treatment. Despite lower seroprevalence and humoral response to the SARS-CoV-2 vaccine in patients treated with anti-TNF, the cellular response to the vaccine did not differ significantly from that patients without IBD therapy.


Asunto(s)
COVID-19 , Enfermedades Inflamatorias del Intestino , Humanos , Vacunas contra la COVID-19 , Estudios Seroepidemiológicos , Inhibidores del Factor de Necrosis Tumoral , SARS-CoV-2 , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Vacunación , Inmunoglobulina G
5.
Cancers (Basel) ; 15(2)2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36672279

RESUMEN

(1) Despite the effectiveness of immune checkpoint inhibitors (ICIs) in lung cancer, there is a lack of knowledge about predictive biomarkers. The objective of our study is to analyze different subsets of T-lymphocytes and natural killer (NK) cells as predictive biomarkers in a cohort of patients with nonsmall cell lung cancer (NSCLC) treated with ICI. (2) This is an observational, prospective study with 55 NSCLC patients treated with ICI. A total of 43 T and NK cell subsets are analyzed in peripheral blood, including the main markers of exhaustion, differentiation, memory, activation, and inhibition. (3) Regarding the descriptive data, Granzyme B+CD4+ Treg lymphocytes stand out (median 17.4%), and within the NK populations, most patients presented cytotoxic NK cells (CD56+CD3-CD16+GranzymeB+; median 94.8%), and about half of them have highly differentiated adaptive-like NK cells (CD56+CD3-CD16+CD57+ (mean 59.8%). A statistically significant difference was observed between the expression of PD1 within the CD56bright NK cell subpopulation (CD56+CD3-CD16-PD-1+) (p = 0.047) and a better OS. (4) Circulating immune cell subpopulations are promising prognostic biomarkers for ICI. Pending on validation with a larger sample, here we provide an analysis of the major circulating T and NK cell subsets involved in cancer immunity, with promising results despite a small sample size.

6.
Oncoimmunology ; 11(1): 2096359, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35813574

RESUMEN

The contribution of the T cell-related inhibitory checkpoint PD-1 to the regulation of NK cell activity is still not clear with contradictory results concerning its expression and role in the modulation of NK cell cytotoxicity. We provide novel key findings on the mechanism involved in the regulation of PD-1 expression on NK cell membrane and its functional consequences for the elimination of cancer cells. In contrast to freshly isolated NK cells from cancer patients, those from healthy donors did not express PD-1 on the cell membrane. However, when healthy NK cells were incubated with tumor target cells, membrane PD-1 expression increased, concurrent with the CD107a surface mobilization. This finding suggested that PD-1 was translocated to the cell membrane during NK cell degranulation after contact with target cells. Indeed, cytosolic PD-1 was expressed in freshly-isolated-NK cells and partly co-localized with CD107a and GzmB, confirming that membrane PD-1 corresponded to a pool of preformed PD-1. Moreover, NK cells that had mobilized PD-1 to the cell membrane presented a significantly reduced anti-tumor activity on PD-L1-expressing-tumor cells in vitro and in vivo, which was partly reversed by using anti-PD-1 blocking antibodies. Our results indicate that NK cells from healthy individuals express cytotoxic granule-associated PD-1, which is rapidly mobilized to the cell membrane after interaction with tumor target cells. This novel finding helps to understand how PD-1 expression is regulated on NK cell membrane and the functional consequences of this expression during the elimination of tumor cells, which will help to design more efficient NK cell-based cancer immunotherapies.


Asunto(s)
Antineoplásicos , Neoplasias , Membrana Celular/metabolismo , Humanos , Inmunoterapia , Células Asesinas Naturales/metabolismo , Activación de Linfocitos
7.
Front Immunol ; 13: 896228, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35651603

RESUMEN

NK cells are key mediators of immune cell-mediated cytotoxicity toward infected and transformed cells, being one of the main executors of cell death in the immune system. NK cells recognize target cells through an array of inhibitory and activating receptors for endogenous or exogenous pathogen-derived ligands, which together with adhesion molecules form a structure known as immunological synapse that regulates NK cell effector functions. The main and best characterized mechanisms involved in NK cell-mediated cytotoxicity are the granule exocytosis pathway (perforin/granzymes) and the expression of death ligands. These pathways are recognized as activators of different cell death programmes on the target cells leading to their destruction. However, most studies analyzing these pathways have used pure recombinant or native proteins instead of intact NK cells and, thus, extrapolation of the results to NK cell-mediated cell death might be difficult. Specially, since the activation of granule exocytosis and/or death ligands during NK cell-mediated elimination of target cells might be influenced by the stimulus received from target cells and other microenvironment components, which might affect the cell death pathways activated on target cells. Here we will review and discuss the available experimental evidence on how NK cells kill target cells, with a special focus on the different cell death modalities that have been found to be activated during NK cell-mediated cytotoxicity; including apoptosis and more inflammatory pathways like necroptosis and pyroptosis. In light of this new evidence, we will develop the new concept of cell death induced by NK cells as a new regulatory mechanism linking innate immune response with the activation of tumour adaptive T cell responses, which might be the initiating stimulus that trigger the cancer-immunity cycle. The use of the different cell death pathways and the modulation of the tumour cell molecular machinery regulating them might affect not only tumour cell elimination by NK cells but, in addition, the generation of T cell responses against the tumour that would contribute to efficient tumour elimination and generate cancer immune memory preventing potential recurrences.


Asunto(s)
Células Asesinas Naturales , Neoplasias , Inmunidad Adaptativa , Citotoxicidad Inmunológica , Humanos , Ligandos , Microambiente Tumoral
8.
Front Immunol ; 13: 890836, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35747143

RESUMEN

Background: Colorectal cancer (CRC) is a heterogeneous disease with variable mutational profile and tumour microenvironment composition that influence tumour progression and response to treatment. While chemoresistant and poorly immunogenic CRC remains a challenge, the development of new strategies guided by biomarkers could help stratify and treat patients. Allogeneic NK cell transfer emerges as an alternative against chemoresistant and poorly immunogenic CRC. Methods: NK cell-related immunological markers were analysed by transcriptomics and immunohistochemistry in human CRC samples and correlated with tumour progression and overall survival. The anti-tumour ability of expanded allogeneic NK cells using a protocol combining cytokines and feeder cells was analysed in vitro and in vivo and correlated with CRC mutational status and the expression of ligands for immune checkpoint (IC) receptors regulating NK cell activity. Results: HLA-I downmodulation and NK cell infiltration correlated with better overall survival in patients with a low-stage (II) microsatellite instability-high (MSI-H) CRC, suggesting a role of HLA-I as a prognosis biomarker and a potential benefit of NK cell immunotherapy. Activated allogeneic NK cells were able to eliminate CRC cultures without PD-1 and TIM-3 restriction but were affected by HLA-I expression. In vivo experiments confirmed the efficacy of the therapy against both HLA+ and HLA- CRC cell lines. Concomitant administration of pembrolizumab failed to improve tumour control. Conclusions: Our results reveal an immunological profile of CRC tumours in which immunogenicity (MSI-H) and immune evasion mechanisms (HLA downmodulation) favour NK cell immunosurveillance at early disease stages. Accordingly, we have shown that allogeneic NK cell therapy can target tumours expressing mutations conferring poor prognosis regardless of the expression of T cell-related inhibitory IC ligands. Overall, this study provides a rationale for a new potential basis for CRC stratification and NK cell-based therapy.


Asunto(s)
Neoplasias Colorrectales , Inestabilidad de Microsatélites , Neoplasias Colorrectales/patología , Humanos , Inmunoterapia/métodos , Células Asesinas Naturales , Ligandos , Microambiente Tumoral
9.
FEBS J ; 289(15): 4398-4415, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34174027

RESUMEN

Cytotoxic lymphocytes (CLs), and more specifically Tc and NK cells, are the main executors of cell death in the immune system, playing a key role during both immunosurveillance and immunotherapy. These cells induce regulated cell death (RCD) by different mechanisms, being granular exocytosis and expression of death ligands the most prominent and best characterized ones. Apoptosis, a traditionally considered low-inflammatory type of cell death, has been accepted for years as the paradigm of RCD induced by CLs. However, several recent studies have demonstrated that NK cells and Tc cells can also induce more inflammatory forms of cell death, namely, necroptosis, pyroptosis, and ferroptosis. Activation of these highly inflammatory types of cell death appears to critically contribute to the activation of a successful antitumour immune response. Additionally, the role of specific cell death pathways in immunogenic cell death is still under intense debate, especially considering the interconnections with other inflammatory forms of cell death. These evidences, together with the advent of new cancer immunotherapies, highlight the necessity to deepen our understanding of the link between the cell death triggered by CLs and inflammation. This knowledge will be instrumental to maximize the antitumour potential of immunotherapies, minimizing deleterious effects associated with these treatments. In this review, we will briefly summarize the main features of apoptosis, necroptosis, pyroptosis and ferroptosis, to subsequently discuss the most recent evidences about the role of these RCD pathways during the elimination of cancer cells mediated by CLs and its modulation to increase the efficacy of cancer immunotherapy.


Asunto(s)
Antineoplásicos , Piroptosis , Apoptosis , Muerte Celular , Células Asesinas Naturales , Necroptosis
10.
Theranostics ; 11(20): 9873-9883, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34815792

RESUMEN

Aims: Recent in vitro findings suggest that the serine protease Granzyme K (GzmK) may act as a proinflammatory mediator. However, its role in sepsis is unknown. Here we aim to understand the role of GzmK in a mouse model of bacterial sepsis and compare it to the biological relevance of Granzyme A (GzmA). Methods: Sepsis was induced in WT, GzmA-/- and GzmK-/- mice by an intraperitoneal injection of 2x108 CFU from E. coli. Mouse survival was monitored during 5 days. Levels of IL-1α, IL-1ß, TNFα and IL-6 in plasma were measured and bacterial load in blood, liver and spleen was analyzed. Finally, profile of cellular expression of GzmA and GzmK was analyzed by FACS. Results: GzmA and GzmK are not involved in the control of bacterial infection. However, GzmA and GzmK deficient mice showed a lower sepsis score in comparison with WT mice, although only GzmA deficient mice exhibited increased survival. GzmA deficient mice also showed reduced expression of some proinflammatory cytokines like IL1-α, IL-ß and IL-6. A similar result was found when extracellular GzmA was therapeutically inhibited in WT mice using serpinb6b, which improved survival and reduced IL-6 expression. Mechanistically, active extracellular GzmA induces the production of IL-6 in macrophages by a mechanism dependent on TLR4 and MyD88. Conclusions: These results suggest that although both proteases contribute to the clinical signs of E. coli-induced sepsis, inhibition of GzmA is sufficient to reduce inflammation and improve survival irrespectively of the presence of other inflammatory granzymes, like GzmK.


Asunto(s)
Granzimas/metabolismo , Sepsis/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Sepsis/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Theranostics ; 11(8): 3781-3795, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33664861

RESUMEN

Aims: Peritonitis is one of the most common causes of sepsis, a serious syndrome characterized by a dysregulated systemic inflammatory response. Recent evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed by NK and T cells, could act as a proinflammatory mediator and could play an important role in the pathogenesis of sepsis. This work aims to analyze the role and the therapeutic potential of GzmA in the pathogenesis of peritoneal sepsis. Methods: The level of extracellular GzmA as well as GzmA activity were analyzed in serum from healthy volunteers and patients with confirmed peritonitis and were correlated with the Sequential Organ Failure Assessment (SOFA) score. Peritonitis was induced in C57Bl/6 (WT) and GzmA-/- mice by cecal ligation and puncture (CLP). Mice were treated intraperitoneally with antibiotics alone or in combination serpinb6b, a specific GzmA inhibitor, for 5 days. Mouse survival was monitored during 14 days, levels of some proinflammatory cytokines were measured in serum and bacterial load and diversity was analyzed in blood and spleen at different times. Results: Clinically, elevated GzmA was observed in serum from patients with abdominal sepsis suggesting that GzmA plays an important role in this pathology. In the CLP model GzmA deficient mice, or WT mice treated with an extracellular GzmA inhibitor, showed increased survival, which correlated with a reduction in proinflammatory markers in both serum and peritoneal lavage fluid. GzmA deficiency did not influence bacterial load in blood and spleen and GzmA did not affect bacterial replication in macrophages in vitro, indicating that GzmA has no role in bacterial control. Analysis of GzmA in lymphoid cells following CLP showed that it was mainly expressed by NK cells. Mechanistically, we found that extracellular active GzmA acts as a proinflammatory mediator in macrophages by inducing the TLR4-dependent expression of IL-6 and TNFα. Conclusions: Our findings implicate GzmA as a key regulator of the inflammatory response during abdominal sepsis and provide solid evidences about its therapeutic potential for the treatment of this severe pathology.


Asunto(s)
Granzimas/antagonistas & inhibidores , Peritonitis/tratamiento farmacológico , Peritonitis/enzimología , Sepsis/tratamiento farmacológico , Sepsis/enzimología , Anciano , Anciano de 80 o más Años , Animales , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Granzimas/sangre , Granzimas/deficiencia , Granzimas/genética , Humanos , Mediadores de Inflamación/sangre , Interleucina-6/biosíntesis , Células Asesinas Naturales/enzimología , Macrófagos/enzimología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Terapia Molecular Dirigida , Peritonitis/etiología , Medicina de Precisión , Sepsis/etiología , Serpinas/farmacología , Receptor Toll-Like 4/metabolismo
12.
Front Oncol ; 10: 568939, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117698

RESUMEN

The advances in molecular biology and the emergence of Next Generation Sequencing (NGS) have revealed that microbiome composition is closely related with health and disease, including cancer. This relationship affects different levels of cancer such as development, progression, and response to treatment including immunotherapy. The efficacy of immune checkpoint inhibitors (ICIs) may be influenced by the concomitant use of antibiotics before, during or shortly after treatment with ICIs. Nevertheless, the linking mechanism between microbiote, host immunity and cancer is not clear and the role of microbiota manipulation and analyses in cancer management has not been clinically validated yet. Regarding the use of microbiome as biomarker to predict ICI efficacy it has been recently shown that the use of biochemical serum markers to monitor intestinal permeability and loss of barrier integrity, like citrulline, could be useful to monitor microbiota changes and predict ICI efficacy. There are still many unknowns about the role of these components, their relationship with the microbiota, with the use of antibiotics and the response to immunotherapy. The next challenge in microbiome research will be to identify individual microbial species that causally affect lung cancer phenotypes and response to ICI and disentangle the underlying mechanisms. Thus, further analyses in patients with lung cancer receiving treatment with ICIs and its correlation with the composition of the microbiota in different organs including the respiratory tract, peripheral blood and intestinal tract could be useful to predict the efficacy of ICIs and its modulation with antibiotic use.

13.
Cell Rep ; 32(1): 107847, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32640217

RESUMEN

If not properly regulated, the inflammatory immune response can promote carcinogenesis, as evident in colorectal cancer (CRC). Aiming to gain mechanistic insight into the link between inflammation and CRC, we perform transcriptomics analysis of human CRC, identifying a strong correlation between expression of the serine protease granzyme A (GzmA) and inflammation. In a dextran sodium sulfate and azoxymethane (DSS/AOM) mouse model, deficiency and pharmacological inhibition of extracellular GzmA both attenuate gut inflammation and prevent CRC development, including the initial steps of cell transformation and epithelial-to-mesenchymal transition. Mechanistically, extracellular GzmA induces NF-κB-dependent IL-6 production in macrophages, which in turn promotes STAT3 activation in cultured CRC cells. Accordingly, colon tissues from DSS/AOM-treated, GzmA-deficient animals present reduced levels of pSTAT3. By identifying GzmA as a proinflammatory protease that promotes CRC development, these findings provide information on mechanisms that link immune cell infiltration to cancer progression and present GzmA as a therapeutic target for CRC.


Asunto(s)
Carcinogénesis/patología , Colon/patología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Espacio Extracelular/enzimología , Granzimas/metabolismo , Inflamación/patología , Enfermedad Aguda , Animales , Azoximetano , Carcinogénesis/genética , Enfermedad Crónica , Neoplasias Colorrectales/genética , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Sulfato de Dextran , Progresión de la Enfermedad , Granzimas/antagonistas & inhibidores , Granzimas/genética , Humanos , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-6/biosíntesis , Ratones Noqueados , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Trends Cancer ; 6(2): 86-97, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32061309

RESUMEN

Microbiota have emerged as key modulators of both the carcinogenic process and the immune response against cancer cells, and, thus, it seems to influence the efficacy of immunotherapy. While most studies have focused on analyzing the influence of gut microbiota, its composition substantially differs from that in the lung. Here, we describe how microbial life in the lungs is associated with host immune status in the lungs and, thus, how the identification of the microbial populations in the lower respiratory tract rather than in the gut might be key to understanding the lung carcinogenic process and to predict the efficacy of different treatments. Understanding the influence of lung microbiota on host immunity may identify new therapeutic targets and help to design new immunotherapy approaches to treat lung cancer.


Asunto(s)
Carcinogénesis/inmunología , Disbiosis/complicaciones , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/microbiología , Microbiota/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Antibacterianos/efectos adversos , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/metabolismo , Carcinogénesis/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/patología , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad Innata/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Microbiota/efectos de los fármacos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Supervivencia sin Progresión , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología
15.
Cancer Cell ; 36(3): 237-249.e6, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31447348

RESUMEN

Deficiency in DNA double-strand break (DSB) repair mechanisms has been widely exploited for the treatment of different malignances, including homologous recombination (HR)-deficient breast and ovarian cancers. Here we demonstrate that diffuse large B cell lymphomas (DLBCLs) expressing LMO2 protein are functionally deficient in HR-mediated DSB repair. Mechanistically, LMO2 inhibits BRCA1 recruitment to DSBs by interacting with 53BP1 during repair. Similar to BRCA1-deficient cells, LMO2-positive DLBCLs and T cell acute lymphoblastic leukemia (T-ALL) cells exhibit a high sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors. Furthermore, chemotherapy and PARP inhibitors synergize to inhibit the growth of LMO2-positive tumors. Together, our results reveal that LMO2 expression predicts HR deficiency and the potential therapeutic use of PARP inhibitors in DLBCL and T-ALL.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas con Dominio LIM/metabolismo , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Reparación del ADN por Recombinación/efectos de los fármacos , Mutaciones Letales Sintéticas/efectos de los fármacos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína BRCA1/metabolismo , Biopsia , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Tonsila Palatina/patología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/metabolismo , Cultivo Primario de Células , Reparación del ADN por Recombinación/genética , Proteína 1 de Unión al Supresor Tumoral P53 , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Front Immunol ; 10: 3010, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998304

RESUMEN

Immune checkpoint receptors (IC) positively or negatively regulate the activation of the host immune response, preventing unwanted reactions against self-healthy tissues. In recent years the term IC has been mainly used for the inhibitory ICs, which are critical to control Natural Killer (NK) and Cytotoxic CD8+ T cells due to its high cytotoxic potential. Due to the different nature of the signals that regulate T and NK cell activation, specific ICs have been described that mainly regulate either NK cell or T cell activity. Thus, strategies to modulate NK cell activity are raising as promising tools to treat tumors that do not respond to T cell-based immunotherapies. NK cell activation is mainly regulated by ICs and receptors from the KIR, NKG2 and NCRs families and the contribution of T cell-related ICs is less clear. Recently, NK cells have emerged as contributors to the effect of inhibitors of T cell-related ICs like CTLA4, LAG3 or the PD1/PD-L1 axes in cancer patients, suggesting that these ICs also regulate the activity of NK cells under pathological conditions. Strikingly, in contrast to NK cells from cancer patients, the level of expression of these ICs is low on most subsets of freshly isolated and in vitro activated NK cells from healthy patients, suggesting that they do not control NK cell tolerance and thus, do not act as conventional ICs under non-pathological conditions. The low level of expression of T cell-related ICs in "healthy" NK cells suggest that they should not be restricted to the detrimental effects of these inhibitory mechanisms in the cancer microenvironment. After a brief introduction of the regulatory mechanisms that control NK cell anti-tumoral activity and the conventional ICs controlling NK cell tolerance, we will critically discuss the potential role of T cell-related ICs in the control of NK cell activity under both physiological and pathological (cancer) conditions. This discussion will allow to comprehensively describe the chances and potential limitations of using allogeneic NK cells isolated from a healthy environment to overcome immune subversion by T cell-related ICs and to improve the efficacy of IC inhibitors (ICIs) in a safer way.


Asunto(s)
Antígenos CD/inmunología , Antígeno CTLA-4/inmunología , Células Asesinas Naturales/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Traslado Adoptivo/métodos , Animales , Linfocitos T CD8-positivos/inmunología , Humanos , Microambiente Tumoral/inmunología
17.
Cancer Lett ; 382(1): 1-10, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27565383

RESUMEN

The proteasome inhibitor bortezomib is now the cornerstone of combination therapy of multiple myeloma (MM). Carfilzomib, a second-generation inhibitor, has shown a substantial benefit vs bortezomib in combination regimes. Here we have analyzed in detail the mechanism of cell death induced by carfilzomib and its crosstalk with autophagy and applied the results to the in vivo treatment of MM in a mouse model. Carfilzomib induced apoptosis essentially by the intrinsic pathway, through the up-regulation of Puma and Noxa proteins followed by the interaction of Puma, Noxa and Bim with Bax and of Noxa with Bak. Carfilzomib also produces an increase in the formation of autophagosomes but, as apoptosis progresses, autophagy is disrupted, probably owing to Beclin 1 and p62 inactivation. Cotreatment with chloroquine, which blocks autophagy, strongly potentiated apoptosis in vitro and in vivo. Accordingly, combination therapy with carfilzomib plus chloroquine was highly effective in the treatment of MM in a mouse xenograft model. Chloroquine also enhanced carfilzomib-induced calreticulin exposure in MM cells undergoing apoptosis, increasing the immunogenic ability of carfilzomib. These results support design of trials combining carfilzomib with chloroquine to improve MM therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Cloroquina/farmacología , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos/farmacología , Inhibidores de Proteasoma/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1/metabolismo , Calreticulina/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Ratones Desnudos , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Biochem Cell Biol ; 49: 42-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24440757

RESUMEN

NK cell is an innate immune system lymphocyte lineage with natural cytotoxicity. Its optimal use in the clinic requires in vitro expansion and activation. Cytokines and encounter with target cells activate NK cells and induce proliferation, and this could depend on the presence of other immune cells. Here we activated PBMCs during 5 days with IL-2, with IL-2 plus the tumor cell line K562 and with the lymphoblastoid cell line R69 and perform integrated analyses of microRNA and mRNA expression profiles of purified NK cells. The samples cluster depending on the stimuli and not on the donor, indicating that the pattern of NK cell stimulation is acutely well conserved between individuals. Regulation of mRNA expression is tighter than that of miRNA expression. All stimuli induce a common preserved genetic remodeling. In addition, encounter with target cells mainly activates pathways related to metabolism. Different target cells induce different NK cell remodeling which affects cytokine response and cytotoxicity, supporting the notion that encounter with different target cells significantly changing the activation pattern. We validate our analysis by showing that activation down regulates miR-23a, which is a negative regulator of cathepsin C (CTSC) mRNA, a gene up regulated by all stimuli. The peptidase CTSC activates the granzymes, the main effector proteases involved in NK cell cytotoxicity. All-trans retinoic acid (ATRA), which induces miR-23a expression, decreases CTSC expression and granzyme B activity leading to impaired NK cell cytotoxicity in an in vivo mouse model.


Asunto(s)
Catepsina C/genética , Citotoxicidad Inmunológica/efectos de los fármacos , Granzimas/genética , Células Asesinas Naturales/efectos de los fármacos , MicroARNs/genética , Tretinoina/farmacología , Animales , Western Blotting , Catepsina C/metabolismo , Línea Celular , Células Cultivadas , Análisis por Conglomerados , Femenino , Granzimas/metabolismo , Humanos , Interleucina-2/farmacología , Células Jurkat , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Componente Principal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
19.
Biochem Pharmacol ; 87(3): 410-23, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24269628

RESUMEN

Granulysin is a protein present in the granules of human CTL and NK cells, with cytolytic activity against microbes and tumors. Previous work demonstrated that granulysin caused cell death through mitochondrial damage with release of AIF and cytochrome c. However, the molecular mechanism and, especially, the type of cell death were still not well defined. In the present work we show that granulysin-induced cell death is apoptotic, with phosphatidylserine exposure preceding membrane breakdown and with caspase 3 activation. Granulysin-induced apoptosis is prevented in Jurkat cells over-expressing Bcl-xL or Bcl2, or lacking Bak and Bax or Bim expression, suggesting a central role of the mitochondrial apoptotic pathway. This apoptotic process is initiated by intracellular Ca(2+) increase and mitochondrial ROS generation. We have tested granulysin against other hematological tumor cells such as multiple myeloma cell lines, and cells from B cell chronic lymphocytic leukemia (B-CLL) patients, finding different degrees of sensitivity. We also show that granulysin induces the cleavage of Atg5 in the complex formed with Atg12, without affecting autophagy. In conclusion, granulysin induces apoptosis on hematological tumor cells and on cells from B-CLL patients, opening the door to research on its use as a new anti-tumoral treatment.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Apoptosis/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Antígenos de Diferenciación de Linfocitos T/genética , Proteína 5 Relacionada con la Autofagia , Calcio/metabolismo , Humanos , Células Jurkat , Leucemia de Células B/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno
20.
Rev. cuba. invest. bioméd ; 28(1)ene.-mar. 2009. graf
Artículo en Español | LILACS | ID: lil-532156

RESUMEN

El veneno del alacrán azul, Rophalurus junceus es comercializado con el nombre de Escozul. Este producto natural se emplea en el tratamiento de diferentes patologías. Este trabajo evaluó el efecto citotóxico in vitro de este producto en las líneas tumorales P3-X63/AG8/653 y Dunning R3327-G provenientes de mieloma murino y próstata de rata, respectivamente. La citotoxicidad fue evaluada mediante la cinética de crecimiento celular y el daño metabólico. Se utilizaron dosis de 1, 10, 20, 50, 100 y 200 mg/mL. El veneno presentó un efecto citostático dependiente de la línea tumoral en cuestión. Las dosis efectivas variaron entre las líneas celulares ensayadas. Se estudió además la estabilidad del producto almacenado durante 30 días a temperaturas de -20 y 4ºC, se evidenció la pérdida de la actividad biológica. El trabajo demostró la citotoxicidad del veneno crudo del alacrán azul en cultivos celulares.


Poison of blue scorpion (Rophalurus junceus) is marketed as Escozul. This natural product is used in treatment of different pathologies. Present paper evaluated the in vitro cytotoxic effect of this product in P3-X63/AG8/653 and Dunning R3327-G tumor lines from murine myeloma and rat prostate, respectively. Cytotoxic effect was evaluated by means of cellular growing kinetics and the metabolic damage. Doses of 1,10, 20, 50, 100, and 200 ìg/mL. Poison had a cytostatic effect dependent of tumor line at issue. Doses effective varied among the cellular lines assessed. We studied also stability of product stored during 30 days at temperatures of -20° and 4° C, evidenced the loss of biological activity. We showed cytotoxic effect of crude poison of blue scorpion in cellular cultures.


Asunto(s)
Técnicas In Vitro , Venenos de Artrópodos/toxicidad , Venenos de Artrópodos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA