Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 11(6): 792-809, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37070661

RESUMO

The pioneering design of chimeric antigen receptor (CAR) T-cell therapy demonstrated the potential of reprogramming the immune system. Nonetheless, T-cell exhaustion, toxicity, and suppressive microenvironments limit their efficacy in solid tumors. We previously characterized a subset of tumor-infiltrating CD4+ T cells expressing the FcγRI receptor. Herein, we detail engineering of a receptor, based on the FcγRI structure, allowing T cells to target tumor cells using antibody intermediates. These T cells showed effective and specific cytotoxicity only when an appropriate antibody was added. Only target-bound antibodies activated these cells, while free antibodies were internalized without activation. Their cytotoxic activity was correlated to target protein density, therefore targeting tumor cells with high antigen density while sparing normal cells with low or no expression. This activation mechanism prevented premature exhaustion. Furthermore, during antibody-dependent cytotoxicity these cells secreted attenuated cytokine levels compared with CAR T cells, thereby enhancing their safety profile. These cells eradicated established melanomas, infiltrated the tumor microenvironment, and facilitated host immune cell recruitment in immunocompetent mice. In NOD/SCID gamma mice the cells infiltrate, persist, and eradicate tumors. As opposed to CAR T-cell therapies, which require changing the receptor across different types of cancer, our engineered T cells remain the same across tumor types, while only the injected antibody changes. Overall, we generated a highly flexible T-cell therapy capable of binding a wide range of tumor cells with high affinity, while preserving the cytotoxic specificity only to cells expressing high density of tumor-associated antigens and using a single manufacturing process.


Assuntos
Imunoterapia Adotiva , Melanoma , Animais , Camundongos , Receptores de IgG , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos SCID , Camundongos Endogâmicos NOD , Melanoma/terapia , Imunoglobulinas , Linhagem Celular Tumoral , Microambiente Tumoral
2.
Elife ; 112022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36124553

RESUMO

Despite the remarkable successes of cancer immunotherapies, the majority of patients will experience only partial response followed by relapse of resistant tumors. While treatment resistance has frequently been attributed to clonal selection and immunoediting, comparisons of paired primary and relapsed tumors in melanoma and breast cancers indicate that they share the majority of clones. Here, we demonstrate in both mouse models and clinical human samples that tumor cells evade immunotherapy by generating unique transient cell-in-cell structures, which are resistant to killing by T cells and chemotherapies. While the outer cells in this cell-in-cell formation are often killed by reactive T cells, the inner cells remain intact and disseminate into single tumor cells once T cells are no longer present. This formation is mediated predominantly by IFNγ-activated T cells, which subsequently induce phosphorylation of the transcription factors signal transducer and activator of transcription 3 (STAT3) and early growth response-1 (EGR-1) in tumor cells. Indeed, inhibiting these factors prior to immunotherapy significantly improves its therapeutic efficacy. Overall, this work highlights a currently insurmountable limitation of immunotherapy and reveals a previously unknown resistance mechanism which enables tumor cells to survive immune-mediated killing without altering their immunogenicity.


Cancer immunotherapies use the body's own immune system to fight off cancer. But, despite some remarkable success stories, many patients only see a temporary improvement before the immunotherapy stops being effective and the tumours regrow. It is unclear why this occurs, but it may have to do with how the immune system attacks cancer cells. Immunotherapies aim to activate a special group of cells known as killer T-cells, which are responsible for the immune response to tumours. These cells can identify cancer cells and inject toxic granules through their membranes, killing them. However, killer T-cells are not always effective. This is because cancer cells are naturally good at avoiding detection, and during treatment, their genes can mutate, giving them new ways to evade the immune system. Interestingly, when scientists analysed the genes of tumour cells before and after immunotherapy, they found that many of the genes that code for proteins recognized by T-cells do not change significantly. This suggests that tumours' resistance to immune attack may be physical, rather than genetic. To investigate this hypothesis, Gutwillig et al. developed several mouse tumour models that stop responding to immunotherapy after initial treatment. Examining cells from these tumours revealed that when the immune system attacks, they reorganise by getting inside one another. This allows some cancer cells to hide under many layers of cell membrane. At this point killer T-cells can identify and inject the outer cell with toxic granules, but it cannot reach the cells inside. This ability of cancer cells to hide within one another relies on them recognising when the immune system is attacking. This happens because the cancer cells can detect certain signals released by the killer T-cells, allowing them to hide. Gutwillig et al. identified some of these signals, and showed that blocking them stopped cancer cells from hiding inside each other, making immunotherapy more effective. This new explanation for how cancer cells escape the immune system could guide future research and lead to new cancer treatments, or approaches to boost existing treatments. Understanding the process in more detail could allow scientists to prevent it from happening, by revealing which signals to block, and when, for best results.


Assuntos
Formação de Célula em Célula , Melanoma , Animais , Humanos , Fatores Imunológicos , Imunoterapia , Melanoma/terapia , Camundongos , Recidiva , Fator de Transcrição STAT3
3.
Cell Immunol ; 361: 104281, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33453508

RESUMO

Dendritic cells (DCs) mature upon an inflammatory trigger. However, an inflammatory trigger can lead to a semi-mature phenotype, allowing DCs to evoke tolerance and expedite the resolution of inflammation. This duality likely involves context-dependent modulation of inflammatory signaling. Human α1-antitrypsin (hAAT) promotes semimature DCs. We examined changes in a wide spectrum of signaling cascades in stimulated murine bone marrow-derived cells with hAAT. Upon stimulation by IL-1ß+IFNγ, hAAT-treated cells depicted an attenuated calcium flux. Disrupting PKA or NF-κB pathways revoked only some hAAT-mediated outcomes. hAAT-treated cells exhibited a distict pattern of kinase phosphorylation. hAAT-mediated increase in Treg cells in-vitro required intact inflammatory signaling pathways. Taken together, hAAT appears to require a stimulated microenvironment to promote inflammatory resolution, setting it aside from classical anti-inflammatory agents. Further studies are required to identify the specific molecules targeted by hAAT that mediate these and other outcomes.


Assuntos
Células Dendríticas/metabolismo , Transdução de Sinais/efeitos dos fármacos , alfa 1-Antitripsina/farmacologia , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/metabolismo , Cálcio/metabolismo , Células Cultivadas , Tolerância Imunológica/imunologia , Inflamação/metabolismo , Interleucina-1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Receptores CCR7/imunologia , Receptores CCR7/metabolismo , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , alfa 1-Antitripsina/metabolismo
4.
Cancer Res ; 80(10): 1942-1956, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32127354

RESUMO

The recent success of checkpoint blockade therapies has established immunotherapy as one of the most promising treatments for melanoma. Nonetheless, a complete curative response following immunotherapy is observed only in a fraction of patients. To identify what factors limit the efficacy of immunotherapies, we established mouse models that cease to respond to immunotherapies once their tumors exceed a certain stage. Analysis of the immune systems of the organisms revealed that the numbers of tumor-infiltrating dendritic cells (TIDC) drastically decreased with time. Further, in contrast to the current paradigm, once melanoma was established, TIDC did not migrate into sentinel lymph nodes. Instead, they underwent local cell death due to excessive phagocytosis of lysosomes. Importantly, TIDC were required to license the cytotoxic activity of tumor CD8+ T cells, and in their absence, T cells did not lyse melanoma cells. Our results offer a paradigm shift regarding the role of TIDC and a framework to increase the efficacy of immunotherapies. SIGNIFICANCE: This work redefines the role of monocyte-derived dendritic cells in melanoma and provides a novel strategy to increase the efficacy of T-cell-based immunotherapies in nonresponding individuals. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/10/1942/F1.large.jpg.


Assuntos
Células Dendríticas/patologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Lisossomos , Melanoma/imunologia , Animais , Apoptose/imunologia , Linfócitos T CD8-Positivos/imunologia , Humanos , Imunoterapia , Ativação Linfocitária/imunologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL
5.
J Clin Invest ; 129(10): 4151-4164, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31449054

RESUMO

While a high frequency of Th1 cells in tumors is associated with improved cancer prognosis, this benefit has been attributed mainly to support of cytotoxic activity of CD8+ T cells. By attempting to potentiate antibody-driven immunity, we found a remarkable synergy between CD4+ T cells and tumor-binding antibodies. This surprising synergy was mediated by a small subset of tumor-infiltrating CD4+ T cells that express the high-affinity Fcγ receptor for IgG (FcγRI) in both mouse and human patients. These cells efficiently lyse tumor cells coated with antibodies through concomitant crosslinking of their T cell receptor (TCR) and FcγRI. By expressing FcγRI and its signaling chain in conventional CD4+ T cells, we successfully employed this mechanism to treat established solid cancers. Overall, this discovery sheds new light on the biology of this T cell subset, their function during tumor immunity, and the means to utilize their unique killing signals in immunotherapy.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Receptores de IgG/metabolismo , Células Th1/classificação , Células Th1/imunologia , Animais , Linfócitos T CD4-Positivos/classificação , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Imunoterapia Adotiva , Masculino , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subpopulações de Linfócitos T/imunologia
6.
Front Immunol ; 10: 590, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001247

RESUMO

Background: Human α1-antitrypsin (hAAT) is a circulating anti-inflammatory serine-protease inhibitor that rises during acute phase responses. in vivo, hAAT reduces bacterial load, without directly inhibiting bacterial growth. In conditions of excess nitric-oxide (NO), hAAT undergoes S-nitrosylation (S-NO-hAAT) and gains antibacterial capacity. The impact of S-NO-hAAT on immune cells has yet to be explored. Aim: Study the effects of S-NO-hAAT on immune cells during bacterial infection. Methods: Clinical-grade hAAT was S-nitrosylated and then compared to unmodified hAAT, functionally, and structurally. Intracellular bacterial clearance by THP-1 macrophages was assessed using live Salmonella typhi. Murine peritoneal macrophages were examined, and signaling pathways were evaluated. S-NO-hAAT was also investigated after blocking free mambranal cysteine residues on cells. Results: S-NO-hAAT (27.5 uM) enhances intracellular bacteria elimination by immunocytes (up to 1-log reduction). S-NO-hAAT causes resting macrophages to exhibit a pro-inflammatory and antibacterial phenotype, including release of inflammatory cytokines and induction of inducible nitric oxide synthase (iNOS) and TLR2. These pro-inflammatory effects are dependent upon cell surface thiols and activation of MAPK pathways. Conclusions: hAAT duality appears to be context-specific, involving S-nitrosylation in a nitric oxide rich environment. Our results suggest that S-nitrosylation facilitates the antibacterial activity of hAAT by promoting its ability to activate innate immune cells. This pro-inflammatory effect may involve transferring of nitric oxide from S-NO-hAAT to a free cysteine residue on cellular targets.


Assuntos
Imunidade Inata , Macrófagos Peritoneais/imunologia , Óxido Nítrico/imunologia , Salmonella typhi/imunologia , alfa 1-Antitripsina/imunologia , Animais , Feminino , Macrófagos Peritoneais/microbiologia , Camundongos , alfa 1-Antitripsina/genética
7.
J Vis Exp ; (135)2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29912184

RESUMO

Dendritic cells (DC) are heterogeneous cell populations that differ in their cell membrane markers, migration patterns and distribution, and in their antigen presentation and T cell activation capacities. Since most vaccinations of experimental tumor models require millions of DC, they are widely isolated from the bone marrow or spleen. However, these DC significantly differ from blood and tumor DC in their responses to immune complexes (IC), and presumably to other Syk-coupled lectin receptors. Importantly, given the sensitivity of DC to danger-associated molecules, the presence of endotoxins or antibodies that crosslink activation receptors in one of the isolating steps could result in the priming of DC and thus affect the parameters, or at least the dosage, required to activate them. Therefore, here we describe a detailed protocol for isolating MoDC from blood and tumors while avoiding their premature activation. In addition, a protocol is provided for MoDC activation with tumor IC, and their subsequent analyses.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Células Dendríticas/imunologia , Monócitos/metabolismo , Neoplasias/imunologia , Animais , Humanos , Camundongos
8.
J Immunol ; 198(4): 1395-1402, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28167650

RESUMO

Over the last decade, danger-associated molecular pattern molecules, or alarmins, have been recognized as signaling mediators of sterile inflammatory responses after trauma and injury. In contrast with the accepted passive release models suggested by the "danger hypothesis," it was recently shown that alarmins can also directly sense and report damage by signaling to the environment when released from live cells undergoing physiological stress, even without loss of subcellular compartmentalization. In this article, we review the involvement of alarmins such as IL-1α, IL-33, IL-16, and high-mobility group box 1 in cellular and physiological stress, and suggest a novel activity of these molecules as central initiators of sterile inflammation in response to nonlethal stress, a function we denote "stressorins." We highlight the role of posttranslational modifications of stressorins as key regulators of their activity and propose that targeted inhibition of stressorins or their modifiers could serve as attractive new anti-inflammatory treatments for a broad range of diseases.


Assuntos
Alarminas/fisiologia , Estresse Fisiológico , Animais , Proteína HMGB1/metabolismo , Humanos , Inflamação , Interleucina-16/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-33/metabolismo , Camundongos , Processamento de Proteína Pós-Traducional , Estresse Fisiológico/imunologia , Estresse Fisiológico/fisiologia
9.
Front Immunol ; 7: 559, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28003813

RESUMO

α1-antitrypsin (AAT), a circulating glycoprotein that rises during acute phase responses and healthy pregnancies, exhibits immunomodulatory properties in several T-cell-dependent immune pathologies. However, AAT does not directly interfere with T-cell responses; instead, it facilitates polarization of macrophages and dendritic cells towards M2-like and tolerogenic cells, respectively. AAT also allows NK cell responses against tumor cells, while attenuating DC-dependent induction of autoimmune NK cell activities. Since AAT-treated macrophages bear resemblance to cancer-promoting tumor-associated macrophages (TAMs), it became imperative to examine the possible induction of tumor permissive conditions by AAT. Here, AAT treatment is examined for its effect on tumor development, metastatic spread, and tumor immunology. Systemic AAT treatment of mice inoculated with B16-F10 melanoma cells resulted in significant inhibition of tumor growth and metastatic spread. Using NK cell-resistant RMA cells, we show that AAT interferes with tumor development in a CD8+ T-cell-dependent manner. Unexpectedly, upon analysis of tumor cellular composition, we identified functional tumor-infiltrating CD8+ T-cells alongside M1-like TAMs in AAT-treated mice. Based on the ability of AAT to undergo chemical modifications, we emulated conditions of elevated reactive nitrogen and oxygen species. Indeed, macrophages were stimulated by treatment with nitrosylated AAT, and IFNγ transcripts were significantly elevated in tumors extracted soon after ischemia-reperfusion challenge. These context-specific changes may explain the differential effects of AAT on immune responses towards tumor cells versus benign antigenic targets. These data suggest that systemically elevated levels of AAT may accommodate its physiological function in inflammatory resolution, without compromising tumor-targeting immune responses.

11.
Int J Cell Biol ; 2016: 9259646, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28083070

RESUMO

Proinflammatory cytokines are potent mediators of numerous biological processes and are tightly regulated in the body. Chronic uncontrolled levels of such cytokines can initiate and derive many pathologies, including incidences of autoimmunity and cancer. Therefore, therapies that regulate the activity of inflammatory cytokines, either by supplementation of anti-inflammatory recombinant cytokines or by neutralizing them by using blocking antibodies, have been extensively used over the past decades. Over the past few years, new innovative biological agents for blocking and regulating cytokine activities have emerged. Here, we review some of the most recent approaches of cytokine targeting, focusing on anti-TNF antibodies or recombinant TNF decoy receptor, recombinant IL-1 receptor antagonist (IL-1Ra) and anti-IL-1 antibodies, anti-IL-6 receptor antibodies, and TH17 targeting antibodies. We discuss their effects as biologic drugs, as evaluated in numerous clinical trials, and highlight their therapeutic potential as well as emphasize their inherent limitations and clinical risks. We suggest that while systemic blocking of proinflammatory cytokines using biological agents can ameliorate disease pathogenesis and progression, it may also abrogate the hosts defense against infections. Moreover, we outline the rational need to develop new therapies, which block inflammatory cytokines only at sites of inflammation, while enabling their function systemically.

12.
Biomed Res Int ; 2015: 184574, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26583093

RESUMO

The traditional model of T helper differentiation describes the naïve T cell as choosing one of several subsets upon stimulation and an added reciprocal inhibition aimed at maintaining the chosen subset. However, to date, evidence is mounting to support the presence of subset plasticity. This is, presumably, aimed at fine-tuning adaptive immune responses according to local signals. Reprograming of cell phenotype is made possible by changes in activation of master transcription factors, employing epigenetic modifications that preserve a flexible mode, permitting a shift between activation and silencing of genes. The acute phase response represents an example of peripheral changes that are critical in modulating T cell responses. α1-antitrypsin (AAT) belongs to the acute phase responses and has recently surfaced as a tolerogenic agent in the context of adaptive immune responses. Nonetheless, AAT does not inhibit T cell responses, nor does it shutdown inflammation per se; rather, it appears that AAT targets non-T cell immunocytes towards changing the cytokine environment of T cells, thus promoting a regulatory T cell profile. The present review focuses on this intriguing two-way communication between innate and adaptive entities, a crosstalk that holds important implications on potential therapies for a multitude of immune disorders.


Assuntos
Proteínas de Fase Aguda/metabolismo , Imunidade Adaptativa , Imunidade Inata , alfa 1-Antitripsina/metabolismo , Proteínas de Fase Aguda/imunologia , Diferenciação Celular/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , alfa 1-Antitripsina/imunologia
13.
Sci Rep ; 5: 14756, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26439902

RESUMO

Environmental signals can be translated into chromatin changes, which alter gene expression. Here we report a novel concept that cells can signal chromatin damage from the nucleus back to the surrounding tissue through the cytokine interleukin-1alpha (IL-1α). Thus, in addition to its role as a danger signal, which occurs when the cytokine is passively released by cell necrosis, IL-1α could directly sense DNA damage and act as signal for genotoxic stress without loss of cell integrity. Here we demonstrate localization of the cytokine to DNA-damage sites and its subsequent secretion. Interestingly, its nucleo-cytosolic shuttling after DNA damage sensing is regulated by histone deacetylases (HDAC) and IL-1α acetylation. To demonstrate the physiological significance of this newly discovered mechanism, we used IL-1α knockout mice and show that IL-1α signaling after UV skin irradiation and DNA damage is important for triggering a sterile inflammatory cascade in vivo that contributes to efficient tissue repair and wound healing.


Assuntos
Dano ao DNA/fisiologia , Imunidade Inata/fisiologia , Inflamação/genética , Interleucina-1alfa/metabolismo , Acetilação , Animais , Linhagem Celular , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Histona Desacetilases/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-1alfa/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pele/metabolismo , Pele/efeitos da radiação
14.
J Immunol ; 195(4): 1705-12, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26157171

RESUMO

Both IL-1α and IL-1ß are highly inflammatory cytokines mediating a wide spectrum of diseases. A recombinant form of the naturally occurring IL-1R antagonist (IL-1Ra), which blocks IL-1R1, is broadly used to treat autoimmune and autoinflammatory diseases; however, blocking IL-1 increases the risk of infection. In this study, we describe the development of a novel form of recombinant IL-1Ra, termed chimeric IL-1Ra. This molecule is a fusion of the N-terminal peptide of IL-1ß and IL-1Ra, resulting in inactive IL-1Ra. Because the IL-1ß N-terminal peptide contains several protease sites clustered around the caspase-1 site, local proteases at sites of inflammation can cleave chimeric IL-1Ra and turn IL-1Ra active. We demonstrate that chimeric IL-1Ra reduces IL-1-mediated inflammation in vitro and in vivo. This unique approach limits IL-1 receptor blockade to sites of inflammation, while sparing a multitude of desired IL-1-related activities, including host defense against infections and IL-1-mediated repair.


Assuntos
Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Receptores de Interleucina-1/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linhagem Celular , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-1beta/química , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Elastase Pancreática/metabolismo , Fragmentos de Peptídeos , Domínios e Motivos de Interação entre Proteínas/genética , Proteólise , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
15.
Immunology ; 2014 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25308894

RESUMO

The autoimmune destruction of pancreatic ß-cells is the hallmark of type 1 diabetes (T1D). Failure of anti-CD3 antibodies to provide long-lasting reversal of T1D and the expression of an NK cell ligand on ß-cells suggest that NK cells play a role in disease pathogenesis. Indeed, killing of ß-cells by NK cells has been shown to occur, mediated by activation of the NK cell activating receptor, NKp46. α1-antitrypsin (AAT), an anti-inflammatory and immunomodulatory glycoprotein, protects ß-cells from injurious immune responses and is currently evaluated as a therapeutic for recent onset T1D. While isolated T lymphocytes are not inhibited by AAT, dendritic cells (DCs) become tolerogenic in its presence and other innate immune cells become less inflammatory. Yet a comprehensive profile of NK cell responses in the presence of AAT has yet to be described. In the present study, we demonstrate that AAT significantly reduces NK cell degranulation against ß-cells, albeit in the whole animal and not in isolated NK cell cultures. AAT-treated mice, and not isolated cultured ß-cells, exhibited a marked reduction in NKp46 ligand levels on ß-cells. In related experiments, AAT-treated DCs exhibited reduced inducible DC-expressed IL-15 levels and evoked a weaker NK cell response. NK cell depletion in a T1D mouse model resulted in improved ß-cell function and survival, similar to the effects observed by AAT treatment alone; nonetheless, the two approaches were non-synergistic. Our data suggest that AAT is a selective immunomodulator that retains pivotal NK cell responses, while diverting their activities away from islet ß-cells. This article is protected by copyright. All rights reserved.

16.
Cell Mol Immunol ; 11(4): 377-86, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25000533

RESUMO

Although islet transplantation for individuals with type 1 diabetes has been shown to yield superior blood glucose control, it remains inadequate for long-term control. This is partly due to islet injuries and stresses that can lead to beta cell loss. Inhibition of excess IL-1ß activity might minimize islet injuries, thus preserving function. The IL-1 receptor antagonist (IL-1Ra), an endogenous inhibitor of IL-1ß, protects islets from cytokine-induced necrosis and apoptosis. Therefore, an imbalance between IL-1ß and IL-1Ra might influence the courses of allogeneic and autoimmune responses to islets. Our group previously demonstrated that the circulating serine-protease inhibitor human alpha-1-antitrypsin (hAAT), the levels of which increase in circulation during acute-phase immune responses, exhibits anti-inflammatory and islet-protective properties, as well as immunomodulatory activity. In the present study, we sought to determine whether the pancreatic islet allograft-protective activity of hAAT was mediated by IL-1Ra induction. Our results demonstrated that hAAT led to a 2.04-fold increase in IL-1Ra expression in stimulated macrophages and that hAAT-pre-treated islet grafts exhibited a 4.851-fold increase in IL-1Ra transcript levels, which were associated with a moderate inflammatory profile. Unexpectedly, islets that were isolated from IL-1Ra-knockout mice and pre-treated with hAAT before grafting into wild-type mice yielded an increase in intragraft IL-1Ra expression that was presumably derived from infiltrating host cells, albeit in the absence of hAAT treatment of the host. Indeed, hAAT-pre-treated islets generated hAAT-free conditioned medium that could induce IL-1Ra production in cultured macrophages. Finally, we demonstrated that hAAT promoted a distinct phosphorylation and nuclear translocation pattern for p65, a key transcription factor required for IL-1Ra expression.


Assuntos
Rejeição de Enxerto/prevenção & controle , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Transplante das Ilhotas Pancreáticas , Macrófagos/efeitos dos fármacos , Complicações Pós-Operatórias/prevenção & controle , Fator de Transcrição RelA/metabolismo , alfa 1-Antitripsina/administração & dosagem , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Células Cultivadas , Rejeição de Enxerto/etiologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1/genética , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fator de Transcrição RelA/genética , Regulação para Cima/efeitos dos fármacos
17.
Gut ; 63(4): 598-609, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23793223

RESUMO

OBJECTIVE: The differential role of the IL-1 agonists, IL-1α, which is mainly cell-associated versus IL-1ß, which is mostly secreted, was studied in colon inflammation. DESIGN: Dextran sodium sulfate (DSS) colitis was induced in mice globally deficient in either IL-1α or IL-1ß, and in wild-type mice, or in mice with conditional deletion of IL-1α in intestinal epithelial cells (IECs). Bone marrow transplantation experiments were performed to assess the role of IL-1α or IL-1ß of myeloid versus colon non-hematopoietic cells in inflammation and repair in acute colitis. RESULTS: IL-1α released from damaged IECs acts as an alarmin by initiating and propagating colon inflammation, as IL-1α deficient mice exhibited mild disease symptoms with improved recovery. IL-1ß is involved in repair of IECs and reconstitution of the epithelial barrier during the resolution of colitis; its deficiency correlates with disease exacerbation. Neutralisation of IL-1α in control mice during acute colitis led to alleviation of clinical and histological manifestations, whereas treatment with rIL-1Ra or anti-IL-1ß antibodies was not effective. Repair after colitis correlated with accumulation of CD8 and regulatory T cells in damaged crypts. CONCLUSIONS: The role of IL-1α and IL-1ß differs in DSS-induced colitis in that IL-1α, mainly of colon epithelial cells is inflammatory, whereas IL-1ß, mainly of myeloid cell origin, promotes healing and repair. Given the dissimilar functions of each IL-1 agonistic molecule, an IL-1 receptor blockade would not be as therapeutically effective as specific neutralising of IL-1α, which leaves IL-1ß function intact.


Assuntos
Colite/fisiopatologia , Interleucina-1alfa/fisiologia , Interleucina-1beta/fisiologia , Animais , Colite/induzido quimicamente , Colite/patologia , Colo/efeitos dos fármacos , Colo/patologia , Colo/fisiopatologia , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Interleucina-1/agonistas , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia , Infiltração Leucêmica/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/fisiologia
18.
Semin Immunol ; 25(6): 430-8, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24183701

RESUMO

Although the IL-1α molecule has long been recognized, information about its distinct role in various diseases is limited, since most clinical studies have focused on the role of IL-1ß. Despite triggering the same IL-1 receptor as does IL-1ß, there is, however, a distinct role for IL-1α in some inflammatory diseases. IL-1α is a unique cytokine since it is constitutively present intracellularly in nearly all resting non-hematopoietic cells in health as well as being up-regulated during hypoxia. During cell necrosis, IL-1α functions as an alarm molecule and thus plays a critical role early in inflammation. Following its release from damage tissue cells, IL-1α mediates neutrophil recruitment to the site of injury, inducing IL-1ß, other cytokines and chemokines from surrounding resident cells. Another unique attribute of IL-1α is its nuclear localization sequence present in the N-terminal half of the precursor termed the propiece. The IL-1α propiece translocates into the nucleus and participates in the regulation of transcription. Therefore, IL-1α, like IL-1 family members IL-33 and IL-37, is a 'dual-function' cytokine binding to chromatin as well as to its cell surface receptor. Some cancer cells can express membrane IL-1α, which can increase immunogenicity of tumor cells and serve in anti-tumor immune surveillance and tumor regression. However, in the tumor microenvironment, precursor IL-1α released from dying tumor cells is inflammatory and, similar to IL-1ß, increases tumor invasiveness and angiogenesis.


Assuntos
Inflamação/imunologia , Interleucina-1alfa/imunologia , Animais , Hipóxia Celular/imunologia , Transformação Celular Neoplásica/patologia , Cromatina , Humanos , Interleucina-1alfa/genética , Camundongos , Necrose/patologia , Invasividade Neoplásica/patologia , Neoplasias/imunologia , Infiltração de Neutrófilos/imunologia , Receptores de Interleucina-1/imunologia
19.
Front Immunol ; 4: 177, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23847618

RESUMO

Interleukin-1 (IL-1) is a major "alarm" upstream pro-inflammatory cytokine that also affects immunity and hematopoiesis by inducing cytokine cascades. In the tumor arena, IL-1 is produced by malignant or microenvironmental cells. As a pleiotropic cytokine, IL-1 is involved in tumorigenesis and tumor invasiveness but also in the control of anti-tumor immunity. IL-1α and IL-1ß are the major agonists of IL-1, while IL-1Ra is a physiological inhibitor of pre-formed IL-1. In their secreted form, IL-1α and IL-1ß bind to the same receptors and induce the same biological functions, but IL-1α and IL-1ß differ in their compartmentalization within the producing cell or the microenvironment. IL-1ß is only active in its processed, secreted form, and mediates inflammation, which promotes carcinogenesis, tumor invasiveness, and immunosuppression, whereas IL-1α is mainly cell-associated and in the tumor context, when expressed on the cell membrane, it stimulates anti-tumor cell immunity manifested by tumor regression. In the tumor milieu, extracellular levels of IL-1α are usually low and do not stimulate broad inflammation that promotes progression. Immunosuppression induced by IL-1ß in the tumor microenvironment, mainly through MDSC induction, usually inhibits or masks anti-tumor cell immunity induced by cell-associated IL-1α. However, in different tumor systems, redundant or unique patterns of IL-1α and IL-1ß expression and function have been observed. Recent breakthroughs in inflammasome biology and IL-1ß processing/secretion have spurred the development of novel anti-IL-1 agents, which are being used in clinical trials in patients with diverse inflammatory diseases. Better understanding of the integrative role of IL-1α and IL-1ß in distinct malignancies will facilitate the application of novel IL-1 modulation approaches at the bedside, in cancer patients with minimal residual disease (MRD), as an adjunct to conventional approaches to reduce the tumor burden.

20.
J Immunol ; 190(7): 3500-9, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23475218

RESUMO

In this study, we assessed the involvement of IL-1ß in early angiogenic responses induced by malignant cells using Matrigel plugs supplemented with B16 melanoma cells. We found that during the angiogenic response, IL-1ß and vascular endothelial growth factor (VEGF) interact in a newly described autoinduction circuit, in which each of these cytokines induces the other. The IL-1ß and VEGF circuit acts through interactions between bone marrow-derived VEGF receptor 1(+)/IL-1R1(+) immature myeloid cells and tissue endothelial cells. Myeloid cells produce IL-1ß and additional proinflammatory cytokines, which subsequently activate endothelial cells to produce VEGF and other proangiogenic factors and provide the inflammatory microenvironment for angiogenesis and tumor progression. These mechanisms were also observed in a nontumor early angiogenic response elicited in Matrigel plugs by either rIL-1ß or recombinant VEGF. We have shown that IL-1ß inhibition stably reduces tumor growth by limiting inflammation and inducing the maturation of immature myeloid cells into M1 macrophages. In sharp contrast, only transient inhibition of tumor growth was observed after VEGF neutralization, followed by tumor recurrence mediated by rebound angiogenesis. This occurs via the reprogramming of VEGF receptor 1(+)/IL-1R1(+) cells to express hypoxia inducible factor-1α, VEGF, and other angiogenic factors, thereby directly supporting proliferation of endothelial cells and blood vessel formation in a paracrine manner. We suggest using IL-1ß inhibition as an effective antitumor therapy and are currently optimizing the conditions for its application in the clinic.


Assuntos
Interleucina-1beta/metabolismo , Melanoma Experimental/metabolismo , Neovascularização Patológica/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Citocinas/farmacologia , Progressão da Doença , Expressão Gênica , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Melanoma Experimental/genética , Camundongos , Camundongos Knockout , Células Mieloides/metabolismo , Neovascularização Patológica/genética , Fenótipo , Microambiente Tumoral/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...