Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38612760

RESUMEN

IL-1α is a dual function cytokine that affects inflammatory and immune responses and plays a pivotal role in cancer. The effects of intracellular IL-1α on the development of triple negative breast cancer (TNBC) in mice were assessed using the CRISPR/Cas9 system to suppress IL-1α expression in 4T1 breast cancer cells. Knockout of IL-1α in 4T1 cells modified expression of multiple genes, including downregulation of cytokines and chemokines involved in the recruitment of tumor-associated pro-inflammatory cells. Orthotopical injection of IL-1α knockout (KO) 4T1 cells into BALB/c mice led to a significant decrease in local tumor growth and lung metastases, compared to injection of wild-type 4T1 (4T1/WT) cells. Neutrophils and myeloid-derived suppressor cells were abundant in tumors developing after injection of 4T1/WT cells, whereas more antigen-presenting cells were observed in the tumor microenvironment after injection of IL-1α KO 4T1 cells. This switch correlated with increased infiltration of CD3+CD8+ and NKp46+cells. Engraftment of IL-1α knockout 4T1 cells into immunodeficient NOD.SCID mice resulted in more rapid tumor growth, with increased lung metastasis in comparison to engraftment of 4T1/WT cells. Our results suggest that tumor-associated IL-1α is involved in TNBC progression in mice by modulating the interplay between immunosuppressive pro-inflammatory cells vs. antigen-presenting and cytotoxic cells.


Asunto(s)
Neoplasias Pulmonares , Neoplasias de la Mama Triple Negativas , Animales , Ratones , Humanos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias de la Mama Triple Negativas/genética , Microambiente Tumoral , Neoplasias Pulmonares/genética , Interleucina-1alfa/genética
2.
PLoS Biol ; 22(1): e3002486, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38236896

RESUMEN

Acute gastrointestinal infection with intracellular pathogens like Salmonella Typhimurium triggers the release of the proinflammatory cytokine interleukin 1ß (IL-1ß). However, the role of IL-1ß in intestinal defense against Salmonella remains unclear. Here, we show that IL-1ß production is detrimental during Salmonella infection. Mice lacking IL-1ß (IL-1ß -/-) failed to recruit neutrophils to the gut during infection, which reduced tissue damage and prevented depletion of short-chain fatty acid (SCFA)-producing commensals. Changes in epithelial cell metabolism that typically support pathogen expansion, such as switching energy production from fatty acid oxidation to fermentation, were absent in infected IL-1ß -/- mice which inhibited Salmonella expansion. Additionally, we found that IL-1ß induces expression of complement anaphylatoxins and suppresses the complement-inactivator carboxypeptidase N (CPN1). Disrupting this process via IL-1ß loss prevented mortality in Salmonella-infected IL-1ß -/- mice. Finally, we found that IL-1ß expression correlates with expression of the complement receptor in patients suffering from sepsis, but not uninfected patients and healthy individuals. Thus, Salmonella exploits IL-1ß signaling to outcompete commensal microbes and establish gut colonization. Moreover, our findings identify the intersection of IL-1ß signaling and the complement system as key host factors involved in controlling mortality during invasive Salmonellosis.


Asunto(s)
Interleucina-1beta , Infecciones por Salmonella , Animales , Humanos , Ratones , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Virulencia
3.
Colloids Surf B Biointerfaces ; 223: 113173, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36724562

RESUMEN

Genistein, an isoflavone from soybean, has attracted attention due to its health benefits, particularly antioxidant and anti-inflammatory activities. Clinical applications of genistein, however, have been limited due to the considerable hydrophobicity and lower bioavailability of the molecule. In this study, carbon dots (C-dots) synthesized from genistein as the carbonaceous precursor exhibit antioxidant properties in test-tube and cell experiments. Anti-inflammatory activity of the genistein-C-dots was also recorded in LPS stimulated macrophages, manifested in inhibition of pro-inflammatory cytokine levels and enhancement anti-inflammatory cytokine expression. The antioxidant and anti-inflammatory effects of the genistein-C-dots, particularly in comparison to the parent genistein molecules, likely account to the display of functional genistein residues on the C-dots' surfaces, and low band gap energy facilitating electron scavenging. Importantly, the genistein-C-dots featured biocompatibility and low cytotoxicity, underlining their potential as a therapeutic vehicle against inflammatory conditions.


Asunto(s)
Antioxidantes , Genisteína , Genisteína/química , Antioxidantes/farmacología , Glycine max/química , Antiinflamatorios/farmacología , Citocinas/metabolismo
4.
J Innate Immun ; 15(1): 531-547, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36809756

RESUMEN

Probiotic fermented foods are perceived as contributing to human health; however, solid evidence for their presumptive therapeutic systemic benefits is generally lacking. Here we report that tryptophol acetate and tyrosol acetate, small-molecule metabolites secreted by the probiotic milk-fermented yeast Kluyveromyces marxianus, inhibit hyperinflammation (e.g., "cytokine storm"). Comprehensive in vivo and in vitro analyses, employing LPS-induced hyperinflammation models, reveal dramatic effects of the molecules, added in tandem, on mice morbidity, laboratory parameters, and mortality. Specifically, we observed attenuated levels of the proinflammatory cytokines IL-6, IL-1α, IL-1ß, and TNF-α and reduced reactive oxygen species. Importantly, tryptophol acetate and tyrosol acetate did not completely suppress proinflammatory cytokine generation, rather brought their concentrations back to baseline levels, thus maintaining core immune functions, including phagocytosis. The anti-inflammatory effects of tryptophol acetate and tyrosol acetate were mediated through downregulation of TLR4, IL-1R, and TNFR signaling pathways and increased A20 expression, leading to NF-kB inhibition. Overall, this work illuminates phenomenological and molecular details underscoring anti-inflammatory properties of small molecules identified in a probiotic mixture, pointing to potential therapeutic avenues against severe inflammation.


Asunto(s)
Citocinas , Probióticos , Animales , Humanos , Ratones , Citocinas/metabolismo , Antiinflamatorios , Probióticos/farmacología
5.
Immunology ; 168(2): 203-216, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35462425

RESUMEN

In recent years, pro-oncogenic mechanisms of the tumour microenvironment (ТМЕ) have been actively discussed. One of the main cytokines of the TМЕ is interleukin-1 beta (IL-1ß), which exhibits proinflammatory properties. Some studies have shown an association between an increase in IL-1ß levels and tumour progression. The purpose of this review is to analyse the pathogenic mechanisms induced by IL-1ß in the TМЕ, as well as the diagnostic significance of the presence of IL-1ß in patients with cancer and the efficacy of treatment with IL-1ß inhibitors. According to the literature, IL-1ß can induce an increase in tumour angiogenesis due to its effects on the differentiation of epithelial cells, pro-angiogenic molecule secretion and expression of adhesion molecules, thus increasing tumour growth and metastasis. IL-1ß is also involved in the suppression of anti-tumour immune responses. The expression and secretion of IL-1ß has been noted in various types of tumours. In some clinical studies, an elevated level of IL-1ß was found to be associated with low efficacy of anti-cancer therapy and a poor prognosis. In most experimental and clinical studies, the use of IL-1ß inhibitors contributed to a decrease in tumour mass and an increase in the response to anti-tumour drugs.


Asunto(s)
Relevancia Clínica , Neoplasias , Humanos , Citocinas , Interleucina-1beta , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
6.
Cancer Immunol Res ; 10(12): 1525-1541, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36206577

RESUMEN

During melanoma metastasis, tumor cells originating in the skin migrate via lymphatic vessels to the sentinel lymph node (sLN). This process facilitates tumor cell spread across the body. Here, we characterized the innate inflammatory response to melanoma in the metastatic microenvironment of the sLN. We found that macrophages located in the subcapsular sinus (SS) produced protumoral IL1α after recognition of tumoral antigens. Moreover, we confirmed that the elimination of LN macrophages or the administration of an IL1α-specific blocking antibody reduced metastatic spread. To understand the mechanism of action of IL1α in the context of the sLN microenvironment, we applied single-cell RNA sequencing to microdissected metastases obtained from animals treated with the IL1α-specific blocking antibody. Among the different pathways affected, we identified STAT3 as one of the main targets of IL1α signaling in metastatic tumor cells. Moreover, we found that the antitumoral effect of the anti-IL1α was not mediated by lymphocytes because Il1r1 knockout mice did not show significant differences in metastasis growth. Finally, we found a synergistic antimetastatic effect of the combination of IL1α blockade and STAT3 inhibition with stattic, highlighting a new immunotherapy approach to preventing melanoma metastasis.


Asunto(s)
Vasos Linfáticos , Melanoma , Ganglio Linfático Centinela , Neoplasias Cutáneas , Animales , Ratones , Biopsia del Ganglio Linfático Centinela , Ganglio Linfático Centinela/patología , Metástasis Linfática/patología , Melanoma/patología , Macrófagos/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Ganglios Linfáticos/patología , Neoplasias Cutáneas/patología , Microambiente Tumoral
7.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35292516

RESUMEN

BACKGROUND: Although the mitogen-activated protein kinases (MAPK) pathway is hyperactive in head and neck cancer (HNC), inhibition of MEK1/2 in HNC patients has not shown clinically meaningful activity. Therefore, we aimed to characterize the effect of MEK1/2 inhibition on the tumor microenvironment (TME) of MAPK-driven HNC, elucidate tumor-host interaction mechanisms facilitating immune escape on treatment, and apply rationale-based therapy combination immunotherapy and MEK1/2 inhibitor to induce tumor clearance. METHODS: Mouse syngeneic tumors and xenografts experiments were used to analyze tumor growth in vivo. Single-cell cytometry by time of flight, flow cytometry, and tissue stainings were used to profile the TME in response to trametinib (MEK1/2 inhibitor). Co-culture of myeloid-derived suppressor cells (MDSC) with CD8+ T cells was used to measure immune suppression. Overexpression of colony-stimulating factor-1 (CSF-1) in tumor cells was used to show the effect of tumor-derived CSF-1 on sensitivity to trametinib and anti-programmed death- 1 (αPD-1) in mice. In HNC patients, the ratio between CSF-1 and CD8A was measured to test the association with clinical benefit to αPD-1 and αPD-L1 treatment. RESULTS: Using preclinical HNC models, we demonstrated that treatment with trametinib delays HNC initiation and progression by reducing tumor cell proliferation and enhancing the antitumor immunity of CD8+ T cells. Activation of CD8+ T cells by supplementation with αPD-1 antibody eliminated tumors and induced an immune memory in the cured mice. Mechanistically, an early response to trametinib treatment sensitized tumors to αPD-1-supplementation by attenuating the expression of tumor-derived CSF-1, which reduced the abundance of two CSF-1R+CD11c+ MDSC populations in the TME. In contrast, prolonged treatment with trametinib abolished the antitumor activity of αPD-1, because tumor cells undergoing the epithelial to mesenchymal transition in response to trametinib restored CSF-1 expression and recreated an immune-suppressive TME. CONCLUSION: Our findings provide the rationale for testing the trametinib/αPD-1 combination in HNC and highlight the importance of sensitizing tumors to αPD-1 by using MEK1/2 to interfere with the tumor-host interaction. Moreover, we describe the concept that treatment of cancer with a targeted therapy transiently induces an immune-active microenvironment, and supplementation of immunotherapy during this time further activates the antitumor machinery to cause tumor elimination.


Asunto(s)
Neoplasias de Cabeza y Cuello , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Inmunoterapia , Ratones
8.
Sci Rep ; 11(1): 7676, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33828163

RESUMEN

IL-2 is the master-regulator cytokine for T cell dependent responses and is crucial for proliferation and survival of T cells. However, IL-2-based treatments remained marginal, in part due to short half-life. Thus, we aimed to extend IL-2 half-life by flanking the IL-2 core with sequences derived from the extensively glycosylated hinge region of the NCR2 receptor. We termed this modified IL-2: "S2A". Importantly, S2A blood half-life was extended 14-fold compared to the clinical grade IL-2, Proleukin. Low doses inoculation of S2A significantly enhanced induction of Tregs (CD4+ Regulatory T cells) in vivo, as compared to Proleukin, while both S2A and Proleukin induced low levels of CD8+ T cells. In a B16 metastatic melanoma model, S2A treatment was unable to reduce the metastatic capacity of B16 melanoma, while enhancing induction and recruitment of Tregs, compared to Proleukin. Conversely, in two autoimmune models, rheumatoid arthritis and DSS-induced colitis, S2A treatment significantly reduced the progression of disease compared to Proleukin. Our results suggest new avenues for generating long-acting IL-2 for long-standing treatment and a new technique for manipulating short-life proteins for clinical and research uses.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Interleucina-2/análogos & derivados , Receptor 2 Gatillante de la Citotoxidad Natural/química , Linfocitos T Reguladores/efectos de los fármacos , Animales , Artritis Reumatoide/prevención & control , Preparaciones de Acción Retardada , Evaluación Preclínica de Medicamentos , Glicosilación , Semivida , Interleucina-2/administración & dosificación , Interleucina-2/farmacocinética , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Mol Cancer Res ; 17(11): 2331-2340, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31409628

RESUMEN

Multiple myeloma (MM) is a plasma cell neoplasia commonly treated with proteasome inhibitors such as bortezomib. Although bortezomib has demonstrated enhanced survival benefit, some patients relapse and subsequently develop resistance to such therapy. Here, we investigate the mechanisms underlying relapse and refractory MM following bortezomib treatment. We show that bortezomib-exposed proinflammatory macrophages promote an enrichment of MM-tumor-initiating cells (MM-TIC) both in vitro and in vivo. These effects are regulated in part by IL1ß, as blocking the IL1ß axis by a pharmacologic or genetic approach abolishes bortezomib-induced MM-TIC enrichment. In MM patients treated with bortezomib, high proinflammatory macrophages in the bone marrow negatively correlate with survival rates (HR, 1.722; 95% CI, 1.138-2.608). Furthermore, a positive correlation between proinflammatory macrophages and TICs in the bone marrow was also found. Overall, our results uncover a protumorigenic cross-talk involving proinflammatory macrophages and MM cells in response to bortezomib therapy, a process that enriches the MM-TIC population. IMPLICATIONS: Our findings suggest that proinflammatory macrophages in bone marrow biopsies represent a potential prognostic biomarker for acquired MM resistance to bortezomib therapy.


Asunto(s)
Antineoplásicos/farmacología , Bortezomib/farmacología , Resistencia a Antineoplásicos , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteasoma/farmacología , Adulto , Animales , Biopsia , Médula Ósea/patología , Línea Celular Tumoral , Femenino , Humanos , Macrófagos/patología , Ratones , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Células Madre Neoplásicas/patología , Recurrencia , Adulto Joven
10.
Proc Natl Acad Sci U S A ; 116(4): 1361-1369, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30545915

RESUMEN

Interleukin-1ß (IL-1ß) is abundant in the tumor microenvironment, where this cytokine can promote tumor growth, but also antitumor activities. We studied IL-1ß during early tumor progression using a model of orthotopically introduced 4T1 breast cancer cells. Whereas there is tumor progression and spontaneous metastasis in wild-type (WT) mice, in IL-1ß-deficient mice, tumors begin to grow but subsequently regress. This change is due to recruitment and differentiation of inflammatory monocytes in the tumor microenvironment. In WT mice, macrophages heavily infiltrate tumors, but in IL-1ß-deficient mice, low levels of the chemokine CCL2 hamper recruitment of monocytes and, together with low levels of colony-stimulating factor-1 (CSF-1), inhibit their differentiation into macrophages. The low levels of macrophages in IL-1ß-deficient mice result in a relatively high percentage of CD11b+ dendritic cells (DCs) in the tumors. In WT mice, IL-10 secretion from macrophages is dominant and induces immunosuppression and tumor progression; in contrast, in IL-1ß-deficient mice, IL-12 secretion by CD11b+ DCs prevails and supports antitumor immunity. The antitumor immunity in IL-1ß-deficient mice includes activated CD8+ lymphocytes expressing IFN-γ, TNF-α, and granzyme B; these cells infiltrate tumors and induce regression. WT mice with 4T1 tumors were treated with either anti-IL-1ß or anti-PD-1 Abs, each of which resulted in partial growth inhibition. However, treating mice first with anti-IL-1ß Abs followed by anti-PD-1 Abs completely abrogated tumor progression. These data define microenvironmental IL-1ß as a master cytokine in tumor progression. In addition to reducing tumor progression, blocking IL-1ß facilitates checkpoint inhibition.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Antígeno CD11b/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Factores Estimulantes de Colonias/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Granzimas/farmacología , Humanos , Terapia de Inmunosupresión/métodos , Inflamación/metabolismo , Interferón gamma/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
12.
mSystems ; 3(3)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29766049

RESUMEN

Inflammatory bowel diseases (IBD) are a group of chronic inflammatory disorders of the intestine, with as-yet-unclear etiologies, affecting over a million people in the United States alone. With the emergence of microbiome research, numerous studies have shown a connection between shifts in the gut microbiota composition (dysbiosis) and patterns of IBD development. In a previous study, we showed that interleukin 1α (IL-1α) deficiency in IL-1α knockout (KO) mice results in moderate dextran sodium sulfate (DSS)-induced colitis compared to that of wild-type (WT) mice, characterized by reduced inflammation and complete healing, as shown by parameters of weight loss, disease activity index (DAI) score, histology, and cytokine expression. In this study, we tested whether the protective effects of IL-1α deficiency on DSS-induced colitis correlate with changes in the gut microbiota and whether manipulation of the microbiota by cohousing can alter patterns of colon inflammation. We analyzed the gut microbiota composition in both control (WT) and IL-1α KO mice under steady-state homeostasis, during acute DSS-induced colitis, and after recovery using 16S rRNA next-generation sequencing. Additionally, we performed cohousing of both mouse groups and tested the effects on the microbiota and clinical outcomes. We demonstrate that host-derived IL-1α has a clear influence on gut microbiota composition, as well as on severity of DSS-induced acute colon inflammation. Cohousing both successfully changed the gut microbiota composition and increased the disease severity of IL-1α-deficient mice to levels similar to those of WT mice. This study shows a strong and novel correlation between IL-1α expression, microbiota composition, and clinical outcomes of DSS-induced colitis. IMPORTANCE Here, we show a connection between IL-1α expression, microbiota composition, and clinical outcomes of DSS-induced colitis. Specifically, we show that the mild colitis symptoms seen in IL-1α-deficient mice following administration of DSS are correlated with the unique gut microbiota compositions of the mice. However, when these mice are exposed to WT microbiota by cohousing, their gut microbiota composition returns to resemble that of WT mice, and their disease severity increases significantly. As inflammatory bowel diseases are such common diseases, with limited effective treatments to date, there is a great need to better understand the interactions between microbiota composition, the immune system, and colitis. This study shows correlation between microbiota composition and DSS resistance; it may potentially lead to the development of improved probiotics for IBD treatment.

13.
Curr Pharm Des ; 23(32): 4893-4905, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28606052

RESUMEN

The importance of anti-tumor immunity in the outcome of cancer is now unequivocally established and recent achivements in the field have stimulated the development of new immunotherapeutical approaches. In invasive tumors, widespread inflammation promotes invasiveness and concomitantly also inhibits anti-tumor immune responses. We suggest that efficient tumor treatment should target both the malignant cells and the tumor microenvironment. Interleukin-1 (IL-1) is a pro-inflammatory as well as an immunostimulatory cytokine that is abundant in the tumor microenvironment. Manipulation of IL-1 can thus serve as an immunotherapeutical approach to reduce inflammation/immunosuppression and thus enhance anti-tumor immunity. The two major IL-1 agonistic molecules are IL-1α and IL-1ß, which bind to the same IL-1 signaling receptor and induce the same array of biological activities. The IL-1 receptor antagonist (IL-Ra) is a physiological inhibitor of IL-1 that binds to its receptor without transmition of activation signals and thus serves as a decoy target. We have demonstrated that IL-1α and IL-1ß are different in terms of the producing cells and their compartmentalization and the amount. IL-1α is mainly expressed intracellularly, in the cytosol, in the nucleus or exposed on the cell membrane, however, it is rarely secreted. IL-1ß is active only as a secreted molecule that is mainly produced by activated myeloid cells. We have shown different functions of IL-1α and IL-1ß in the malignant process. Thus, in its membrane- associated form, IL-1α is mainly immunostimulatory, while IL-1ß that is secreted into the tumor microenvironment is mainly pro-inflammatory and promotes tumorigenesis, tumor invasiveness and immunosuppression. These distinct functions of the IL-1 agonistic molecules are mainly manifested in early stages of tumor development and the patterns of their expression dictate the direction of the malignant process. Here, we suggest that IL-1 modulation can serve as an effective mean to tilt the balance between inflammation and immunity in tumor sites, towards the latter. Different agents that neutralize IL-1, mainly the IL-Ra and specific antibodies, exist. They are safe and FDA-approved. The IL-1Ra has been widely and successfully used in patients with Rheumatoid arthritis, autoinflammatory diseases and various other diseases that have an inflammatory component. Here, we provide the rationale and experimental evidence for the use of anti-IL-1 agents in cancer patients, following first line therapy to debulk the major tumor's mass. The considerations and constraints of using anti-IL-1 treatments in cancer are also discussed. We hope that this review will stimulate studies that will fasten the application of IL-1 neutralization at the bedside of cancer patients.


Asunto(s)
Interleucina-1/antagonistas & inhibidores , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Humanos , Inmunoterapia/métodos , Inflamación/inmunología , Inflamación/terapia , Interleucina-1/inmunología , Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Invasividad Neoplásica/inmunología , Neoplasias/inmunología , Neoplasias/patología
14.
J Leukoc Biol ; 102(2): 293-306, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28522598

RESUMEN

IL-1 is a pleiotropic cytokine that controls inflammation, immunity, and hemopoiesis. The major IL-1 agonistic molecules are IL-1α and IL-1ß, which bind to IL-1R type I (IL-1R1) and induce similar biologic functions. The IL-1R antagonist (IL-1Ra) is a physiologic inhibitor of IL-1R1 signaling. In the tumor microenvironment, IL-1 is expressed by malignant, stromal, and infiltrating cells and supports tumor invasiveness and progression. We have shown that in the tumor microenvironment, the IL-1 agonistic molecules act different as a result of their local amounts and their compartmentalization within the producing cells. IL-1ß is produced mainly by myeloid cells upon inflammatory stimulation and is active as a mature, secreted molecule. The precursor of IL-1α (ProIL-1α) is biologically active; it is constitutively expressed in diverse tissue cells in basal levels, and its expression increases during stress or inflammation. ProIL-1α is mainly located in the cytosol or it is membrane associated. ProIL-1α also translocates into the nucleus and binds to chromatin. ProIL-1α is rarely actively secreted but is released from necrotizing tissues and serves as "alarmin" for initiation of inflammation. In the tumor microenvironment, IL-1ß promotes tumorigenesis, tumor invasiveness, and immunosuppression. On the other hand, membrane-associated forms of IL-1α support the development of anti-tumor immunity. In cancer patients, both IL-1 agonistic molecules coexist and interact with each other. Here, we discuss the role of IL-1 agonistic molecules in tumor progression and their potential to serve as targets in anti-tumor immunotherapeutic approaches. Our notion on the optimal conditions for IL-1 manipulation is also discussed.


Asunto(s)
Inmunoterapia/métodos , Interleucina-1/inmunología , Microambiente Tumoral/inmunología , Animales , Humanos , Interleucina-1/antagonistas & inhibidores
15.
J Immunol ; 198(4): 1395-1402, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28167650

RESUMEN

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.


Asunto(s)
Alarminas/fisiología , Estrés Fisiológico , Animales , Proteína HMGB1/metabolismo , Humanos , Inflamación , Interleucina-16/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-33/metabolismo , Ratones , Procesamiento Proteico-Postraduccional , Estrés Fisiológico/inmunología , Estrés Fisiológico/fisiología
16.
Oncotarget ; 7(22): 31847-61, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-26870993

RESUMEN

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy characterized by extremely heterogeneous molecular and biologic abnormalities that hamper the development of effective targeted treatment modalities. While AML cells are highly sensitive to cytotoxic Ca2+ overload, the feasibility of Ca2+- targeted therapy of this disease remains unclear. Here, we show that apoptotic response of AML cells to the synergistically acting polyphenols curcumin (CUR) and carnosic acid (CA), combined at low, non-cytotoxic doses of each compound was mediated solely by disruption of cellular Ca2+ homeostasis. Specifically, activation of caspase cascade in CUR+CA-treated AML cells resulted from sustained elevation of cytosolic Ca2+ (Ca2+cyt) and was not preceded by endoplasmic reticulum stress or mitochondrial damage. The CUR+CA-induced Ca2+cyt rise did not involve excessive influx of extracellular Ca2+ but, rather, occurred due to massive Ca2+ release from intracellular stores concomitant with inhibition of Ca2+cyt extrusion through the plasma membrane. Notably, the CUR+CA combination did not alter Ca2+ homeostasis and viability in non-neoplastic hematopoietic cells, suggesting its cancer-selective action. Most importantly, co-administration of CUR and CA to AML-bearing mice markedly attenuated disease progression in two animal models. Collectively, our results provide the mechanistic and translational basis for further characterization of this combination as a prototype of novel Ca2+-targeted pharmacological tools for the treatment of AML.


Asunto(s)
Abietanos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Curcumina/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Caspasas/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Células HL-60 , Homeostasis , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones Endogámicos C57BL , Ratones SCID , Factores de Tiempo , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Microenviron ; 8(3): 187-200, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26686225

RESUMEN

Interleukin-1 (IL-1) is a major "alarm" upstream pro-inflammatory cytokine that mainly acts by inducing cascades of cytokine and inflammation-promoting mediators. In the tumor arena, IL-1 is produced by both malignant and microenvironmental cells. IL-1α and IL-1ß are the major agonists of IL-1, while IL-1Ra is a physiological inhibitor of pre-formed IL-1. IL-1α and IL-1ß differ in their compartmentalization and in the producing cells. IL-1ß is only active in its inflammasome dependent processed and secreted form and has been considered as the major mediator of inflammation. On the other hand, IL-1α is mainly cell-associated in tissue resident cells, being also active in its precursor form. The role of the IL-1 molecules in the unique microenvironment in the colon is largely unknown. Here, we described the role of IL-1α and IL-1ß in colon homeostasis, colon inflammation, colon carcinogenesis and invasiveness of colorectal cancer. Understanding of the integrative role of IL-1α and IL-1ß in these processes will facilitate the application of novel IL-1 modulating approaches.

18.
J Control Release ; 217: 102-12, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26297207

RESUMEN

There is currently no effective means to prevent or control metastatic dissemination of cancer cells. E-selectin, an adhesion molecule expressed exclusively on inflamed and angiogenic blood vessels, plays an important role in several rate-limiting steps of cancer metastasis. In this study, we assessed the in vivo antitumor efficacy of N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers conjugated to an E-selectin binding peptide (Esbp, DITWDQLWDLMK) and equipped with the chemotherapeutic drug doxorubicin (P-(Esbp)-DOX) or with the proapoptotic peptide D(KLAKLAK)2 (P-(Esbp)-KLAK). Following a single intravenous injection, P-(Esbp)-DOX reduced tumor growth rate and prolonged the survival of mice bearing primary Lewis lung carcinoma (3LL) tumors significantly more than treatment with a non-targeted copolymer (P-DOX) or with free DOX. In an experimental B16-F10 lung metastasis model, a single intravenous dose of P-(Esbp)-DOX or P-(Esbp)-KLAK prolonged mice survival time significantly more than the non-targeted copolymers or the free drugs, and the percentage of complete tumor regression increased with increasing doses and with dosing frequency. In addition, mice pretreated with an E-selectin-targeted "drug-free" copolymer (P-(Esbp)-FITC) exhibited significantly fewer B16-F10 tumor foci in the lungs as compared with non-treated mice, demonstrating the anti-metastatic properties of the copolymer and its ability to control cancer spread through E-selectin-mediated interactions. Biodistribution analysis further confirmed the preferential accumulation of the E-selectin-targeted near-infrared fluorescently-labeled copolymer P-(Esbp)-IR783 in B16-F10 lung metastases. Taken together, this study demonstrates, for the first time, that the E-selectin targeted copolymer-drug conjugates can inhibit primary tumor growth and prevent metastases in vivo.


Asunto(s)
Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Selectina E/metabolismo , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico , Polímeros/uso terapéutico , Acrilamidas/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Femenino , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Neoplasias/patología , Péptidos/química , Péptidos/farmacocinética , Polímeros/química , Polímeros/farmacocinética , Carga Tumoral/efectos de los fármacos
19.
Mol Cancer Ther ; 14(6): 1385-94, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25887886

RESUMEN

Acquired resistance to therapy is a major obstacle in clinical oncology, and little is known about the contributing mechanisms of the host response to therapy. Here, we show that the proinflammatory cytokine IL1ß is overexpressed in response to paclitaxel chemotherapy in macrophages, subsequently promoting the invasive properties of malignant cells. In accordance, blocking IL1ß, or its receptor, using either genetic or pharmacologic approach, results in slight retardation of primary tumor growth; however, it accelerates metastasis spread. Tumors from mice treated with combined therapy of paclitaxel and the IL1 receptor antagonist anakinra exhibit increased number of M2 macrophages and vessel leakiness when compared with paclitaxel monotherapy-treated mice, indicating a prometastatic role of M2 macrophages in the IL1ß-deprived microenvironment. Taken together, these findings demonstrate the dual effects of blocking the IL1 pathway on tumor growth. Accordingly, treatments using "add-on" drugs to conventional therapy should be investigated in appropriate tumor models consisting of primary tumors and their metastases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Interleucina-1beta/genética , Neoplasias Experimentales/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Interleucina-1beta/sangre , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/prevención & control , Paclitaxel/administración & dosificación , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Tumoral/efectos de los fármacos
20.
Cytokine ; 73(1): 138-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25748836

RESUMEN

IL-1α and IL-1ß are synthesized as 31kDa cell-associated precursors following TLR-4 stimulation, but their processing to the mature form and secretion require a second intracellular stimulus. The unique localization of the precursor of IL-1α (pro-IL-1α) to the nucleus suggested a role in transcriptional regulation of inflammatory cytokines. We explored the hypothesis that pro-IL-1α is involved in regulation of IL-1ß expression following TLR-4 stimulation. IL-1ß mRNA and protein levels were specifically decreased in macrophages from IL-1α-deficient mice following TLR-1/2, TLR-4 or TLR-9 stimulation, supporting the hypothesis. However, activation of the main upstream regulators of IL-1ß expression, IRF3, NFkB and p38/JNK, were not reduced in macrophages from IL-1α-deficient mice. In order to assess the specific role of IL-1α in macrophages, we generated mice with myeloid cell deficiency of IL-1α (LyzMCre-loxp). Despite over 90% knockdown of IL-1α, TLR-4 stimulated macrophages from LyzMCre-loxp mice did not produce lower levels of IL-1ß compared to IL-1α-loxp-flanked mice. In order to overcome the possibility that effects are caused by the incomplete deficiency of IL-1α, we generated new whole-body IL-1α knockout mice (GeneralCre-IL-1α) and the findings were similar to myeloid cell-deficient IL-1α. Collectively, our findings do not support the previously suggested role of nuclear IL-1α in gene regulation of IL-1ß. Rather, they suggest that IL-1α acts mainly as an alarmin that is sequestered in the nucleus following stimulation with TLR-4.


Asunto(s)
Técnicas de Silenciamiento del Gen , Interleucina-1alfa/metabolismo , Interleucina-1beta/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...