Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 25(6)2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38542331

RESUMO

Colorectal cancer (CRC) is a major life-threatening disease, being the third most common cancer and a leading cause of death worldwide. Enhanced adiposity, particularly visceral fat, is a major risk factor for CRC, and obesity-associated alterations in metabolic, inflammatory and immune profiles in visceral adipose tissue (VAT) strongly contribute to promoting or sustaining intestinal carcinogenesis. The role of diet and nutrition in obesity and CRC has been extensively demonstrated, and AT represents the main place where diet-induced signals are integrated. Among the factors introduced with diet and processed or enriched in AT, ω3/ω6 polyunsaturated fatty acids (PUFAs) are endowed with pro- or anti-inflammatory properties and have been shown to exert either promoting or protective roles in CRC. In this study, we investigated the impact of ex vivo exposure to the ω3 and ω6 PUFAs docosahexaenoic and arachidonic acids on VAT adipocyte whole transcription in healthy lean, obese and CRC-affected individuals. High-throughput sequencing of protein-coding and long non-coding RNAs allowed us to identify specific pathways and regulatory circuits controlled by PUFAs and highlighted an impaired responsiveness of obese and CRC-affected individuals as compared to the strong response observed in healthy lean subjects. This further supports the role of healthy diets and balanced ω3/ω6 PUFA intake in the primary prevention of obesity and cancer.


Assuntos
Neoplasias Colorretais , Ácidos Graxos Ômega-3 , RNA Longo não Codificante , Humanos , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Insaturados , Adipócitos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Neoplasias Colorretais/genética
2.
Crit Rev Food Sci Nutr ; 56(5): 737-47, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-24941314

RESUMO

Marked changes in socioeconomic status, cultural traditions, population growth, and agriculture have been affecting diets worldwide. Nutrition is known to play a pivotal role in the pathogenesis of several chronic diseases, and the use of bioactive food compounds at pharmacologic doses is emerging as a preventive and/or therapeutic approach to target metabolic dysregulations occurring in aging, obesity-related chronic diseases, and cancer. Only recently have data on the effects of specific nutrients or food on the immune system become available, and studies regarding the human immune system are still in their infancy. Beyond providing essential nutrients, diet can actively influence the immune system. Understanding how diet and nutritional status influence the innate and adaptive arms of our immune system represents an area of scientific need, opportunity, and challenge. The insights gleaned should help to address several pressing global health problems. Recently, biologically active polyphenols, which are widespread constituents of fruit and vegetables, have gained importance as complex regulators of various cellular processes, critically involved in the maintenance of body homeostasis. This review outlines the potential effects of polyphenols on the function of dendritic cells (DCs), key players in the orchestration of the immune response. Their effects on different aspects of DC biology including differentiation, maturation, and DC capacity to shift immune response toward tolerance or immune activation will be outlined.


Assuntos
Células Dendríticas/citologia , Dieta , Polifenóis/química , Animais , Disponibilidade Biológica , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Frutas/química , Marcadores Genéticos , Humanos , Verduras/química
3.
BMC Genomics ; 16: 480, 2015 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-26116514

RESUMO

BACKGROUND: MicroRNAs (miRs) are an abundant class of small non-coding RNAs (~22 nt) that reprogram gene expression by targeting mRNA degradation and translational disruption. An emerging concept implicates miR coupling with transcription factors in myeloid cell development and function, thus contributing to host defense and inflammation. The important role that these molecules play in the pathogenesis of HIV-1 is only now emerging. RESULTS: We provide evidence that exposure of monocyte-derived dendritic cells (MDDCs) to recombinant HIV-1 R5 gp120, but not to CCR5 natural ligand CCL4, influences the expression of a panel of miRs (i.e., miR-21, miR-155 and miR-181b) regulated by STAT3 and potentially targeting genes belonging to the STAT3 signaling pathway. The blockage of gp120-induced STAT3 activation impairs gp120 capacity to modulate the expression level of above mentioned miRs. Predictive analysis of miR putative targets emphasizes that these miRs share common target genes. Furthermore, gene ontology and pathway enrichment analysis outline that these genes mainly belong to biological processes related to regulation of transcription, in a complex network of interactions involving pathways relevant to HIV-DC interaction. CONCLUSIONS: Overall, these results point to gp120-triggered modulation of miR expression via STAT3 activation as a novel molecular mechanism exploited by HIV-1 to affect DC biology and thus modulate the immune response through complex regulatory loops involving, at the same time, miRs and transcription factors.


Assuntos
Células Dendríticas/metabolismo , Expressão Gênica/genética , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , MicroRNAs/genética , Monócitos/metabolismo , Fator de Transcrição STAT3/genética , Células Dendríticas/virologia , Humanos , Monócitos/virologia , Transdução de Sinais/genética
4.
RNA ; 19(3): 365-79, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23329697

RESUMO

MicroRNAs (miRNAs) are a class of small noncoding RNAs acting as post-transcriptional gene expression regulators in many physiological and pathological conditions. During the last few years, many novel mammalian miRNAs have been predicted experimentally with bioinformatics approaches and validated by next-generation sequencing. Although these strategies have prompted the discovery of several miRNAs, the total number of these genes still seems larger. Here, by exploiting the species conservation of human, mouse, and rat hairpin miRNAs, we discovered a novel rat microRNA, mir-155. We found that mature miR-155 is overexpressed in rat spleen myeloid cells treated with LPS, similarly to humans and mice. Rat mir-155 is annotated only on the alternate genome, suggesting the presence of other "hidden" miRNAs on this assembly. Therefore, we comprehensively extended the homology search also to mice and humans, finally validating 34 novel mammalian miRNAs (two in humans, five in mice, and up to 27 in rats). Surprisingly, 15 of these novel miRNAs (one for mice and 14 for rats) were found only on the alternate and not on the reference genomic assembly. To date, our findings indicate that the choice of genomic assembly, when mapping small RNA reads, is an important option that should be carefully considered, at least for these animal models. Finally, the discovery of these novel mammalian miRNA genes may contribute to a better understanding of already acquired experimental data, thereby paving the way to still unexplored investigations and to unraveling the function of miRNAs in disease models.


Assuntos
Genoma , MicroRNAs/genética , Animais , Sequência de Bases , Biologia Computacional , Genômica , Humanos , Camundongos , MicroRNAs/metabolismo , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Ratos , Análise de Sequência de RNA
5.
J Virol ; 88(19): 11045-55, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25008924

RESUMO

UNLABELLED: Dendritic cells (DCs) are fundamental for the initiation of immune responses and are important players in AIDS immunopathogenesis. The modulation of DC functional activities represents a strategic mechanism for HIV-1 to evade immune surveillance. Impairment of DC function may result from bystander effects of HIV-1 envelope proteins independently of direct HIV-1 infection. In this study, we report that exposure of immature monocyte-derived DCs (MDDCs) to HIV-1 R5 gp120 resulted in the CCR5-dependent production of interleukin-6 (IL-6) via mitogen-activated protein kinase (MAPK)/NF-κB pathways. IL-6 in turn activated STAT3 by an autocrine loop. Concomitantly, gp120 promoted an early activation of STAT3 that further contributed to IL-6 induction. This activation paralleled a concomitant upregulation of the STAT3 inhibitor PIAS3. Notably, STAT3/IL-6 pathway activation was not affected by the CCR5-specific ligand CCL4. These results identify STAT3 as a key signaling intermediate activated by gp120 in MDDCs and highlight the existence of a virus-induced dysregulation of the IL-6/STAT3 axis. HIV-1 gp120 signaling through STAT3 may provide an explanation for the impairment of DC function observed upon HIV exposure. IMPORTANCE: This study provides new evidence for the molecular mechanisms and signaling pathways triggered by HIV-1 gp120 in human DCs in the absence of productive infection, emphasizing a role of aberrant signaling in early virus-host interaction, contributing to viral pathogenesis. We identified STAT3 as a key component in the gp120-mediated signaling cascade involving MAPK and NF-κB components and ultimately leading to IL-6 secretion. STAT3 now is recognized as a key regulator of DC functions. Thus, the identification of this transcription factor as a signaling molecule mediating some of gp120's biological effects unveils a new mechanism by which HIV-1 may deregulate DC functions and contribute to AIDS pathogenesis.


Assuntos
Células Dendríticas/imunologia , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Interleucina-6/imunologia , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/imunologia , Comunicação Autócrina , Diferenciação Celular , Células Cultivadas , Quimiocina CCL4/genética , Quimiocina CCL4/imunologia , Células Dendríticas/virologia , Regulação da Expressão Gênica , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Interleucina-6/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/imunologia , Chaperonas Moleculares/genética , Chaperonas Moleculares/imunologia , Monócitos/imunologia , Monócitos/virologia , NF-kappa B/genética , NF-kappa B/imunologia , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/imunologia , Receptores CCR5/genética , Receptores CCR5/imunologia , Fator de Transcrição STAT3/genética
6.
Cancers (Basel) ; 15(1)2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36612048

RESUMO

Colorectal cancer (CRC) is a leading cause of death worldwide. Despite advances in surgical and therapeutic management, tumor metastases and resistance to therapy still represent major hurdles. CRC risk is highly modifiable by lifestyle factors, including diet, which strongly influences both cancer incidence and related mortality. Galectin-3 (Gal-3) is a multifaceted protein involved in multiple pathophysiological pathways underlying chronic inflammation and cancer. Its versatility is given by the ability to participate in a wide range of tumor-promoting processes, including cell-cell/cell-matrix interactions, cell growth regulation and apoptosis, and the immunosuppressive tumor microenvironment. This review provides an updated summary of preclinical and observational human studies investigating the pathogenetic role of Gal-3 in intestinal inflammation and CRC, as well as the potential of Gal-3 activity inhibition by plant-source food-derived bioactive compounds to control CRC onset/growth. These studies highlight both direct and immuno-mediated effects of Gal-3 on tumor growth and invasiveness and its potential role as a CRC prognostic biomarker. Substantial evidence indicates natural food-derived Gal-3 inhibitors as promising candidates for CRC prevention and therapy. However, critical issues, such as their bioavailability and efficacy, in controlled human studies need to be addressed to translate research progress into clinical applications.

7.
Blood ; 114(4): 796-806, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19465691

RESUMO

Toll-like receptor (TLR) signaling activation by pathogens is critical to the induction of immune responses, and demands tight regulation. We describe in this study that CC chemokine ligand 2 (CCL2) secretion triggered by TLR4 or TLR8 engagement is strongly inhibited upon simultaneous activation of both TLRs in human monocyte-derived dendritic cells (DCs). Impaired CCL2 secretion occurs concomitantly to interleukin-12 up-regulation, being part of a complex regulatory circuit ensuring optimal T helper type 1 polarization. Interestingly, triggering selected TLRs or their combinations differently affects nuclear factor-kappaB p65 activation and microRNA expression. Overall, these results indicate that CCL2 supplies an important immunomodulatory role to DCs, and may contribute to dictate the cytokine profile in T helper type 1 responses induced by DCs.


Assuntos
Polaridade Celular/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Receptores Toll-Like/agonistas , Células Cultivadas , Quimiocina CCL2/genética , Células Dendríticas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Combinação de Medicamentos , Humanos , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Interferon beta/metabolismo , Interleucina-10/metabolismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Poli I-C/administração & dosagem , Poli I-C/farmacologia , RNA Mensageiro/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
8.
Cancers (Basel) ; 13(18)2021 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-34572726

RESUMO

Colorectal cancer (CRC) is a major cancer type and a leading cause of death worldwide. Despite advances in therapeutic management, the current medical treatments are not sufficient to control metastatic disease. Treatment-related adverse effects and drug resistance strongly contribute to therapy failure and tumor recurrence. Combination therapy, involving cytotoxic treatments and non-toxic natural compounds, is arousing great interest as a promising more effective and safer alternative. Polyphenols, a heterogeneous group of bioactive dietary compounds mainly found in fruit and vegetables, have received great attention for their capacity to modulate various molecular pathways active in cancer cells and to affect host anticancer response. This review provides a summary of the most recent (i.e., since 2016) preclinical and clinical studies using polyphenols as adjuvants for CRC therapies. These studies highlight the beneficial effects of dietary polyphenols in combination with cytotoxic drugs or irradiation on both therapy outcome and drug resistance. Despite substantial preclinical evidence, data from a few pilot clinical trials are available to date with promising but still inconclusive results. Larger randomized controlled studies and polyphenol formulations with improved bioavailability are needed to translate the research progress into clinical applications and definitively prove the added value of these molecules in CRC management.

9.
Cells ; 10(7)2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34359908

RESUMO

Colorectal cancer (CRC) is among the major threatening diseases worldwide, being the third most common cancer, and a leading cause of death, with a global incidence expected to increase in the coming years. Enhanced adiposity, particularly visceral fat, is a major risk factor for the development of several tumours, including CRC, and represents an important indicator of incidence, survival, prognosis, recurrence rates, and response to therapy. The obesity-associated low-grade chronic inflammation is thought to be a key determinant in CRC development, with the adipocytes and the adipose tissue (AT) playing a significant role in the integration of diet-related endocrine, metabolic, and inflammatory signals. Furthermore, AT infiltrating immune cells contribute to local and systemic inflammation by affecting immune and cancer cell functions through the release of soluble mediators. Among the factors introduced with diet and enriched in AT, fatty acids (FA) represent major players in inflammation and are able to deeply regulate AT homeostasis and immune cell function through gene expression regulation and by modulating the activity of several transcription factors (TF). This review summarizes human studies on the effects of dietary FA on AT homeostasis and immune cell functions, highlighting the molecular pathways and TF involved. The relevance of FA balance in linking diet, AT inflammation, and CRC is also discussed. Original and review articles were searched in PubMed without temporal limitation up to March 2021, by using fatty acid as a keyword in combination with diet, obesity, colorectal cancer, inflammation, adipose tissue, immune cells, and transcription factors.


Assuntos
Tecido Adiposo/patologia , Neoplasias Colorretais/patologia , Dieta , Ácidos Graxos/efeitos adversos , Homeostase , Imunidade , Obesidade/patologia , Humanos
10.
Cancers (Basel) ; 12(1)2020 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-31936360

RESUMO

ß-glucans represent a heterogeneous group of naturally occurring and biologically active polysaccharides found in many kinds of edible mushrooms, baker's yeast, cereals and seaweeds, whose health-promoting effects have been known since ancient times. These compounds can be taken orally as food supplements or as part of daily diets, and are safe to use, nonimmunogenic and well tolerated. A main feature of ß-glucans is their capacity to function as biological response modifiers, exerting regulatory effects on inflammation and shaping the effector functions of different innate and adaptive immunity cell populations. The potential to interfere with processes involved in the development or control of cancer makes ß-glucans interesting candidates as adjuvants in antitumor therapies as well as in cancer prevention strategies. Here, the regulatory effects of dietary ß-glucans on human innate immunity cells are reviewed and their potential role in cancer control is discussed.

11.
Crit Rev Oncol Hematol ; 145: 102834, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31790930

RESUMO

Colorectal cancer (CRC) is a leading cause of cancer-related mortality in the world. Patterns and trends in CRC incidence and mortality correlate with increasing adoption of Western lifestyles and with the overweight/obesity epidemic. Both genetic background and a range of modifiable environmental/lifestyle factors play a role in CRC etiology. Among these the links of body weight, dietary patterns and physical activity (PA) behavior with CRC risk are some of the strongest for any type of cancer, with a different impact in women and men. Nonetheless, gender disparities still represent a neglected aspect of CRC management. This review sheds light on gender-related association of obesity and different dietary/PA habits with CRC risk, highlighting the importance of lifestyle modifications in the prevention of this neoplastic disease. In this scenario, intervention studies are strongly recommended to define the most effective dietary/PA regimens for primary prevention of cancer in women and men.


Assuntos
Neoplasias Colorretais , Estilo de Vida , Obesidade , Neoplasias Colorretais/epidemiologia , Dieta , Comportamento Alimentar , Feminino , Humanos , Masculino , Fatores de Risco , Fatores Sexuais
12.
Front Oncol ; 10: 1089, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32714872

RESUMO

Obesity, and the obesity-associated inflammation, represents a major risk factor for the development of chronic diseases, including colorectal cancer (CRC). Dysfunctional visceral adipose tissue (AT) is now recognized as key player in obesity-associated morbidities, although the biological processes underpinning the increased CRC risk in obese subjects are still a matter of debate. Recent findings have pointed to specific alterations in the expression pattern of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), and long non-coding RNAs (lncRNAs), as mechanisms underlying dysfunctional adipocyte phenotype in obesity. Nevertheless, the regulatory networks and interrelated processes relevant for adipocyte functions, that may contribute to a tumor-promoting microenvironment, are poorly known yet. To this end, based on RNA sequencing data, we identified lncRNAs and miRNAs, which are aberrantly expressed in visceral adipocytes from obese and CRC subjects, as compared to healthy lean control, and validated a panel of modulated ncRNAs by real-time qPCR. Furthermore, by combining the differentially expressed lncRNA and miRNA profiles with the transcriptome analysis dataset of adipocytes from lean and obese subjects affected or not by CRC, lncRNA-miRNA-mRNA adipocyte networks were defined for obese and CRC subjects. This analysis highlighted several ncRNAs modulation that are common to both obesity and CRC or unique of each disorder. Functional enrichment analysis of network-related mRNA targets, revealed dysregulated pathways associated with metabolic processes, lipid and energy metabolism, inflammation, and cancer. Moreover, adipocytes from obese subjects affected by CRC exhibited a higher complexity, in terms of number of genes, lncRNAs, miRNAs, and biological processes found to be dysregulated, providing evidence that the transcriptional and post-transcriptional program of adipocytes from CRC patients is deeply affected by obesity. Overall, this study adds further evidence for a central role of visceral adipocyte dysfunctions in the obesity-cancer relationship.

13.
Front Immunol ; 10: 265, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30838002

RESUMO

Obesity, a low-grade inflammatory condition, represents a major risk factor for the development of several pathologies including colorectal cancer (CRC). Although the adipose tissue inflammatory state is now recognized as a key player in obesity-associated morbidities, the underlying biological processes are complex and not yet precisely defined. To this end, we analyzed transcriptome profiles of human visceral adipocytes from lean and obese subjects affected or not by CRC by RNA sequencing (n = 6 subjects/category), and validated selected modulated genes by real-time qPCR. We report that obesity and CRC, conditions characterized by the common denominator of inflammation, promote changes in the transcriptional program of adipocytes mostly involving pathways and biological processes linked to extracellular matrix remodeling, and metabolism of pyruvate, lipids and glucose. Interestingly, although the transcriptome of adipocytes shows several alterations that are common to both disorders, some modifications are unique under obesity (e.g., pathways associated with inflammation) and CRC (e.g., TGFß signaling and extracellular matrix remodeling) and are influenced by the body mass index (e.g., processes related to cell adhesion, angiogenesis, as well as metabolism). Indeed, cancer-induced transcriptional program is deeply affected by obesity, with adipocytes from obese individuals exhibiting a more complex response to the tumor. We also report that in vitro exposure of adipocytes to ω3 and ω6 polyunsaturated fatty acids (PUFA) endowed with either anti- or pro-inflammatory properties, respectively, modulates the expression of genes involved in processes potentially relevant to carcinogenesis, as assessed by real-time qPCR. All together our results suggest that genes involved in pyruvate, glucose and lipid metabolism, fibrosis and inflammation are central in the transcriptional reprogramming of adipocytes occurring in obese and CRC-affected individuals, as well as in their response to PUFA exposure. Moreover, our results indicate that the transcriptional program of adipocytes is strongly influenced by the BMI status in CRC subjects. The dysregulation of these interrelated processes relevant for adipocyte functions may contribute to create more favorable conditions to tumor establishment or favor tumor progression, thus linking obesity and colorectal cancer.


Assuntos
Adipócitos/fisiologia , Carcinogênese/genética , Neoplasias Colorretais/genética , Ácidos Graxos Insaturados/genética , Obesidade/genética , Transcriptoma/genética , Tecido Adiposo/fisiologia , Adulto , Idoso , Fenômenos Biológicos/genética , Índice de Massa Corporal , Ácidos Graxos Ômega-3/genética , Feminino , Humanos , Inflamação/genética , Metabolismo dos Lipídeos/genética , Masculino , Pessoa de Meia-Idade
14.
Front Immunol ; 9: 2556, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30455701

RESUMO

Colorectal cancer (CRC) is the third most common cancer worldwide and a leading cause of death, with burden expected to increase in the coming years. Enhanced adiposity, particularly visceral fat, is associated with increased cancer incidence representing an important indicator of survival, prognosis, recurrence rates, and response to therapy for several tumors including CRC. Compelling evidence has been achieved that the low-grade chronic inflammation characterizing obesity represents a main factor that can favor carcinogenesis. Adipocytes and adipose tissue (AT) infiltrating immune cells contribute to obesity-related inflammation by releasing soluble factors affecting, both locally and systemically, the function of several cell types, including immune and cancer cells. The unbalanced production of immune mediators as well as the profound changes in the repertoire and activation state of immune cells in AT of obese subjects represent key events in the processes that set the basis for a pro-tumorigenic microenvironment. AT harbors a unique profile of immune cells of different origin that play an important role in tissue homeostasis. Among these, tissue-resident innate lymphocytes are emerging as important AT components whose depletion/aberrant activation occurring in obesity could have an impact on inflammation and immune-surveillance against tumors. However, a direct link between obesity-induced dysfunction and cancer development has not been demonstrated yet. In this review, we provide an overview of human obesity- and CRC-induced alterations of blood and adipose tissue-associated innate lymphocytes, and discuss how the adipose tissue microenvironment in obesity might influence the development of CRC.


Assuntos
Adipócitos/citologia , Tecido Adiposo/citologia , Neoplasias Colorretais/imunologia , Imunidade Inata/imunologia , Linfócitos/imunologia , Obesidade/imunologia , Tecido Adiposo/imunologia , Neoplasias Colorretais/patologia , Homeostase/imunologia , Humanos , Linfócitos/citologia , Obesidade/patologia , Microambiente Tumoral
15.
Front Microbiol ; 8: 597, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28424679

RESUMO

Environmental and lifestyle factors, including diet and nutritional habits have been strongly linked to colorectal cancer (CRC). Of note, unhealthy dietary habits leading to adiposity represent a main risk factor for CRC and are associated with a chronic low-grade inflammatory status. Inflammation is a hallmark of almost every type of cancer and can be modulated by several food compounds exhibiting either protective or promoting effects. However, in spite of an extensive research, the underlying mechanisms by which dietary patterns or bioactive food components may influence tumor onset and outcome have not been fully clarified yet. Growing evidence indicates that diet, combining beneficial substances and potentially harmful ingredients, has an impact on the expression of key regulators of gene expression such as the non-coding RNA (ncRNA). Since the expression of these molecules is deranged in chronic inflammation and cancer, modulating their expression may strongly influence the cancer phenotype and outcomes. In addition, the recently acquired knowledge on the existence of intricate inter-kingdom communication networks, is opening new avenues for a deeper understanding of the intimate relationships linking diet to CRC. In this novel scenario, diet-modulated ncRNA may represent key actors in the interaction between plant and animal kingdoms, capable of influencing disease onset and outcome. In this review, we will summarize the studies demonstrating a link between bioactive food components, including food-derived, microbiota-processed, secondary metabolites, and host ncRNA. We will focus on microRNA, highlighting how this plant/animal inter-kingdom cross-talk may have an impact on CRC establishment and progression.

16.
Front Immunol ; 8: 643, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649243

RESUMO

Visceral adipose tissue (VAT) is a main site where metabolic and immunologic processes interplay to regulate, at local and systemic level, the inflammatory status and immune response. Obesity-associated inflammation and immune dysfunctions are inextricably linked to tumor but, in spite of intense efforts, the mechanisms underpinning this association remain elusive. In this report, we characterized the profile of VAT-associated and circulating innate lymphocyte and regulatory T (Treg) cell subsets underlying inflammatory conditions, such as obesity and colorectal cancer (CRC). Analysis of NK, NKT-like, γδ T, and Treg cell populations in VAT and blood of healthy lean subjects revealed that CD56hi NK and OX40+ Treg cells are more abundant in VAT with respect to blood. Conversely, CD56dim NK and total Treg cells are most present in the circulation, while γδ T lymphocytes are uniformly distributed in the two compartments. Interestingly, a reduced frequency of circulating activated Treg cells, and a concomitant preferential enrichment of OX40-expressing Treg cells in VAT, were selectively observed in obese (Ob) subjects, and directly correlated with body mass index. Likewise, CRC patients were characterized by a specific enrichment of VAT-associated NKT-like cells. In addition, Ob and CRC-affected individuals shared a significant reduction of the Vγ9Vδ2/γδ T cell ratio at systemic level. The alterations in the relative proportions of Treg and NKT-like cells in VAT were found to correlate with the content of pro- and anti-inflammatory polyunsaturated fatty acids (PUFA), respectively. Overall, these results provide evidence for distinct alterations of the immune cell repertoire in the periphery with respect to the VAT microenvironment that uniquely characterize or are shared by different inflammatory conditions, such as obesity and CRC, and suggest that VAT PUFA composition may represent one of the factors that contribute to shape the immune phenotypes.

17.
J Leukoc Biol ; 99(1): 97-105, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26307548

RESUMO

The Toll-like receptors are the first line of the host response to pathogens, representing an essential component of the innate and adaptive immune response. They recognize different pathogens and trigger responses directed at eliminating the invader and at developing immunologic long-term memory, ultimately affecting viral pathogenesis. In viral infections, sensing of nucleic acids and/or viral structural proteins generally induces a protective immune response. Thus, it is not surprising that many viruses have developed strategies to evade or counteract signaling through the Toll-like receptor pathways, to survive the host defense machinery and ensure propagation. Thus, Toll-like receptor engagement can also be part of viral pathogenic mechanisms. Evidence for a direct interaction of Toll-like receptors with human immunodeficiency virus type 1 (HIV-1) structures has started to be achieved, and alterations of their expression and function have been described in HIV-1-positive subjects. Furthermore, Toll-like receptor triggering by bacterial and viral ligands have been described to modulate HIV-1 replication and host response, leading to protective or detrimental effects. This review covers major advances in the field of HIV-1 interplay with Toll-like receptors, focusing on human myeloid cells (e.g., monocytes/macrophages and dendritic cells). The role of this interaction in the dysregulation of myeloid cell function and in dictating aspects of the multifaceted pathogenesis of acquired immunodeficiency syndrome will be discussed.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Células Mieloides/imunologia , Células Mieloides/metabolismo , Receptores Toll-Like/metabolismo , Apresentação de Antígeno , Antígenos Virais/imunologia , Infecções por HIV/virologia , Humanos , Imunidade Inata , Ligantes , Ligação Proteica , Receptores de Quimiocinas/metabolismo , Receptores Toll-Like/agonistas , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Replicação Viral
18.
J Leukoc Biol ; 100(3): 599-606, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26992429

RESUMO

Highly active antiretroviral therapy has significantly improved the prognosis of HIV-infected subjects. However, patients treated long term still manifest increased mortality and, even with undetectable plasma viremia, often experience persistent immune activation. Furthermore, liver-related mortality is now the most common cause of non-AIDS-related death in HIV-infected individuals on highly active antiretroviral therapy through accelerated fibrosis progression. TLRs are the first line of the host response to pathogens and play an important role in human host defense against viruses through sensing of viral structural proteins. Growing evidence points to TLR4 as a key player in chronic immune activation, HIV recognition/replication, and liver fibrosis progression, suggesting that HIV triggering of TLR4 may dictate some aspects of the multifaceted AIDS pathogenesis. In this study, we provide evidence for an interplay between host TLR4 and HIV-1 gp120 in human monocyte-derived macrophages and hepatic stellate cells, leading to intracellular pathways and biologic activities that mediate proinflammatory and profibrogenic signals. Finally, we hypothesize that CCR5 and TLR4 are likely part of a common receptor cluster, as the blocking of CCR5 by specific antagonists impairs the macrophage capacity to produce chemokines in response to LPS. Chronic immune activation and liver fibrosis remain important obstacles for highly active antiretroviral therapy success. Thus, the identification of gp120-TLR4 axis as a novel determinant of immune system and hepatic stellate cell biology opens new perspectives to the management of HIV infection and disease.


Assuntos
Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/imunologia , Células Estreladas do Fígado/imunologia , Cirrose Hepática/imunologia , Macrófagos/imunologia , Receptor 4 Toll-Like/metabolismo , Terapia Antirretroviral de Alta Atividade , Células Cultivadas , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/imunologia , Células Estreladas do Fígado/patologia , Células Estreladas do Fígado/virologia , Humanos , Cirrose Hepática/patologia , Cirrose Hepática/virologia , Macrófagos/patologia , Macrófagos/virologia , Receptores CCR5/metabolismo , Transdução de Sinais
19.
Oncotarget ; 7(39): 63093-63105, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27494857

RESUMO

Obesity is a low-grade chronic inflammatory state representing an important risk factor for colorectal cancer (CRC). Adipocytes strongly contribute to inflammation by producing inflammatory mediators. In this study we investigated the role of human visceral fat adipocytes in regulating the functions of innate immunity cells. Adipocyte-conditioned media (ACM) from obese (n = 14) and CRC (lean, n = 14; obese, n = 13) subjects released higher levels of pro-inflammatory/immunoregulatory factors as compared to ACM from healthy lean subjects (n = 13). Dendritic cells (DC), differentiated in the presence of ACM from obese and CRC subjects, expressed elevated levels of the inhibitory molecules PD-L1 and PD-L2, and showed a reduced IL-12/IL-10 ratio in response to both TLR ligand- and γδ T lymphocyte-induced maturation. Furthermore, CRC patient-derived ACM inhibited DC-mediated γδ T cell activation. The immunosuppressive signals delivered by ACM from obese and CRC individuals were associated with a pro-inflammatory secretory and ω6 polyunsaturated fatty acid profile of adipocytes. Interestingly, STAT3 activation in adipocytes correlated with dihomo-γlinolenic acid content and was further induced by arachidonic acid, which conversely down-modulated PPARγ. These results provide novel evidence for a cross-talk between human adipocytes and innate immunity cells whose alteration in obesity and CRC may lead to immune dysfunctions, thus setting the basis for cancer development.


Assuntos
Adipócitos/química , Neoplasias Colorretais/metabolismo , Ácidos Graxos Ômega-6/metabolismo , Ácidos Graxos Insaturados/metabolismo , Tolerância Imunológica , Obesidade/metabolismo , Adipócitos/citologia , Ácido Araquidônico/química , Quimiocinas/metabolismo , Meios de Cultivo Condicionados/química , Citocinas/metabolismo , Células Dendríticas/citologia , Progressão da Doença , Humanos , Imunidade Inata , Terapia de Imunossupressão , Inflamação , Gordura Intra-Abdominal/citologia , Monócitos/citologia , PPAR gama/metabolismo , Fenótipo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Fatores de Risco , Fator de Transcrição STAT3/metabolismo
20.
Toxins (Basel) ; 7(12): 5472-83, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26694465

RESUMO

Lactoferrin (LF) exhibits a wide range of immunomodulatory activities including modulation of cytokine and chemokine secretion. In this study, we demonstrate that bovine LF (bLF) up-modulates, in a concentration- and time-dependent manner, CCL1 secretion in monocytes (Mo) at the early stage of differentiation toward dendritic cells (DCs), and in fully differentiated immature Mo-derived DCs (MoDCs). In both cell types, up-modulation of CCL1 secretion is an early event following bLF-mediated enhanced accumulation of CCL1 transcripts. Notably, bLF-mediated up-regulation of CCL1 involves the engagement of distinct surface receptors in MoDCs and their Mo precursors. We show that bLF-mediated engagement of CD36 contributes to CCL1 induction in differentiating Mo. Conversely, toll-like receptor (TLR)2 blocking markedly reduces bLF-induced CCL1 production in MoDCs. These findings add further evidence for cell-specific differential responses elicited by bLF through the engagement of distinct TLRs and surface receptors. Furthermore, the different responses observed at early and late stages of Mo differentiation towards DCs may be relevant in mediating bLF effects in specific body districts, where these cell types may be differently represented in physiopathological conditions.


Assuntos
Quimiocina CCL1/metabolismo , Células Dendríticas/efeitos dos fármacos , Lactoferrina/farmacologia , Monócitos/efeitos dos fármacos , Animais , Bovinos , Células Cultivadas , Quimiocina CCL1/genética , Células Dendríticas/metabolismo , Humanos , Monócitos/citologia , Monócitos/metabolismo , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa