Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Leukoc Biol ; 115(6): 993-995, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38498617

RESUMO

The tumor microenvironment provides a unique opportunity to gain insight into the relationship and crosstalk between different cell types. In this context, little is known about the interaction between γδ T cells and neutrophils, which are innate immune cells abundant in the tumor microenvironment. Interestingly, both γδ T cells and neutrophils are heterogeneous, may play diverse regulatory roles, and have been shown to have both protumor and antitumor functions. In this editorial, we discuss recent advances in the understanding of interplay between γδ T cells and neutrophils in cancer and provide insights and future directions highlighting the role these interactions may play in cancer.


Assuntos
Neoplasias , Neutrófilos , Receptores de Antígenos de Linfócitos T gama-delta , Microambiente Tumoral , Humanos , Neutrófilos/imunologia , Neutrófilos/patologia , Neoplasias/imunologia , Neoplasias/patologia , Microambiente Tumoral/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Animais , Comunicação Celular/imunologia , Linfócitos T/imunologia , Linfócitos Intraepiteliais/imunologia
3.
J Leukoc Biol ; 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38334307

RESUMO

Poly (ADP ribose) polymerase family member 11(PARP11) has important immune regulatory functions in viral infection and tumor immune response. Particularly, PARP11 showed protumor activities in multiple preclinical murine models. However, no systematic pan-cancer analysis has been conducted to explore PARP11 function. In this study we used multiple databases to assess PARP11 expression, which associations with clinical outcomes, immune checkpoint factors, prognostic significance, genomic characteristics, and immunological aspects. The analysis revealed varying expression levels of PARP11 across different cancer types and a significant correlation between its expression and immune cell infiltration. Insights from the CellMiner database suggest a strong link between PARP11 expression and sensitivity to anticancer drugs, highlighting its potential as a therapeutic target. Moreover, PARP11 expression correlates with patient survival during anti-PD1 and anti-CTLA4 treatments, suggested that PARP11 would be a predictor of immune checkpoint inhibitor (ICI) treatment. In summary, PARP11 would be a potential immunoregulatory target and a diagnosis and prognosis marker for certain types of cancers. The detailed mechanisms of PARP11 in tumor immune responses need to be further investigated.

4.
Neuro Oncol ; 26(5): 858-871, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38153426

RESUMO

BACKGROUND: Intrinsic or environmental stresses trigger the accumulation of improperly folded proteins in the endoplasmic reticulum (ER), leading to ER stress. To cope with this, cells have evolved an adaptive mechanism named the unfolded protein response (UPR) which is hijacked by tumor cells to develop malignant features. Glioblastoma (GB), the most aggressive and lethal primary brain tumor, relies on UPR to sustain growth. We recently showed that IRE1 alpha (referred to IRE1 hereafter), 1 of the UPR transducers, promotes GB invasion, angiogenesis, and infiltration by macrophage. Hence, high tumor IRE1 activity in tumor cells predicts a worse outcome. Herein, we characterized the IRE1-dependent signaling that shapes the immune microenvironment toward monocytes/macrophages and neutrophils. METHODS: We used human and mouse cellular models in which IRE1 was genetically or pharmacologically invalidated and which were tested in vivo. Publicly available datasets from GB patients were also analyzed to confirm our findings. RESULTS: We showed that IRE1 signaling, through both the transcription factor XBP1s and the regulated IRE1-dependent decay controls the expression of the ubiquitin-conjugating E2 enzyme UBE2D3. In turn, UBE2D3 activates the NFκB pathway, resulting in chemokine production and myeloid infiltration in tumors. CONCLUSIONS: Our work identifies a novel IRE1/UBE2D3 proinflammatory axis that plays an instrumental role in GB immune regulation.


Assuntos
Neoplasias Encefálicas , Endorribonucleases , Glioblastoma , Células Mieloides , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Glioblastoma/patologia , Glioblastoma/metabolismo , Humanos , Camundongos , Endorribonucleases/metabolismo , Endorribonucleases/genética , Animais , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Células Mieloides/metabolismo , Células Mieloides/patologia , Resposta a Proteínas não Dobradas , Microambiente Tumoral , Células Tumorais Cultivadas , Estresse do Retículo Endoplasmático
5.
Front Immunol ; 13: 1003871, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275643

RESUMO

Neutrophils play critical roles in a broad spectrum of clinical conditions. Accordingly, manipulation of neutrophil function may provide a powerful immunotherapeutic approach. However, due to neutrophils characteristic short half-life and their large population number, this possibility was considered impractical. Here we describe the identification of peptides which specifically bind either murine or human neutrophils. Although the murine and human neutrophil-specific peptides are not cross-reactive, we identified CD177 as the neutrophil-expressed binding partner in both species. Decorating nanoparticles with a neutrophil-specific peptide confers neutrophil specificity and these neutrophil-specific nanoparticles accumulate in sites of inflammation. Significantly, we demonstrate that encapsulating neutrophil modifying small molecules within these nanoparticles yields specific modulation of neutrophil function (ROS production, degranulation, polarization), intracellular signaling and longevity both in vitro and in vivo. Collectively, our findings demonstrate that neutrophil specific targeting may serve as a novel mode of immunotherapy in disease.


Assuntos
Nanopartículas , Neutrófilos , Camundongos , Humanos , Animais , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Inflamação/metabolismo
6.
Genome Med ; 14(1): 120, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36266692

RESUMO

BACKGROUND: Drug resistance continues to be a major limiting factor across diverse anti-cancer therapies. Contributing to the complexity of this challenge is cancer plasticity, in which one cancer subtype switches to another in response to treatment, for example, triple-negative breast cancer (TNBC) to Her2-positive breast cancer. For optimal treatment outcomes, accurate tumor diagnosis and subsequent therapeutic decisions are vital. This study assessed a novel approach to characterize treatment-induced evolutionary changes of distinct tumor cell subpopulations to identify and therapeutically exploit anticancer drug resistance. METHODS: In this research, an information-theoretic single-cell quantification strategy was developed to provide a high-resolution and individualized assessment of tumor composition for a customized treatment approach. Briefly, this single-cell quantification strategy computes cell barcodes based on at least 100,000 tumor cells from each experiment and reveals a cell-specific signaling signature (CSSS) composed of a set of ongoing processes in each cell. RESULTS: Using these CSSS-based barcodes, distinct subpopulations evolving within the tumor in response to an outside influence, like anticancer treatments, were revealed and mapped. Barcodes were further applied to assign targeted drug combinations to each individual tumor to optimize tumor response to therapy. The strategy was validated using TNBC models and patient-derived tumors known to switch phenotypes in response to radiotherapy (RT). CONCLUSIONS: We show that a barcode-guided targeted drug cocktail significantly enhances tumor response to RT and prevents regrowth of once-resistant tumors. The strategy presented herein shows promise in preventing cancer treatment resistance, with significant applicability in clinical use.


Assuntos
Antineoplásicos , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Linhagem Celular Tumoral , Transdução de Sinais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
7.
Front Immunol ; 13: 849701, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35911772

RESUMO

Breast tumors and their derived circulating cancer cells express the leukocyte ß2 integrin ligand Intercellular adhesion molecule 1 (ICAM-1). We found that elevated ICAM-1 expression in breast cancer cells results in a favorable outcome and prolonged survival of breast cancer patients. We therefore assessed the direct in vivo contribution of ICAM-1 expressed by breast cancer cells to breast tumorigenesis and lung metastasis in syngeneic immunocompetent mice hosts using spontaneous and experimental models of the lung metastasis of the C57BL/6-derived E0771 cell line, a luminal B breast cancer subtype. Notably, the presence of ICAM-1 on E0771 did not alter tumor growth or the leukocyte composition in the tumor microenvironment. Interestingly, the elimination of Tregs led to the rapid killing of primary tumor cells independently of tumor ICAM-1 expression. The in vivo elimination of a primary E0771 tumor expressing the ovalbumin (OVA) model neoantigen by the OVA-specific OVA-tcr-I mice (OT-I) transgenic cytotoxic T lymphocytes (CTLs) also took place normally in the absence of ICAM-1 expression by E0771 breast cancer target cells. The whole lung imaging of these cells by light sheet microscopy (LSM) revealed that both Wild type (WT)- and ICAM-1-deficient E0771 cells were equally disseminated from resected tumors and accumulated inside the lung vasculature at similar magnitudes. ICAM-1-deficient breast cancer cells developed, however, much larger metastatic lesions than their control counterparts. Strikingly, the vast majority of these cells gave rise to intravascular tumor colonies both in spontaneous and experimental metastasis models. In the latter model, ICAM-1 expressing E0771- but not their ICAM-1-deficient counterparts were highly susceptible to elimination by neutrophils adoptively transferred from E0771 tumor-bearing donor mice. Ex vivo, neutrophils derived from tumor-bearing mice also killed cultured E0771 cells via ICAM-1-dependent interactions. Collectively, our results are a first indication that ICAM-1 expressed by metastatic breast cancer cells that expand inside the lung vasculature is involved in innate rather than in adaptive cancer cell killing. This is also a first indication that the breast tumor expression of ICAM-1 is not required for CTL-mediated killing but can function as a suppressor of intravascular breast cancer metastasis to lungs.


Assuntos
Neoplasias Pulmonares , Linfócitos T Citotóxicos , Animais , Linhagem Celular Tumoral , Molécula 1 de Adesão Intercelular/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina , Microambiente Tumoral
8.
Methods Mol Biol ; 2470: 629-639, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35881379

RESUMO

Plasmodium falciparum, which causes the deadliest form of human malaria, is able to evade antibody-mediated immune responses through switches in expression of surface antigens. Thus, over the years, the focus of most research has been on the role of the adaptive immune response in the course of malaria. However, in recent years there is mounting evidence for the role of the innate immune response to Plasmodium infections. In this context, very little is known on the protective role of neutrophils against blood-stage parasites and the mechanisms by which they recognize and eliminate infected red blood cells. Here we describe several useful methodologies that enable the study and quantification of the interactions between human neutrophils and P. falciparum-infected red blood cells.


Assuntos
Malária Falciparum , Malária , Eritrócitos , Humanos , Malária Falciparum/parasitologia , Neutrófilos , Plasmodium falciparum
9.
J Exp Med ; 219(6)2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35522219

RESUMO

Neutrophils are the first responders to infection and inflammation and are thus a critical component of innate immune defense. Understanding the behavior of neutrophils as they act within various inflammatory contexts has provided insights into their role in sterile and infectious diseases; however, the field of neutrophils in cancer is comparatively young. Here, we summarize key concepts and current knowledge gaps related to the diverse roles of neutrophils throughout cancer progression. We discuss sources of neutrophil heterogeneity in cancer and provide recommendations on nomenclature for neutrophil states that are distinct in maturation and activation. We address discrepancies in the literature that highlight a need for technical standards that ought to be considered between laboratories. Finally, we review emerging questions in neutrophil biology and innate immunity in cancer. Overall, we emphasize that neutrophils are a more diverse population than previously appreciated and that their role in cancer may present novel unexplored opportunities to treat cancer.


Assuntos
Neoplasias , Neutrófilos , Humanos , Imunidade Inata , Inflamação , Neoplasias/genética , Fenótipo
10.
iScience ; 25(5): 104246, 2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35494247

RESUMO

Nucleotide repeat expansions are a hallmark of over 40 neurodegenerative diseases and cause RNA toxicity and multisystemic symptoms that worsen with age. Through an unclear mechanism, RNA toxicity can trigger severe disease manifestation in infants if the repeats are inherited from their mother. Here we use Caenorhabditis elegans bearing expanded CUG repeats to show that this asymmetric intergenerational inheritance of toxicity contributes to disease pathogenesis. In addition, we show that this mechanism is dependent on small RNA pathways with maternal repeat-derived small RNAs causing transcriptomic changes in the offspring, reduced motility, and shortened lifespan. We rescued the toxicity phenotypes in the offspring by perturbing the RNAi machinery in the affected hermaphrodites. This points to a novel mechanism linking maternal bias and the RNAi machinery and suggests that toxic RNA is transmitted to offspring, causing disease phenotypes through intergenerational epigenetic inheritance.

11.
Biomedicines ; 10(4)2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35453656

RESUMO

Neutrophil-mediated cytotoxicity toward tumor cells requires cell contact and is mediated by hydrogen peroxide. We have recently shown that Cathepsin G expressed on the neutrophil surface interacts with tumor RAGE, and this interaction facilitates neutrophil cytotoxicity. Interruption of the Cathepsin G-RAGE interaction led to 50-80% reduction in cytotoxicity, suggesting that additional interactions are also involved. Here we show that blocking antibodies to the C-type lectin receptors (CLRs) Clec4e and Dectin-1, but not those to NKG2D, attenuated murine neutrophil cytotoxicity towards murine tumor cells, suggesting a contributing role for these CLRs in neutrophil recognition of tumor cells. We further observed that the CLRs interact with tumor Nidogen-1 and Hspg2, two sulfated glycoproteins of the basement membrane. Both Nidogen-1 and Hspg2 were found to be expressed on the tumor cell surface. The knockdown of Nidogen-1, but not that of Hspg2, led to reduced susceptibility of the tumor cells to neutrophil cytotoxicity. Altogether, this study suggests a role for CLR-Nidogen-1 interaction in the recognition of tumor cells by neutrophils, and this interaction facilitates neutrophil-mediated killing of the tumor cells.

12.
EMBO Rep ; 23(6): e53641, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35417070

RESUMO

Plasmodium falciparum, the deadliest form of human malaria, remains one of the major threats to human health in endemic regions. Its virulence is attributed to its ability to modify infected red blood cells (iRBC) to adhere to endothelial receptors by placing variable antigens known as PfEMP1 on the iRBC surface. PfEMP1 expression determines the cytoadhesive properties of the iRBCs and is implicated in severe malaria. To evade antibody-mediated responses, the parasite undergoes continuous switches of expression between different PfEMP1 variants. Recently, it became clear that in addition to antibody-mediated responses, PfEMP1 triggers innate immune responses; however, the role of neutrophils, the most abundant white blood cells in the human circulation, in malaria remains elusive. Here, we show that neutrophils recognize and kill blood-stage P. falciparum isolates. We identify neutrophil ICAM-1 and specific PfEMP1 implicated in cerebral malaria as the key molecules involved in this killing. Our data provide mechanistic insight into the interactions between neutrophils and iRBCs and demonstrate the important influence of PfEMP1 on the selective innate response to cerebral malaria.


Assuntos
Malária Cerebral , Malária Falciparum , Plasmodium falciparum , Eritrócitos/parasitologia , Humanos , Malária Cerebral/genética , Malária Cerebral/metabolismo , Malária Falciparum/genética , Neutrófilos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
14.
Eur J Immunol ; 51(9): 2218-2224, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34268737

RESUMO

Neutrophils play a crucial role in immune defense against and clearance of uropathogenic Escherichia coli (UPEC)-mediated urinary tract infection, the most common bacterial infection in healthy humans. CD300a is an inhibitory receptor that binds phosphatidylserine and phosphatidylethanolamine, presented on the membranes of apoptotic cells. CD300a binding to phosphatidylserine and phosphatidylethanolamine, also known as the "eat me" signal, mediates immune tolerance to dying cells. Here, we demonstrate for the first time that CD300a plays an important role in the neutrophil-mediated immune response to UPEC-induced urinary tract infection. We show that CD300a-deficient neutrophils have impaired phagocytic abilities and despite their increased accumulation at the site of infection, they are unable to reduce bacterial burden in the bladder, which results in significant exacerbation of infection and worse host outcome. Finally, we demonstrate that UPEC's pore forming toxin α-hemolysin induces upregulation of the CD300a ligand on infected bladder epithelial cells, signaling to neutrophils to be cleared.


Assuntos
Infecções por Escherichia coli/prevenção & controle , Neutrófilos/imunologia , Receptores Imunológicos/deficiência , Receptores Imunológicos/imunologia , Infecções Urinárias/imunologia , Escherichia coli Uropatogênica/imunologia , Animais , Apoptose/imunologia , Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/metabolismo , Feminino , Técnicas de Inativação de Genes , Proteínas Hemolisinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/genética , Fagocitose/imunologia , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Receptores Imunológicos/genética , Bexiga Urinária/imunologia , Bexiga Urinária/microbiologia , Bexiga Urinária/patologia , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/crescimento & desenvolvimento
15.
Cells ; 10(6)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34203915

RESUMO

The PD-L1/PD-1 axis mediates immune tolerance and promotes tumor growth and progression via the inhibition of anti-tumor immunity. Blocking the interaction between PD-L1 and PD-1 was clinically shown to be beneficial in maintaining the anti-tumor functions of the adaptive immune system. Still, the consequences of blocking the PD-L1/PD-1 axis on innate immune responses remain largely unexplored. In this context, neutrophils were shown to consist of distinct subpopulations, which possess either pro- or anti-tumor properties. PD-L1-expressing neutrophils are considered pro-tumor as they are able to suppress cytotoxic T cells and are propagated with disease progression. That said, we found that PD-L1 expression is not limited to tumor promoting neutrophils, but is also evident in anti-tumor neutrophils. We show that neutrophil cytotoxicity is effectively and efficiently blocked by tumor cell-expressed PD-1. Furthermore, the blocking of either neutrophil PD-L1 or tumor cell PD-1 maintains neutrophil cytotoxicity. Importantly, we show that tumor cell PD-1 blocks neutrophil cytotoxicity and promotes tumor growth via a mechanism independent of adaptive immunity. Taken together, these findings highlight the therapeutic potential of enhancing anti-tumor innate immune responses via blocking of the PD-L1/PD-1 axis.


Assuntos
Antígeno B7-H1/metabolismo , Neutrófilos/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Imunidade Adaptativa , Animais , Antígeno B7-H1/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Tolerância Imunológica , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Neoplasias/metabolismo , Neoplasias/patologia , Neutrófilos/fisiologia , Receptor de Morte Celular Programada 1/fisiologia
16.
J Mol Cell Cardiol ; 155: 125-137, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33130150

RESUMO

AIMS: One unaddressed aspect of healing after myocardial infarction (MI) is how non-myocyte cells that survived the ischemic injury, keep withstanding additional cellular damage by stress forms typically arising during the post-infarction inflammation. Here we aimed to determine if cell survival is conferred by expression of a mitochondrial protein novel to the cardiac proteome, known as steroidogenic acute regulatory protein, (StAR/STARD1). Further studies aimed to unravel the regulation and role of the non-steroidogenic cardiac StAR after MI. METHODS AND RESULTS: Following permanent ligation of the left anterior descending coronary artery in mouse heart, timeline western blot analyses showed that StAR expression corresponds to the inflammatory response to MI. Following the identification of StAR in mitochondria of cardiac fibroblasts in culture, confocal microscopy immunohistochemistry (IHC) identified StAR expression in left ventricular (LV) activated interstitial fibroblasts, adventitial fibroblasts and endothelial cells. Further work with the primary fibroblasts model revealed that interleukin-1α (IL-1α) signaling via NF-κB and p38 MAPK pathways efficiently upregulates the expression of the Star gene products. At the functional level, IL-1α primed fibroblasts were protected against apoptosis when exposed to cisplatin mimicry of in vivo apoptotic stress; yet, the protective impact of IL-1α was lost upon siRNA mediated StAR downregulation. At the physiological level, StAR expression was nullified during post-MI inflammation in a mouse model with global IL-1α deficiency, concomitantly resulting in a 4-fold elevation of apoptotic fibroblasts. Serial echocardiography and IHC studies of mice examined 24 days after MI revealed aggravation of LV dysfunction, LV dilatation, anterior wall thinning and adverse tissue remodeling when compared with loxP control hearts. CONCLUSIONS: This study calls attention to overlooked aspects of cellular responses evolved under the stress conditions associated with the default inflammatory response to MI. Our observations suggest that LV IL-1α is cardioprotective, and at least one mechanism of this action is mediated by induction of StAR expression in border zone fibroblasts, which renders them apoptosis resistant. This acquired survival feature also has long-term ramifications on the heart recovery by diminishing adverse remodeling and improving the heart function after MI.


Assuntos
Fibroblastos/metabolismo , Regulação da Expressão Gênica , Interleucina-1alfa/metabolismo , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/metabolismo , Fosfoproteínas/genética , Remodelação Ventricular/genética , Animais , Apoptose/genética , Biomarcadores , Células Cultivadas , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Imunofluorescência , Interleucina-1alfa/genética , Masculino , Camundongos , Camundongos Knockout , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Fosfoproteínas/metabolismo , Transdução de Sinais
17.
Nat Commun ; 11(1): 3259, 2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32591509

RESUMO

Fusobacterium nucleatum is an oral anaerobe recently found to be prevalent in human colorectal cancer (CRC) where it is associated with poor treatment outcome. In mice, hematogenous F. nucleatum can colonize CRC tissue using its lectin Fap2, which attaches to tumor-displayed Gal-GalNAc. Here, we show that Gal-GalNAc levels increase as human breast cancer progresses, and that occurrence of F. nucleatum gDNA in breast cancer samples correlates with high Gal-GalNAc levels. We demonstrate Fap2-dependent binding of the bacterium to breast cancer samples, which is inhibited by GalNAc. Intravascularly inoculated Fap2-expressing F. nucleatum ATCC 23726 specifically colonize mice mammary tumors, whereas Fap2-deficient bacteria are impaired in tumor colonization. Inoculation with F. nucleatum suppresses accumulation of tumor infiltrating T cells and promotes tumor growth and metastatic progression, the latter two of which can be counteracted by antibiotic treatment. Thus, targeting F. nucleatum or Fap2 might be beneficial during treatment of breast cancer.


Assuntos
Neoplasias da Mama/microbiologia , Neoplasias da Mama/patologia , Progressão da Doença , Fusobacterium nucleatum/crescimento & desenvolvimento , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Contagem de Colônia Microbiana , DNA Bacteriano/genética , Modelos Animais de Doenças , Feminino , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/genética , Galactosamina/metabolismo , Galactose/metabolismo , Genoma Bacteriano/genética , Humanos , Imunidade/efeitos dos fármacos , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Metástase Neoplásica
18.
Front Immunol ; 10: 1710, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379884

RESUMO

Neutrophils are the most abundant population of white blood cells in the human circulation. They are terminally differentiated myeloid cells which were traditionally associated with fighting infections and inflammatory processes. While this perception of neutrophils is still widely prevalent, in the past decade it has become clear that neutrophils also play a critical role in tumor growth and progression. The unique tumor microenvironment, consisting of the non-malignant stroma that surrounds tumor cells, is shaped by numerous cues emanating from both tumor cells and stromal cells which support the growing tumor. Various immune cells, including neutrophils, make up a significant proportion of the tumor stroma. Immune cells exist for the protection of the host against various threats including the detection and elimination of cancerous cells. However, in the context of cancer immune cells are often coerced into a tumor supportive phenotype. This is also the case for neutrophils, which are often described to possess tumor promoting properties and to associate with poorer prognosis. The fact that neutrophils may contribute to tumor growth and progression suggests they may be targets for anti-cancer therapies. This review discusses the various functions neutrophils may play in cancer and the possibility of targeting these functions as a novel mode of immunotherapy.


Assuntos
Neoplasias/imunologia , Neutrófilos/imunologia , Animais , Humanos , Linfócitos/imunologia , Neoplasias/terapia
19.
Oncoimmunology ; 8(9): e1624129, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31428521

RESUMO

Neutrophils are a heterogeneous population of myeloid cells which may either promote or hinder tumor growth and progression. Anti-tumor neutrophils have the capacity to kill tumor cells in a contact-dependent manner. However, the molecular mechanisms underlying tumor cell recognition by neutrophils remained unexplored. Tumor cells were shown to express aberrant glycosylation patterns and neutrophils are equipped with receptors capable of recognizing such glycosylations. Accordingly, we hypothesized that the receptor for advanced glycation end products (RAGE) may facilitate neutrophil recognition of tumor cells. Indeed, RAGE decoy receptors and RAGE-specific blocking antibodies dramatically reduce tumor cell susceptibility to neutrophil cytotoxicity. Unexpectedly, we found that tumor cell RAGE rather than neutrophil RAGE is important for the killing process. We further identified neutrophil Cathepsin G as the neutrophil component interacting with tumor cell RAGE. Cathepsin G-deficient neutrophils show impaired ability to kill tumor cells, suggesting that RAGE-Cathepsin G interaction is required for neutrophil cytotoxicity. These data unravel new aspects of neutrophil anti-tumor activity and identify a novel role for RAGE and Cathepsin G in neutrophil-mediated cytotoxicity.

20.
Cell Rep ; 27(13): 3902-3915.e6, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31242422

RESUMO

Neutrophils are phenotypically heterogeneous and exert either anti- or pro-metastatic functions. We show that cancer-cell-derived G-CSF is necessary, but not sufficient, to mobilize immature low-density neutrophils (iLDNs) that promote liver metastasis. In contrast, mature high-density neutrophils inhibit the formation of liver metastases. Transcriptomic and metabolomic analyses of high- and low-density neutrophils reveal engagement of numerous metabolic pathways specifically in low-density neutrophils. iLDNs exhibit enhanced global bioenergetic capacity, through their ability to engage mitochondrial-dependent ATP production, and remain capable of executing pro-metastatic neutrophil functions, including NETosis, under nutrient-deprived conditions. We demonstrate that NETosis is an important neutrophil function that promotes breast cancer liver metastasis. iLDNs rely on the catabolism of glutamate and proline to support mitochondrial-dependent metabolism in the absence of glucose, which enables sustained NETosis. These data reveal that distinct pro-metastatic neutrophil populations exhibit a high degree of metabolic flexibility, which facilitates the formation of liver metastases.


Assuntos
Neoplasias Hepáticas/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neutrófilos/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Neutrófilos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...