Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Physiol Rev ; 101(2): 417-425, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32790578

RESUMO

The host immune system shapes the fate of tumor progression. Hence, manipulating patients' immune system to activate host immune responses against cancer pathogenesis is a promising strategy to develop effective therapeutic interventions for metastatic and drug-resistant cancers. Understanding the dynamic mechanisms within the tumor microenvironment (TME) that contribute to heterogeneity and metabolic plasticity is essential to enhance the patients' responsiveness to immune targeted therapies. Riera-Domingo et al. (Riera-Domingo C, Audige A, Granja S, Cheng WC, Ho PC, Baltazar F, Stockmann C, Mazzone, M. Physiol Rev 100: 1-102, 2020) describe the immune landscape within the TME and highlight the significance of metabolic and hypoxic signatures that impact immune function and response to immunotherapy strategies. Current literature in this field confirms that targeting tumor metabolism and the acidic microenvironment commonly associated with tumors may present viable strategies to modulate the host immune system in favor of response to immune targeted therapies. However, development of better tools to understand tumor-immune interactions and identify mechanisms driving nonresponders, more innovative clinical trial design, and new therapies will need to be identified to move the field forward. Personalized immune therapies incorporating metabolic and microbiome-based gene signatures to influence the therapeutic response and novel methods to generate immunologically "hot" tumors are at the forefront of immunotherapy currently. The combination of these approaches with clinically approved immunotherapies will be valuable moving forward.


Assuntos
Imunoterapia/métodos , Neoplasias/terapia , Animais , Antineoplásicos/farmacologia , Humanos , Imunoterapia/tendências , Microambiente Tumoral/imunologia
2.
Proc Natl Acad Sci U S A ; 120(49): e2315096120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38011564

RESUMO

Hidradenitis suppurativa (HS) is a complex inflammatory skin disease with undefined mechanistic underpinnings. Here, we investigated HS epithelial cells and demonstrated that HS basal progenitors modulate their lineage restriction and give rise to pathogenic keratinocyte clones, resulting in epidermal hyperproliferation and dysregulated inflammation in HS. When comparing to healthy epithelial stem/progenitor cells, in HS, we identified changes in gene signatures that revolve around the mitotic cell cycle, DNA damage response and repair, as well as cell-cell adhesion and chromatin remodeling. By reconstructing cell differentiation trajectory and CellChat modeling, we identified a keratinocyte population specific to HS. This population is marked by S100A7/8/9 and KRT6 family members, triggering IL1, IL10, and complement inflammatory cascades. These signals, along with HS-specific proinflammatory cytokines and chemokines, contribute to the recruitment of certain immune cells during the disease progression. Furthermore, we revealed a previously uncharacterized role of S100A8 in regulating the local chromatin environment of target loci in HS keratinocytes. Through the integration of genomic and epigenomic datasets, we identified genome-wide chromatin rewiring alongside the switch of transcription factors (TFs), which mediated HS transcriptional profiles. Importantly, we identified numerous clinically relevant inflammatory enhancers and their coordinated TFs in HS basal CD49fhigh cells. The disruption of the S100A enhancer using the CRISPR/Cas9-mediated approach or the pharmacological inhibition of the interferon regulatory transcription factor 3 (IRF3) efficiently reduced the production of HS-associated inflammatory regulators. Our study not only uncovers the plasticity of epidermal progenitor cells in HS but also elucidates the epigenetic mechanisms underlying HS pathogenesis.


Assuntos
Hidradenite Supurativa , Humanos , Hidradenite Supurativa/genética , Pele/metabolismo , Epigenômica , Epigênese Genética , Células-Tronco/metabolismo , Cromatina/metabolismo
3.
Immunity ; 44(3): 582-596, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26921108

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disorder with increasing incidence. Mitochondrial oxidative stress in alveolar macrophages is directly linked to pulmonary fibrosis. Mitophagy, the selective engulfment of dysfunctional mitochondria by autophagasomes, is important for cellular homeostasis and can be induced by mitochondrial oxidative stress. Here, we show Akt1 induced macrophage mitochondrial reactive oxygen species (ROS) and mitophagy. Mice harboring a conditional deletion of Akt1 in macrophages (Akt1(-/-)Lyz2-cre) and Park2(-/-) mice had impaired mitophagy and reduced active transforming growth factor-ß1 (TGF-ß1). Although Akt1 increased TGF-ß1 expression, mitophagy inhibition in Akt1-overexpressing macrophages abrogated TGF-ß1 expression and fibroblast differentiation. Importantly, conditional Akt1(-/-)Lyz2-cre mice and Park2(-/-) mice had increased macrophage apoptosis and were protected from pulmonary fibrosis. Moreover, IPF alveolar macrophages had evidence of increased mitophagy and displayed apoptosis resistance. These observations suggest that Akt1-mediated mitophagy contributes to alveolar macrophage apoptosis resistance and is required for pulmonary fibrosis development.


Assuntos
Fibrose Pulmonar Idiopática/imunologia , Pulmão/patologia , Macrófagos Alveolares/fisiologia , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/genética , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Fibrose , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Mitofagia/genética , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/genética , Espécies Reativas de Oxigênio/metabolismo , Deleção de Sequência/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
4.
Semin Cell Dev Biol ; 128: 120-129, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35131152

RESUMO

Hidradenitis suppurativa (HS) is characterized by deep-seated, highly inflamed, and painful lumps/abscesses, fistulae, and sinus tracts that grow extensively deep in the dermis and are highly immunogenic in nature. In about one-third of the HS patients there is strong evidence for the role of γ-secretase mutations along with dysregulated Notch signaling. However, the contribution of dysregulated Notch signaling in HS pathogenesis in relation to hair follicle alterations and hyper-activation of the immune system remains undefined. A genome-wide association study (GWAS), proteomic data and functional investigations of identified sequence variants in HS pathology are not fully revealing. The disease initiation or progression may involve bacterial infection besides intrinsic functional defects in keratinocytes, which may be key to further exacerbate immune cell infiltration and cytokine production in and around the lesional tissue. The absence of a suitable animal model that could fully recapitulate the pathogenesis of HS is a major impediment for proper understanding the underlying mechanisms and development of effective treatments. The presence of extracellular matrix (ECM) degradation products along with dysregulation in keratinocytes and, dermal fibroblasts ultimately affect immune regulation and are various components of HS pathogenesis. Bacterial infection further exacerbates the complexity of the disease progression. While anti-TNFα therapy shows partial efficacy, treatment to cure HS is absent. Multiple clinical trials targeting various cytokines, complement C5a and ECM products are in progress. This review provides state-of-the-art information on these aspects with a focus on dysregulated keratinocyte and immune cells; and role of ECM, and Keratin functions in this regard.


Assuntos
Hidradenite Supurativa , Animais , Proteínas do Citoesqueleto/metabolismo , Estudo de Associação Genômica Ampla , Hidradenite Supurativa/genética , Hidradenite Supurativa/patologia , Humanos , Queratinas/genética , Queratinas/metabolismo , Proteômica , Transdução de Sinais/genética
5.
Am J Perinatol ; 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37216974

RESUMO

OBJECTIVE: This study aimed to investigate asthma medication reduction in the periconceptional period as it relates to asthma status and adverse outcomes in pregnancy. STUDY DESIGN: In a prospective cohort study, self-reported current and past asthma medications were collected and analyzes compared measures of asthma status in women who discontinued asthma medication in the 6 months prior to enrollment ("step-down") versus those who did not ("no change"). Evaluation of asthma was done at three study visits (one per trimester) and by daily diaries, including measures of lung function (percent predicted forced expiratory volume in 1 and 6 s [%FEV1, %FEV6], peak expiratory flow [%PEF], forced vital capacity [%FVC], FEV1 to FVC ratio [FEV1/FVC]), lung inflammation (fractional exhaled nitric oxide [FeNO], ppb), rate of asthma symptoms (activity limitation, night symptoms, rescue inhaler use, wheeze, shortness of breath, cough, chest tightness, chest pain), and rate of asthma exacerbations. Adverse pregnancy outcomes were also evaluated. Adjusted regression analyses examined whether adverse outcomes differed by periconceptional asthma medication changes. RESULTS: Of 279 participants included in analyses, 135 (48.4%) did not change asthma medication in the periconceptional period, whereas 144 (51.6%) reported a step down in medication. Those in the step-down group were more likely to have milder disease (88 [61.1%] in the step-down vs. 74 [54.8%] in the no change group), exhibited less activity limitation (rate ratio [RR]: 0.68, 95% confidence interval [CI]: 0.47-0.98), and experienced fewer asthma attacks (RR: 0.53, 95% CI: 0.34-0.84) during pregnancy. The step-down group had a nonsignificant increase in overall odds of experiencing an adverse pregnancy outcome (odds ratio: 1.62, 95% CI: 0.97-2.72). CONCLUSION: Over half of women with asthma reduce asthma medication in the periconceptional period. Although these women typically have milder disease, a step down in medication may be associated with an increased risk of adverse pregnancy outcomes. KEY POINTS: · Many women reduce their asthma medication in pregnancy.. · Reduction is more common among those with mild disease.. · Medication reduction may lead to adverse pregnancy outcomes..

6.
Am J Pathol ; 191(7): 1227-1239, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33887217

RESUMO

Increased apoptosis sensitivity of alveolar type 2 (ATII) cells and increased apoptosis resistance of (myo)fibroblasts, the apoptosis paradox, contributes to the pathogenesis of idiopathic pulmonary fibrosis (IPF). The mechanism underlying the apoptosis paradox in IPF lungs, however, is unclear. Aging is the greatest risk factor for IPF. In this study, we show, for the first time, that ATII cells from old mice are more sensitive, whereas fibroblasts from old mice are more resistant, to apoptotic challenges, compared with the corresponding cells from young mice. The expression of plasminogen activator inhibitor 1 (PAI-1), an important profibrogenic mediator, was significantly increased in both ATII cells and lung fibroblasts from aged mice. In vitro studies using PAI-1 siRNA and active PAI-1 protein indicated that PAI-1 promoted ATII cell apoptosis but protected fibroblasts from apoptosis, likely through dichotomous regulation of p53 expression. Deletion of PAI-1 in adult mice led to a reduction in p53, p21, and Bax protein expression, as well as apoptosis sensitivity in ATII cells, and their increase in the lung fibroblasts, as indicated by in vivo studies. This increase was associated with an attenuation of lung fibrosis after bleomycin challenge. Since PAI-1 is up-regulated in both ATII cells and fibroblasts in IPF, the results suggest that increased PAI-1 may underlie the apoptosis paradox of ATII cells and fibroblasts in IPF lungs.


Assuntos
Células Epiteliais Alveolares/metabolismo , Apoptose/fisiologia , Fibroblastos/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Fatores Etários , Células Epiteliais Alveolares/patologia , Animais , Fibroblastos/patologia , Fibrose Pulmonar Idiopática/patologia , Camundongos
7.
J Immunol ; 205(6): 1695-1708, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32817334

RESUMO

Innate lymphoid and adaptive immune cells are known to regulate epithelial responses, including mucous cell metaplasia (MCM), but their roles in mucoinflammatory airway diseases, such as cystic fibrosis, remain unknown. Scnn1b transgenic (Scnn1b-Tg+) mice, which recapitulate cystic fibrosis-like mucoinflammatory airway disease, deficient in innate lymphoid (Il2rg knockout mice [Il2rg KO]), adaptive immune (Rag1 knockout mice [Rag1 KO]), or both systems (Il2rg KO/Rag1 KO), were employed to investigate their respective contributions in the pathogenesis of mucoinflammatory airway disease. As previously reported, immunocompetent Tg+ juveniles exhibited spontaneous neonatal bacterial infections with robust mucoinflammatory features, including elevated expression of Th2-associated markers accompanied by MCM, elevated MUC5B expression, and airway mucus obstruction. The bacterial burden was increased in Il2rg KO/Tg+ juveniles but returned to significantly lower levels in Il2rg KO/Rag1 KO/Tg+ juveniles. Mechanistically, this improvement reflected reduced production of adaptive immunity-derived IL-10 and, in turn, increased activation of macrophages. Although all the mucoinflammatory features were comparable between the immunocompetent Tg+ and Rag1 KO/Tg+ juveniles, the Il2rg KO/Tg+ and Il2rg KO/Rag1 KO/Tg+ juveniles exhibited suppressed expression levels of Th2 markers, diminished MCM, suppressed MUC5B expression, and reduced mucus obstruction. Collectively, these data indicate that, in the context of airway mucus obstruction, the adaptive immune system suppresses antibacterial macrophage activation, whereas the innate lymphoid system contributes to MCM, mucin production, and mucus obstruction.


Assuntos
Fibrose Cística/imunologia , Células Epiteliais/metabolismo , Inflamação/imunologia , Mucina-5B/metabolismo , Doenças Respiratórias/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/patologia , Canais Epiteliais de Sódio/genética , Proteínas de Homeodomínio/genética , Humanos , Imunidade Inata , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucina-5B/genética , Regulação para Cima
8.
FASEB J ; 34(12): 15687-15700, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33047892

RESUMO

Extracellular trafficking of tumor necrosis factor receptor superfamily (TNFRSF) is tightly regulated, disruption of which triggers various autoinflammatory disorders, including TNF receptor-associated periodic syndrome (TRAPS). Here, we provide thus far unraveled molecular basis of noncysteine mutations in TNFR1 ectodomain where loss of an aromatic moiety in cysteine-rich domain (CRD) 2 results in TRAPS disease-associated phenotype. Our study characterized that a missense mutation on phenylalanine residue located in CRD2 (TNFR1F60V ) causes a delay in TNFR1 transport to cell membrane, leading to sustained receptor responsiveness and downstream NF-κB activation, characteristic of clinical manifestation of a prolonged fever. By creating and characterizing identical mutations on structurally conserved ectodomains of osteoprotegerin (OPG) and decoy receptor 3, other two secreted forms of TNFRSF, we further identified that a conserved aromatic residue at the A1 submodule of CRD2 (A1CRD2) confers structural integrity of ectodomain where aromatic sidechain deletion increases thermal instability, interfering with efficient posttranslational modification and subsequent receptor secretion. Interestingly, our functional analyses indicated that this particular noncysteine mutation is not associated with either protein misfolding or loss of function. Finally, by using a synthetic agonist, we demonstrated gain-of-function of the trafficking defect, suggesting the possibility of rescuing affected pathology in related disorders. Given the structural and topological similarities present in the ectodomains of TNFRSF members, our findings provide mechanistic insights of defects in subcellular trafficking of TNF receptors, reported in various TNFRSF-associated diseases.


Assuntos
Transporte Proteico/genética , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais/genética , Proteínas de Transporte/genética , Linhagem Celular , Linhagem Celular Tumoral , Febre/genética , Células HEK293 , Células HeLa , Humanos , Mutação de Sentido Incorreto/genética , NF-kappa B/genética
9.
Methods ; 177: 27-34, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31953152

RESUMO

Exosomes have been described as promising biomarkers for understanding disease progression and prognosis. These lipid membrane nanoparticles derived from airway cells have been shown to have immunomodulatory effects, such as driving inflammatory responses in asthma. These emerging evidences demonstrating an important pathophysiological role of exosomes warrants the development of novel approaches for isolation and rapid characterization of exosomes, which would be applicable for both translational and clinical studies. In this review article, we describe two methods of rapid exosomes characterization: (1) imaging flow cytometry using ImageStream; and (2) conventional flow cytometry using the BD Symphony A5 platform. We also explore sorting of exosomes using the BD Aria.


Assuntos
Asma/metabolismo , Centrifugação com Gradiente de Concentração/métodos , Exossomos/química , Citometria de Fluxo/métodos , Software , Ultracentrifugação/métodos , Antígenos CD/genética , Antígenos CD/metabolismo , Asma/diagnóstico , Asma/genética , Asma/patologia , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/química , Citometria de Fluxo/instrumentação , Expressão Gênica , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Pulmão/metabolismo , Pulmão/patologia
10.
J Infect Dis ; 222(11): 1882-1893, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-32492702

RESUMO

BACKGROUND: Streptococcus pneumoniae infection can result in bacteremia with devastating consequences including heart damage. Necroptosis is a proinflammatory form of cell death instigated by pore-forming toxins such as S. pneumoniae pneumolysin. Necroptosis-inhibiting drugs may lessen organ damage during invasive pneumococcal disease (IPD). METHODS: In vitro experiments were carried out with human and mouse cardiomyocytes. Long-term cardiac damage was assessed using high-resolution echocardiography in ampicillin-rescued mice 3 months after challenge with S. pneumoniae. Ponatinib, a necroptosis-inhibiting and Food and Drug Administration-approved drug for lymphocytic leukemia treatment, was administered intraperitoneally alongside ampicillin to test its therapeutic efficacy. Histology of heart sections included hematoxylin-eosin staining for overt damage, immunofluorescence for necroptosis, and Sirius red/fast green staining for collagen deposition. RESULTS: Cardiomyocyte death and heart damage was due to pneumolysin-mediated necroptosis. IPD leads to long-term cardiac damage, as evidenced by de novo collagen deposition in mouse hearts and a decrease in fractional shortening. Adjunct necroptosis inhibition reduced the number of S. pneumoniae foci observed in hearts of acutely infected mice and serum levels of troponin I. Ponatinib reduced collagen deposition and protected heart function in convalescence. CONCLUSIONS: Acute and long-term cardiac damage incurred during IPD is due in part to cardiomyocyte necroptosis. Necroptosis inhibitors may be a viable adjunct therapy.


Assuntos
Coração , Necroptose , Pneumonia Pneumocócica/complicações , Animais , Bacteriemia , Morte Celular , Modelos Animais de Doenças , Feminino , Imidazóis , Leucemia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Pneumocócicas , Proteínas Quinases , Piridazinas , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Streptococcus pneumoniae
11.
Lab Invest ; 100(12): 1503-1516, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32572176

RESUMO

Breast cancer (BCa) proliferates within a complex, three-dimensional microenvironment amid heterogeneous biochemical and biophysical cues. Understanding how mechanical forces within the tumor microenvironment (TME) regulate BCa phenotype is of great interest. We demonstrate that mechanical strain enhanced the proliferation and migration of both estrogen receptor+ and triple-negative (TNBC) human and mouse BCa cells. Furthermore, a critical role for exosomes derived from cells subjected to mechanical strain in these pro-tumorigenic effects was identified. Exosome production by TNBC cells increased upon exposure to oscillatory strain (OS), which correlated with elevated cell proliferation. Using a syngeneic, orthotopic mouse model of TNBC, we identified that preconditioning BCa cells with OS significantly increased tumor growth and myeloid-derived suppressor cells (MDSCs) and M2 macrophages in the TME. This pro-tumorigenic myeloid cell enrichment also correlated with a decrease in CD8+ T cells. An increase in PD-L1+ exosome release from BCa cells following OS supported additive T cell inhibitory functions in the TME. The role of exosomes in MDSC and M2 macrophage was confirmed in vivo by cytotracking fluorescent exosomes, derived from labeled 4T1.2 cells, preconditioned with OS. In addition, in vivo internalization and intratumoral localization of tumor-cell derived exosomes was observed within MDSCs, M2 macrophages, and CD45-negative cell populations following direct injection of fluorescently-labeled exosomes. Our data demonstrate that exposure to mechanical strain promotes invasive and pro-tumorigenic phenotypes in BCa cells, indicating that mechanical strain can impact the growth and proliferation of cancer cell, alter exosome production by BCa, and induce immunosuppression in the TME by dampening anti-tumor immunity.


Assuntos
Fenômenos Biomecânicos , Neoplasias da Mama , Estresse Mecânico , Microambiente Tumoral , Animais , Fenômenos Biomecânicos/imunologia , Fenômenos Biomecânicos/fisiologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/fisiopatologia , Carcinogênese , Movimento Celular , Proliferação de Células , Exossomos/metabolismo , Feminino , Humanos , Tolerância Imunológica , Células MCF-7 , Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Microambiente Tumoral/imunologia , Microambiente Tumoral/fisiologia
12.
Respir Res ; 21(1): 104, 2020 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-32375889

RESUMO

BACKGROUND: Recent studies suggest that alterations in lung microbiome are associated with occurrence of chronic lung diseases and transplant rejection. To investigate the host-microbiome interactions, we characterized the airway microbiome and metabolome of the allograft (transplanted lung) and native lung of single lung transplant recipients. METHODS: BAL was collected from the allograft and native lungs of SLTs and healthy controls. 16S rRNA microbiome analysis was performed on BAL bacterial pellets and supernatant used for metabolome, cytokines and acetylated proline-glycine-proline (Ac-PGP) measurement by liquid chromatography-high-resolution mass spectrometry. RESULTS: In our cohort, the allograft airway microbiome was distinct with a significantly higher bacterial burden and relative abundance of genera Acinetobacter & Pseudomonas. Likewise, the expression of the pro-inflammatory cytokine VEGF and the neutrophil chemoattractant matrikine Ac-PGP in the allograft was significantly higher. Airway metabolome distinguished the native lung from the allografts and an increased concentration of sphingosine-like metabolites that negatively correlated with abundance of bacteria from phyla Proteobacteria. CONCLUSIONS: Allograft lungs have a distinct microbiome signature, a higher bacterial biomass and an increased Ac-PGP compared to the native lungs in SLTs compared to the native lungs in SLTs. Airway metabolome distinguishes the allografts from native lungs and is associated with distinct microbial communities, suggesting a functional relationship between the local microbiome and metabolome.


Assuntos
Aloenxertos/fisiologia , Transplante de Pulmão/métodos , Pulmão/fisiologia , Metaboloma/fisiologia , Microbiota/fisiologia , Transplantados , Idoso , Aloenxertos/microbiologia , Feminino , Redes Reguladoras de Genes/fisiologia , Humanos , Pulmão/microbiologia , Masculino , Pessoa de Meia-Idade
13.
J Immunol ; 201(1): 278-295, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29752311

RESUMO

Myeloid-derived suppressor cells (MDSCs) are known suppressors of antitumor immunity, affecting amino acid metabolism and T cell function in the tumor microenvironment. However, it is unknown whether MDSCs regulate B cell responses during tumor progression. Using a syngeneic mouse model of lung cancer, we show reduction in percentages and absolute numbers of B cell subsets including pro-, pre-, and mature B cells in the bone marrow (BM) of tumor-bearing mice. The kinetics of this impaired B cell response correlated with the progressive infiltration of MDSCs. We identified that IL-7 and downstream STAT5 signaling that play a critical role in B cell development and differentiation were also impaired during tumor progression. Global impairment of B cell function was indicated by reduced serum IgG levels. Importantly, we show that anti-Gr-1 Ab-mediated depletion of MDSCs not only rescued serum IgG and IL-7 levels but also reduced TGF-ß1, a known regulator of stromal IL-7, suggesting MDSC-mediated regulation of B cell responses. Furthermore, blockade of IL-7 resulted in reduced phosphorylation of downstream STAT5 and B cell differentiation in tumor-bearing mice and administration of TGF-ß-blocking Ab rescued these IL-7-dependent B cell responses. Adoptive transfer of BM-derived MDSCs from tumor-bearing mice into congenic recipients resulted in significant reductions of B cell subsets in the BM and in circulation. MDSCs also suppressed B cell proliferation in vitro in an arginase-dependent manner that required cell-to-cell contact. Our results indicate that tumor-infiltrating MDSCs may suppress humoral immune responses and promote tumor escape from immune surveillance.


Assuntos
Linfócitos B/imunologia , Interleucina-7/imunologia , Neoplasias Pulmonares/imunologia , Células Supressoras Mieloides/imunologia , Fator de Transcrição STAT5/imunologia , Evasão Tumoral/imunologia , Transferência Adotiva , Animais , Linfócitos B/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Feminino , Imunoglobulina G/sangue , Interleucina-7/sangue , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/transplante , Fosforilação , Transdução de Sinais/imunologia , Fator de Crescimento Transformador beta/sangue , Microambiente Tumoral/imunologia
15.
Am J Respir Cell Mol Biol ; 60(3): 279-288, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30321056

RESUMO

Extracellular vesicles (EVs) are endosome and plasma membrane-derived nano-sized vesicles that participate in intercellular signaling. Although EV cargo may signal via multiple mechanisms, how signaling components on the surface of EVs mediate cellular signaling is less well understood. In this study, we show that fibroblast-derived EVs carry fibronectin on the vesicular surface, as evidenced by mass spectrometry-based proteomics (Sequential Window Acquisition of all Theoretical Mass Spectra) and flow-cytometric analyses. Fibroblasts undergoing replicative senescence or transforming growth factor ß1-induced senescence and fibroblasts isolated from human subjects with an age-related lung disorder, idiopathic pulmonary fibrosis, secreted higher numbers of EVs than their respective controls. Fibroblast-derived EVs induced an invasive phenotype in recipient fibroblasts. This invasive fibroblast phenotype was dependent on EV surface localization of fibronectin, interaction with the fibronectin receptor α5ß1 integrin, and activation of invasion-associated signaling pathways involving focal adhesion kinase and Src family kinases. EVs in the cellular supernatant, unbound to the extracellular matrix, were capable of mediating invasion signaling on recipient fibroblasts, supporting a direct interaction of EV surface fibronectin with the plasma membrane of recipient cells. Together, these studies uncover a novel mechanism of EV signaling of fibroblast invasion that may be relevant in the pathogenesis of fibrotic diseases and cancer.


Assuntos
Vesículas Extracelulares/metabolismo , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Senescência Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Integrina alfa5beta1/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Quinases da Família src/metabolismo
16.
PLoS Pathog ; 13(8): e1006582, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28841717

RESUMO

For over 130 years, invasive pneumococcal disease has been associated with the presence of extracellular planktonic pneumococci, i.e. diplococci or short chains in affected tissues. Herein, we show that Streptococcus pneumoniae that invade the myocardium instead replicate within cellular vesicles and transition into non-purulent biofilms. Pneumococci within mature cardiac microlesions exhibited salient biofilm features including intrinsic resistance to antibiotic killing and the presence of an extracellular matrix. Dual RNA-seq and subsequent principal component analyses of heart- and blood-isolated pneumococci confirmed the biofilm phenotype in vivo and revealed stark anatomical site-specific differences in virulence gene expression; the latter having major implications on future vaccine antigen selection. Our RNA-seq approach also identified three genomic islands as exclusively expressed in vivo. Deletion of one such island, Region of Diversity 12, resulted in a biofilm-deficient and highly inflammogenic phenotype within the heart; indicating a possible link between the biofilm phenotype and a dampened host-response. We subsequently determined that biofilm pneumococci released greater amounts of the toxin pneumolysin than did planktonic or RD12 deficient pneumococci. This allowed heart-invaded wildtype pneumococci to kill resident cardiac macrophages and subsequently subvert cytokine/chemokine production and neutrophil infiltration into the myocardium. This is the first report for pneumococcal biofilm formation in an invasive disease setting. We show that biofilm pneumococci actively suppress the host response through pneumolysin-mediated immune cell killing. As such, our findings contradict the emerging notion that biofilm pneumococci are passively immunoquiescent.


Assuntos
Biofilmes , Macrófagos/imunologia , Miocardite/imunologia , Miocardite/microbiologia , Infecções Pneumocócicas/imunologia , Transcriptoma , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Infecções Pneumocócicas/genética , Análise de Componente Principal , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/imunologia , Virulência/genética , Virulência/imunologia
17.
Immunity ; 33(1): 2-4, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20643332

RESUMO

Follicular dendritic cells (FDCs) are key organizers of B cell follicles and germinal centers. In this issue of Immunity, Suzuki et al. (2010) and Garin et al. (2010) identify the roles of Toll-like receptors in the responses of FDCs, providing a unique link between innate and adaptive immunity.

18.
Am J Respir Crit Care Med ; 198(10): 1288-1301, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29897791

RESUMO

RATIONALE: Cigarette smoking is prevalent in the United States and is the leading cause of preventable diseases. A prominent complication of smoking is an increase in lower respiratory tract infections (LRTIs). Although LRTIs are known to be increased in subjects that smoke, the mechanism(s) by which this occurs is poorly understood. OBJECTIVES: Determine how cigarette smoke (CS) reduces reactive oxygen species (ROS) production by the phagocytic NOX2 (NADPH oxidase 2), which is essential for innate immunity in lung macrophages. METHODS: NOX2-derived ROS and Rac2 (Ras-related C3 botulinum toxin substrate 2) activity were determined in BAL cells from wild-type and Rac2-/- mice exposed to CS or cadmium and in BAL cells from subjects that smoke. Host defense to respiratory pathogens was analyzed in mice infected with Streptococcus pneumoniae. MEASUREMENTS AND MAIN RESULTS: NOX2-derived ROS in BAL cells was reduced in mice exposed to CS via inhibition of the small GTPase Rac2. These mice had greater bacterial burden and increased mortality compared with air-exposed mice. BAL fluid from CS-exposed mice had increased levels of cadmium, which mediated the effect on Rac2. Similar observations were seen in human subjects that smoke. To support the importance of Rac2 in the macrophage immune response, overexpression of constitutively active Rac2 by lentiviral administration increased NOX2-derived ROS, decreased bacterial burden in lung tissue, and increased survival compared with CS-exposed control mice. CONCLUSIONS: These observations suggest that therapies to maintain Rac2 activity in lung macrophages restore host defense against respiratory pathogens and diminish the prevalence of LRTIs in subjects that smoke.


Assuntos
Fumar Cigarros/efeitos adversos , Fumar Cigarros/imunologia , Pneumonia/etiologia , Pneumonia/imunologia , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/imunologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Inata/imunologia , Pulmão/imunologia , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Espécies Reativas de Oxigênio/imunologia , Índice de Gravidade de Doença , Proteína RAC2 de Ligação ao GTP
19.
J Immunol ; 196(5): 2239-48, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26829988

RESUMO

We used two different infection models to investigate the kinetics of the PcpA-dependent pneumococcal disease in mice. In a bacteremic pneumonia model, we observed a PcpA-dependent increase in bacterial burden in the lungs, blood, liver, bronchoalveolar lavage, and spleens of mice at 24 h postinfection. This PcpA-dependent effect on bacterial burden appeared earlier (within 12 h) in the focal pneumonia model, which lacks bacteremia or sepsis. Histological changes show that the ability of pneumococci to make PcpA was associated with unresolved inflammation in both models of infection. Using our bacteremic pneumonia model we further investigated the effects of PcpA on recruitment of innate immune regulatory cells. The presence of PcpA was associated with increased IL-6 levels, suppressed production of TRAIL, and reduced infiltration of polymorphonuclear cells. The ability of pneumococci to make PcpA negatively modulated both the infiltration and apoptosis of macrophages and the recruitment of myeloid-derived suppressor-like cells. The latter have been shown to facilitate the clearance and control of bacterial pneumonia. Taken together, the ability to make PcpA was strongly associated with increased bacterial burden, inflammation, and negative regulation of innate immune cell recruitment to the lung tissue during bacteremic pneumonia.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Transporte/imunologia , Imunomodulação , Células Mieloides/imunologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/imunologia , Animais , Bacteriemia , Carga Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Regulação Bacteriana da Expressão Gênica , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Mutação , Células Mieloides/metabolismo , Pneumonia Pneumocócica/mortalidade , Pneumonia Pneumocócica/patologia , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA