Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Infect Dis ; 218(5): 809-813, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29668950

RESUMO

Staphylococcus aureus is a predominant cause of fatal pneumonia following influenza A virus (IAV) infection. Herein we investigate the influence of antecedent IAV infection on S. aureus virulence gene expression. Using a murine model, comparing the USA300 and USA300ΔsaeR/S strains, we demonstrate that S. aureus pathogenesis following IAV infection is SaeR/S dependent. Furthermore, we show that IAV modulates the lung environment to rapidly up-regulate S. aureus virulence factors containing the SaeR-binding domain. Data demonstrate that the pathogen response to IAV infection impacts host outcome and provides evidence that the ability of S. aureus to sense and respond to the lung environment determines severity of pneumonia.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Orthomyxoviridae/complicações , Pneumonia Estafilocócica/imunologia , Proteínas Quinases/metabolismo , Staphylococcus aureus/patogenicidade , Fatores de Transcrição/metabolismo , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Feminino , Deleção de Genes , Masculino , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/patologia , Pneumonia Estafilocócica/genética , Proteínas Quinases/genética , Staphylococcus aureus/genética , Fatores de Transcrição/genética
2.
Am J Pathol ; 186(2): 259-69, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26687815

RESUMO

Pulmonary hypertension subsequent to an infectious disease can be due to vascular structural remodeling or to functional alterations within various vascular cell types. In our previous mouse model of Pneumocystis-associated pulmonary hypertension, we found that vascular remodeling was not responsible for observed increases in right ventricular pressures. Here, we report that the vascular dysfunction we observed could be explained by an enhanced response to endothelin-1 (20% greater reduction in lumen diameter, P ≤ 0.05), corresponding to an up-regulation of similar magnitude (P ≤ 0.05) of the endothelin A receptor in the lung tissue. This effect was potentially augmented by a decrease in production of the pulmonary vasodilator adrenomedullin of almost 70% (P ≤ 0.05). These changes did not occur in interferon-γ knockout mice similarly treated, which do not develop pulmonary hypertension under these circumstances. Surprisingly, we did not observe any relevant changes in the vascular endothelial nitric oxide synthase vasodilatory response, which is a common potential site of inflammatory alterations to pulmonary vascular function. Our results indicate the diverse mechanisms by which inflammatory responses to prior infections can cause functionally relevant changes in vascular responses in the lung, promoting the development of pulmonary hypertension.


Assuntos
Adrenomedulina/metabolismo , Endotelinas/metabolismo , Hipertensão Pulmonar/metabolismo , Pulmão/metabolismo , Pneumocystis/metabolismo , Artéria Pulmonar/metabolismo , Adrenomedulina/genética , Animais , Hipertensão Pulmonar/fisiopatologia , Interferon gama/metabolismo , Camundongos Endogâmicos BALB C , Óxido Nítrico Sintase Tipo III/metabolismo , Artéria Pulmonar/fisiopatologia , Regulação para Cima , Vasodilatadores/farmacologia
3.
J Immunol ; 195(11): 5347-57, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26519535

RESUMO

Although acquired bone marrow failure (BMF) is considered a T cell-mediated autoimmune disease, few studies have considered contributing roles of innate immune deviations following otherwise innocuous infections as a cause underlying the immune defects that lead to BMF. Type I IFN signaling plays an important role in protecting hematopoiesis during systemic stress responses to the opportunistic fungal pathogen Pneumocystis. During Pneumocystis lung infection, mice deficient in both lymphocytes and type I IFN receptor (IFrag(-/-)) develop rapidly progressing BMF associated with accelerated hematopoietic cell apoptosis. However, the communication pathway eliciting the induction of BMF in response to this strictly pulmonary infection has been unclear. We developed a conditional-null allele of Ifnar1 and used tissue-specific induction of the IFrag(-/-) state and found that, following Pneumocystis lung infection, type I IFNs act not only in the lung to prevent systemic immune deviations, but also within the progenitor compartment of the bone marrow to protect hematopoiesis. In addition, transfer of sterile-filtered serum from Pneumocystis-infected mice as well as i.p. injection of Pneumocystis into uninfected IFrag(-/-) mice induced BMF. Although specific cytokine deviations contribute to induction of BMF, immune-suppressive treatment of infected IFrag(-/-) mice ameliorated its progression but did not prevent loss of hematopoietic progenitor functions. This suggested that additional, noncytokine factors also target and impair progenitor functions; and interestingly, fungal ß-glucans were also detected in serum. In conclusion, our data demonstrate that type 1 IFN signaling protects hematopoiesis within the bone marrow compartment from the damaging effects of proinflammatory cytokines elicited by Pneumocystis in the lung and possibly at extrapulmonary sites via circulating fungal components.


Assuntos
Hematopoese/imunologia , Células-Tronco Hematopoéticas/citologia , Interferon Tipo I/imunologia , Pneumocystis/imunologia , Receptor de Interferon alfa e beta/genética , Anemia Aplástica , Animais , Apoptose , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Hematopoese/genética , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/imunologia , Proteínas de Homeodomínio/genética , Interferon gama/imunologia , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Pneumonia por Pneumocystis/imunologia , Transdução de Sinais/imunologia , beta-Glucanas/sangue
4.
Infect Immun ; 83(2): 743-58, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25452554

RESUMO

HIV infection results in a complex immunodeficiency due to loss of CD4(+) T cells, impaired type I interferon (IFN) responses, and B cell dysfunctions causing susceptibility to opportunistic infections such as Pneumocystis murina pneumonia and unexplained comorbidities, including bone marrow dysfunctions. Type I IFNs and B cells critically contribute to immunity to Pneumocystis lung infection. We recently also identified B cells as supporters of on-demand hematopoiesis following Pneumocystis infection that would otherwise be hampered due to systemic immune effects initiated in the context of a defective type I IFN system. While studying the role of type I IFNs in immunity to Pneumocystis infection, we discovered that mice lacking both lymphocytes and type I IFN receptor (IFrag(-/-)) developed progressive bone marrow failure following infection, while lymphocyte-competent type I IFN receptor-deficient mice (IFNAR(-/-)) showed transient bone marrow depression and extramedullary hematopoiesis. Lymphocyte reconstitution of lymphocyte-deficient IFrag(-/-) mice pointed to B cells as a key player in bone marrow protection. Here we define how B cells protect on-demand hematopoiesis following Pneumocystis lung infection in our model. We demonstrate that adoptive transfer of B cells into IFrag(-/-) mice protects early hematopoietic progenitor activity during systemic responses to Pneumocystis infection, thus promoting replenishment of depleted bone marrow cells. This activity is independent of CD4(+) T cell help and B cell receptor specificity and does not require B cell migration to bone marrow. Furthermore, we show that B cells protect on-demand hematopoiesis in part by induction of interleukin-10 (IL-10)- and IL-27-mediated mechanisms. Thus, our data demonstrate an important immune modulatory role of B cells during Pneumocystis lung infection that complement the modulatory role of type I IFNs to prevent systemic complications.


Assuntos
Linfócitos B/imunologia , Hematopoese/imunologia , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Receptor de Interferon alfa e beta/genética , Transferência Adotiva , Anemia Aplástica , Animais , Linfócitos B/transplante , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Hemoglobinúria Paroxística/imunologia , Hemoglobinúria Paroxística/microbiologia , Interferon Tipo I/genética , Interleucina-10/biossíntese , Interleucina-10/imunologia , Interleucinas/biossíntese , Interleucinas/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID
5.
J Immunol ; 191(7): 3884-95, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23975863

RESUMO

Although acquired bone marrow failure (BMF) is considered a T cell-mediated autoimmune disease, possible innate immune defects as a cause for systemic immune deviations in response to otherwise innocuous infections have not been extensively explored. In this regard, we recently demonstrated an important role of type I IFNs in protecting hematopoiesis during systemic stress responses to the opportunistic fungal pathogen Pneumocystis in lymphocyte-deficient mice. Mice deficient in both lymphocytes and type I IFN receptor (IFrag(-/-) mice) develop rapidly progressing BMF due to accelerated bone marrow (BM) cell apoptosis associated with innate immune deviations in the BM in response to Pneumocystis lung infection. However, the communication pathway between lung and BM eliciting the induction of BMF in response to this strictly pulmonary infection has been unclear. In this study, we report that absence of an intact type I IFN system during Pneumocystis lung infection not only causes BMF in lymphocyte-deficient mice but also transient BM stress in lymphocyte-competent mice. This is associated with an exuberant systemic IFN-γ response. IFN-γ neutralization prevented Pneumocystis lung infection-induced BM depression in type I IFN receptor-deficient mice and prolonged neutrophil survival time in BM from IFrag(-/-) mice. IL-1ß and upstream regulators of IFN-γ, IL-12, and IL-18 were also upregulated in lung and serum of IFrag(-/-) mice. In conjunction, there was exuberant inflammasome-mediated caspase-1 activation in pulmonary innate immune cells required for processing of IL-18 and IL-1ß. Thus, absence of type I IFN signaling during Pneumocystis lung infection may result in deregulation of inflammasome-mediated pulmonary immune activation, causing systemic immune deviations triggering BMF in this model.


Assuntos
Medula Óssea/metabolismo , Homeostase , Inflamassomos/metabolismo , Interferon Tipo I/metabolismo , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Caspase 1/metabolismo , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Homeostase/efeitos dos fármacos , Homeostase/genética , Interferon gama/antagonistas & inibidores , Interferon gama/sangue , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Pneumocystis/patogenicidade , Receptor de Interferon alfa e beta/genética
6.
Am J Pathol ; 181(1): 151-62, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22626807

RESUMO

HIV infection causes loss of CD4(+) T cells and type 1 interferon (IFN)-producing and IFN-responsive dendritic cells, resulting in immunodeficiencies and susceptibility to opportunistic infections, such as Pneumocystis. Osteoporosis and bone marrow failure are additional unexplained complications in HIV-positive patients and patients with AIDS, respectively. We recently demonstrated that mice that lack lymphocytes and IFN a/b receptor (IFrag(-/-)) develop bone marrow failure after Pneumocystis lung infection, whereas lymphocyte-deficient, IFN α/ß receptor-competent mice (RAG(-/-)) had normal hematopoiesis. Interestingly, infected IFrag(-/-) mice also exhibited bone fragility, suggesting loss of bone mass. We quantified bone changes and evaluated the potential connection between progressing bone fragility and bone marrow failure after Pneumocystis lung infection in IFrag(-/-) mice. We found that Pneumocystis infection accelerated osteoclastogenesis as bone marrow failure progressed. This finding was consistent with induction of osteoclastogenic factors, including receptor-activated nuclear factor-κB ligand and the proapoptotic factor tumor necrosis factor-related apoptosis-inducing ligand, in conjunction with their shared decoy receptor osteoprotegerin, in the bone marrow of infected IFrag(-/-) mice. Deregulation of this axis has also been observed in HIV-positive individuals. Biphosphonate treatment of IFrag(-/-) mice prevented bone loss and protected loss of hematopoietic precursor cells that maintained activity in vitro but did not prevent loss of mature neutrophils. Together, these data show that bone loss and bone marrow failure are partially linked, which suggests that the deregulation of the receptor-activated nuclear factor-κB ligand/osteoprotegerin/tumor necrosis factor-related apoptosis-inducing ligand axis may connect the two phenotypes in our model.


Assuntos
Interferon Tipo I/fisiologia , Osteoclastos/fisiologia , Osteoporose/microbiologia , Pneumonia por Pneumocystis/complicações , Síndrome de Resposta Inflamatória Sistêmica/microbiologia , Anemia Aplástica , Animais , Conservadores da Densidade Óssea/uso terapêutico , Medula Óssea/metabolismo , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Diferenciação Celular/fisiologia , Citocinas/metabolismo , Difosfonatos/uso terapêutico , Progressão da Doença , Fêmur/diagnóstico por imagem , Fêmur/patologia , Hemoglobinúria Paroxística/microbiologia , Hemoglobinúria Paroxística/fisiopatologia , Hemoglobinúria Paroxística/prevenção & controle , Camundongos , Camundongos Knockout , Camundongos SCID , Osteoclastos/patologia , Osteoporose/diagnóstico por imagem , Osteoporose/fisiopatologia , Osteoporose/prevenção & controle , Osteoprotegerina/metabolismo , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/fisiopatologia , Ligante RANK/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/complicações , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Microtomografia por Raio-X
7.
J Immunol ; 186(10): 5956-67, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21471447

RESUMO

We recently demonstrated that lack of type I IFN signaling (IFNAR knockout) in lymphocyte-deficient mice (IFrag(-/-)) results in bone marrow (BM) failure after Pneumocystis lung infection, whereas lymphocyte-deficient mice with intact IFNAR (RAG(-/-)) had normal hematopoiesis. In the current work, we performed studies to define further the mechanisms involved in the induction of BM failure in this system. BM chimera experiments revealed that IFNAR expression was required on BM-derived but not stroma-derived cells to prevent BM failure. Signals elicited after day 7 postinfection appeared critical in determining BM cell fate. We observed caspase-8- and caspase-9-mediated apoptotic cell death, beginning with neutrophils. Death of myeloid precursors was associated with secondary oxidative stress, and decreasing colony-forming activity in BM cell cultures. Treatment with N-acetylcysteine could slow the progression of, but not prevent, BM failure. Type I IFN signaling has previously been shown to expand the neutrophil life span and regulate the expression of some antiapoptotic factors. Quantitative RT-PCR demonstrated reduced mRNA abundance for the antiapoptotic factors BCL-2, IAP2, MCL-1, and others in BM cells from IFrag(-/-) compared with that in BM cells from RAG(-/-) mice at day 7. mRNA and protein for the proapoptotic cytokine TNF-α was increased, whereas mRNA for the growth factors G-CSF and GM-CSF was reduced. In vivo anti-TNF-α treatment improved precursor cell survival and activity in culture. Thus, we propose that lack of type I IFN signaling results in decreased resistance to inflammation-induced proapoptotic stressors and impaired replenishment by precursors after systemic responses to Pneumocystis lung infection. Our finding may have implications in understanding mechanisms underlying regenerative BM depression/failure during complex immune deficiencies such as AIDS.


Assuntos
Apoptose , Células da Medula Óssea/fisiologia , Hematopoese , Interferon Tipo I/imunologia , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/fisiopatologia , Acetilcisteína/farmacologia , Animais , Proteína 3 com Repetições IAP de Baculovírus , Células da Medula Óssea/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Genes RAG-1 , Genes bcl-2 , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Proteínas Inibidoras de Apoptose/genética , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Dados de Sequência Molecular , Proteína de Sequência 1 de Leucemia de Células Mieloides , Células Progenitoras Mieloides/imunologia , Neutrófilos/imunologia , Estresse Oxidativo , Pneumocystis/patogenicidade , Pneumonia por Pneumocystis/metabolismo , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/análise , Quimera por Radiação , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Ubiquitina-Proteína Ligases
8.
Am J Respir Cell Mol Biol ; 46(3): 290-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21960549

RESUMO

It is widely held that exposure to pathogens such as fungi can be an agent of comorbidity, such as exacerbation of asthma or chronic obstructive pulmonary disease. Although many studies have examined allergic responses to fungi and their effects on pulmonary function, the possible pathologic implications of the early innate responses to fungal pathogens have not been explored. We examined early responses to the atypical fungus Pneumocystis in two common strains of mice in terms of overall immunological response and related pathology, such as cell damage and airway hyperresponsiveness (AHR). We found a strong strain-specific response in BALB/c mice that included recruitment of neutrophils, NK, NKT, and CD4 T cells. This response was accompanied by elevated indicators of lung damage (bronchoalveolar lavage fluid albumin and LDH) and profound AHR. This early response was absent in C57BL/6 mice, although both strains exhibited a later response associated with the clearance of Pneumocystis. We found that this AHR could not be attributed exclusively to the presence of recruited neutrophils, NKT, NK, or CD4 cells or to the actions of IFN-γ or IL-4. However, in the absence of STAT6 signaling, AHR and inflammatory cell recruitment were virtually absent. Gene expression analysis indicated that this early response included activation of several transcription factors that could be involved in pulmonary remodeling. These results show that exposure to a fungus such as Pneumocystis can elicit pulmonary responses that may contribute to morbidity, even without prior sensitization, in the context of certain genetic backgrounds.


Assuntos
Hiper-Reatividade Brônquica/metabolismo , Imunidade Inata , Pneumopatias Fúngicas/metabolismo , Pulmão/metabolismo , Infecções por Pneumocystis/metabolismo , Fator de Transcrição STAT6/metabolismo , Albuminas/metabolismo , Animais , Antígenos CD1/genética , Antígenos CD1/metabolismo , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/microbiologia , Hiper-Reatividade Brônquica/fisiopatologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/microbiologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Interferon gama/deficiência , Interferon gama/genética , Interleucina-4/metabolismo , L-Lactato Desidrogenase/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/fisiopatologia , Pneumopatias Fúngicas/genética , Pneumopatias Fúngicas/imunologia , Pneumopatias Fúngicas/microbiologia , Pneumopatias Fúngicas/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/microbiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Infecções por Pneumocystis/genética , Infecções por Pneumocystis/imunologia , Infecções por Pneumocystis/microbiologia , Infecções por Pneumocystis/fisiopatologia , Receptores de Interleucina-4/deficiência , Receptores de Interleucina-4/genética , Receptores de Interleucina-8B/deficiência , Receptores de Interleucina-8B/genética , Fator de Transcrição STAT6/deficiência , Fator de Transcrição STAT6/genética , Transdução de Sinais , Especificidade da Espécie , Fatores de Tempo
9.
Infect Immun ; 79(5): 1905-14, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21343358

RESUMO

Infection with the opportunistic fungal pathogen Pneumocystis is assumed to pass without persistent pathology in immunocompetent hosts. However, when immunocompetent BALB/c mice were inoculated with Pneumocystis, a vigorous Th2-like pulmonary inflammation ensued and peaked at 14 days postinfection. This coincided with a 10-fold increase in the number of antigen-presenting cells (APCs) in the lung, and these cells were capable of presenting antigen in vitro, as well as greater uptake of antigen in vivo. When mice were presented with exogenous antigen at the 14-day time point of the infection, they developed respiratory sensitization to that antigen, in the form of increased airway hyperresponsiveness upon a later challenge, whereas mice not infected but presented with antigen did not. Like other forms of collateral sensitization, this response was dependent on interleukin-4 receptor signaling. This ability to facilitate sensitization to exogenous antigen has been previously reported for other infectious disease agents; however, Pneumocystis appears to be uniquely capable in this respect, as a single intranasal dose without added adjuvant, when it was administered at the appropriate time, was sufficient to initiate sensitization. Pneumocystis infection probably occurs in most humans during the first few years of life, and in the vast majority of cases, it fails to cause any overt direct pathology. However, as we show here, Pneumocystis can be an agent of comorbidity at this time by facilitating respiratory sensitization that may relate to the later development or exacerbation of obstructive airway disease.


Assuntos
Antígenos/imunologia , Infecções por Pneumocystis/imunologia , Hipersensibilidade Respiratória/imunologia , Animais , Separação Celular , Citocinas/imunologia , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Ovalbumina/imunologia , Receptores de Interleucina-4/deficiência , Receptores de Interleucina-4/imunologia
10.
Am J Pathol ; 176(6): 2806-18, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395428

RESUMO

Immune-reconstitution after highly active antiretroviral therapy (HAART) is often incomplete, and some HIV-infected individuals fail to regenerate type-I interferon (IFN)-producing pDCs. We recently demonstrated that during Pneumocystis (PC) infection in CD4 T cell-competent mice the absence of type-I IFN signaling results in chronic pulmonary inflammation and fibrosis despite clearance. Because the mechanisms involved are poorly understood, we further characterized the role of type-I IFN signaling in immune responses to PC. We show that type-I IFN signaling around day 7 postinfection is critical to the outcome of inflammation. Microarray analysis of pulmonary CD11c(+) cells revealed that at day 7 post infection, wild-type cells up-regulated type-I IFN-responsive genes as well as SOCS1, which is a critical negative-regulator of type-I IFN and IFN-gamma signaling. This was associated with an eosinophilic lung inflammation, PC clearance, and complete restitution. However, pulmonary CD11c(+) cells from IFNAR(-/-) mice demonstrated increased tumor necrosis factor (TNF)-alpha production and lacked SOCS1-induction at day 7. This was followed by a transient lymphocytic and IFN-gamma response before switching to a chronic eosinophilic inflammation of the lung. Early neutralization of TNF-alpha did not prevent chronic inflammation in IFNAR(-/-) mice, but treatment with an anti-IFN-gamma antibody did. We propose that during PC lung infection type-I IFNs induce SOCS1-associated regulatory mechanisms, which prevent excessive IFN-gamma-mediated responses that cause chronic lung damage. Therefore, partial immune-reconstitution in AIDS, attributable to reduced type-I IFN actions, might disrupt regulatory aspects of inflammation, causing unexplained chronic pulmonary complications as seen in some patients during HAART.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Inflamação , Interferon Tipo I/imunologia , Interferon gama/imunologia , Pulmão , Pneumonia por Pneumocystis , Transdução de Sinais/imunologia , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Antígeno CD11c/genética , Antígeno CD11c/imunologia , Perfilação da Expressão Gênica , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Camundongos SCID , Análise em Microsséries , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/microbiologia , Pneumonia por Pneumocystis/patologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Fator de Necrose Tumoral alfa/imunologia
11.
J Leukoc Biol ; 73(2): 306-14, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12554808

RESUMO

To elucidate the functions of circulating gammadelta T cells, in the absence of antigen stimulation, the differential gene expression of two circulating gammadelta T cell subsets was analyzed. The two subsets, with distinct trafficking phenotypes in young calves, were GD3.5(+), CD8(-), WC1(+) or GD3.5(-), CD2(+), WC1(-), and 90-100% CD8(+) and were sorted based on GD3.5 and gammadelta T cell receptor expression. Results from two different human arrays probed with cDNA from these gammadelta T cell subsets indicated that they have markedly different tissue-specific functions. The genes preferentially expressed by GD3.5(+) (CD8(-)) gammadelta T cells demonstrated that they were highly activated, proliferative, and inflammatory, whereas those expressed by GD3.5(-) (primarily CD8(+)) gammadelta T cells were involved in promoting quiescence, consistent with a role for gammadelta T cells as sentinel mucosal cells, and several were interferon-regulated genes. Gene expression and phenotypic assays indicated that CD8(+) gammadelta T cells were apoptotic, whereas CD8(-) gammadelta T cells were apoptosis-resistant. Differential expression of multiple genes was confirmed in both arrays: That of 14 genes was confirmed by quantitative reverse transcriptase-polymerase chain reaction and that of seven proteins was confirmed by flow cytometry. This novel, genomic analysis of circulating gammadelta T cell subsets, without confounding effects of the tissue microenvironment, offers new insight into the biology and development of neonatal gammadelta T cells.


Assuntos
Perfilação da Expressão Gênica , Receptores de Antígenos de Linfócitos T gama-delta/análise , Subpopulações de Linfócitos T/metabolismo , Animais , Bovinos , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos
12.
Front Oncol ; 4: 169, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25019063

RESUMO

Tumor vessels with resistance to anti-angiogenic therapy are characterized by the normalization of the vascular structures through integration of mature pericytes and smooth muscle cells (SMC) into the vessel wall, a process termed vessel stabilization. Unfortunately, stabilization-associated vascular remodeling can result in reduced sensitivity to subsequent anti-angiogenic therapy. We show here that blockade of VEGF by bevacizumab induces stabilization of angiogenic tumor blood vessels in human tumor specimen by recruiting Nestin-positive cells, whereas mature vessels down-regulated Nestin-expression. Using xenograft tumors growing on bone-marrow (BM) chimera of C57Bl/6 wildtype and Nestin-GFP transgenic mice, we show for first time that Nestin(+) cells inducing the maturation of tumor vessels do not originate from the BM but presumably reside within the adventitia of adult blood vessels. Complementary ex vivo experiments using explants of murine aortas revealed that Nestin(+) multipotent stem cells (MPSCs) are mobilized from their niche and differentiated into pericytes and SMC through the influence of tumor-cell-secreted factors. We conclude that tissue-resident Nestin(+) cells are more relevant than BM-derived cells for vessel stabilization and therefore have to be considered in future strategies for anti-angiogenic therapy. The identification of proteins mediating recruitment or differentiation of local Nestin(+) cells with potential stem cell character to angiogenic blood vessels may allow the definition of new therapeutic targets to reduce tumor resistance against anti-angiogenic drugs.

13.
Front Microbiol ; 5: 671, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25538692

RESUMO

K20 is a novel amphiphilic antifungal aminoglycoside that is synthetically derived from the antibiotic kanamycin A. Reported here are investigations of K20's antimicrobial activities, cytotoxicity, and fungicidal mechanism of action. In vitro growth inhibitory activities against a variety of human and plant pathogenic yeasts, filamentous fungi, and bacteria were determined using microbroth dilution assays and time-kill curve analyses, and hemolytic and animal cell cytotoxic activities were determined. Effects on Cryptococcus neoformans H-99 infectivity were determined with a preventive murine lung infection model. The antifungal mechanism of action was studied using intact fungal cells, yeast lipid mutants, and small unilamellar lipid vesicles. K20 exhibited broad-spectrum in vitro antifungal activities but not antibacterial activities. Pulmonary, single dose-administration of K20 reduced C. neoformans lung infection rates 4-fold compared to controls. Hemolysis and half-maximal cytotoxicities of mammalian cells occurred at concentrations that were 10 to 32-fold higher than fungicidal MICs. With fluorescein isothiocyanate (FITC), 20-25 mg/L K20 caused staining of >95% of C. neoformans and Fusarium graminearum cells and at 31.3 mg/L caused rapid leakage (30-80% in 15 min) of calcein from preloaded small unilamellar lipid vesicles. K20 appears to be a broad-spectrum fungicide, capable of reducing the infectivity of C. neoformans, and exhibits low hemolytic activity and mammalian cell toxicity. It perturbs the plasma membrane by mechanisms that are lipid modulated. K20 is a novel amphiphilic aminoglycoside amenable to scalable production and a potential lead antifungal for therapeutic and crop protection applications.

15.
J Immunol ; 178(10): 6604-15, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17475892

RESUMO

Loss of CD4 T cells is the hallmark of HIV infection. However, type I IFN-producing plasmacytoid dendritic cells may also be lost. This results in susceptibility to an opportunistic infection such as Pneumocystis pneumonia. In addition, regenerative bone marrow failure resulting in pancytopenia is another common problem in advanced stage AIDS. This may be linked to both the failing immune system and recurrent opportunistic infections. We generated lymphocyte-deficient type I IFN receptor-deficient mice (IFrag-/-) to study the effects on Pneumocystis infection of the lung. When IFrag-/- animals were infected with Pneumocystis they died between days 16 and 21 postinfection with minimal pneumonia but severe anemia due to complete bone marrow failure. This included the loss of uncommitted hemopoietic precursor cells. Bone marrow failure was prevented by the reconstitution of IFrag-/- mice with wild-type lymphocytes, especially B cells. T and B cells lacking type I IFN receptor signaling could only partially prevent bone marrow failure in response to Pneumocystis infection. However, the presence of T and B cells lacking type I IFN signaling resulted in compensatory extramedullary hemopoiesis in the liver and spleen. Lymphocyte support of the regenerative capacity of the bone marrow was provided by both type I IFN-dependent and -independent mechanisms that acted synergistically. Our findings point to the requirement of both type I IFNs and lymphocytes in the regenerative capabilities of the hemopoietic system under the pressure of Pneumocystis infection, but not during steady-state hemopoiesis. This may have implications in the management of pancytopenia in AIDS.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Hematopoese Extramedular/imunologia , Interferon-alfa/fisiologia , Interferon beta/fisiologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/patologia , Transdução de Sinais/imunologia , Anemia Aplástica/genética , Anemia Aplástica/imunologia , Anemia Aplástica/prevenção & controle , Animais , Subpopulações de Linfócitos B/patologia , Hematopoese Extramedular/genética , Interferon-alfa/deficiência , Interferon-alfa/genética , Interferon beta/deficiência , Interferon beta/genética , Fígado/citologia , Fígado/imunologia , Linfopenia/genética , Linfopenia/imunologia , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Pneumonia por Pneumocystis/sangue , Pneumonia por Pneumocystis/genética , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/genética , Baço/citologia , Baço/imunologia , Baço/transplante
16.
Am J Pathol ; 168(2): 466-75, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16436661

RESUMO

Pneumocystis spp. pneumonia (PCP) in humans and in surrogate animal species typically occurs in the absence of CD4 T cells, as takes place during acquired immune deficiency syndrome. However, patients treated with highly active anti-retroviral therapy sometimes exhibit an exacerbation of diseases such as PCP that coincides with resurgent CD4 T cells, a phenomenon known as immune reconstitution disease. We used an animal model of PCP using the B-cell-deficient muMT mouse together with antibody-mediated depletion of various T-cell subsets to examine the role of CD4 and CD8 T cells in the development of pathology in PCP. Although overt pathology occurs in the presence of CD4 T cells only, CD8 T cells only, or both, pulmonary injury occurs via different paths, depending on the complement of T cells present. Surprisingly, profound damage occurred when only CD4 T cells were present, and this pathology coincided with enhanced recruitment and activation of eosinophils and strong type 2 cytokine polarization in the alveolar environment. In addition, CD8 T cells can act to moderate this CD4 T cell-mediated pathology, possibly by increasing the ratio of putative CD25+ suppressor CD4 T cells to CD25- effector CD4 T cells.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Eosinófilos/imunologia , Pneumonia por Pneumocystis/patologia , Animais , Linfócitos B/patologia , Lavagem Broncoalveolar , Citometria de Fluxo , Pulmão/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia
17.
J Immunol ; 175(12): 8271-9, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16339567

RESUMO

Pneumocystis, a fungal, extracellular pathogen causes a life-threatening pneumonia in patients with severe immunodeficiencies. In the absence of CD4 T cells, Pneumocystis infection results in vigorous CD8 T cell influx into the alveolar and interstitial spaces of the lung. This response results in lung damage characterized by low pO2 and albumin leakage into the bronchoalveolar lavage fluid similar to other CD8 T cell-mediated interstitial lung diseases. How this extracellular pathogen elicits a CD8 T cell response is not clear, and it was the aim of our study to determine the Ag specificity of the recruited CD8 T cells and to determine whether MHC class I (MHC I) expression was necessary to initiate lung damage. Using an adoptive T cell-transfer model with either polyclonal wild-type CD8 T cells or transgenic influenza virus-specific CD8 T cells we found that CD8 T cell recruitment is Ag-specific and requires the continuous presence of the Pneumocystis pathogen. Bone marrow chimera experiments using Rag-1 and beta2-microglobulin-deficient mice as hosts demonstrated a requirement for MHC I expression on nonbone marrow-derived cells of the lung. This suggests either direct processing of Pneumocystis Ags by nonbone marrow-derived cells of the lung or the induction of lung damage triggered by a lung-specific autoantigen. Using perforin-, Fas-, and IFN-gamma-deficient animals, we showed that these molecules are not directly involved in the CD8-mediated lung damage. However, CD8 T cell-mediated lung damage is Ag-specific is induced by a MHC I-expressing nonbone marrow-derived cell in the lung and is dependent on the continued presence of live Pneumocystis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doenças Pulmonares Intersticiais/etiologia , Pulmão/patologia , Pneumocystis/imunologia , Transferência Adotiva , Animais , Quimiotaxia de Leucócito/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Pulmão/efeitos da radiação , Doenças Pulmonares Intersticiais/metabolismo , Camundongos , Camundongos SCID , Especificidade do Receptor de Antígeno de Linfócitos T
18.
J Immunol ; 174(9): 5462-71, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843544

RESUMO

Despite the advent of highly active antiretroviral therapy, pulmonary complications in AIDS are a common clinical problem. Pneumocystis jiroveci infection causes a life-threatening pneumonia, especially in individuals with CD4 T cell deficiencies as occurs in AIDS. Although Pneumocystis sp. is an extracellular fungal pathogen, CD8 T cells are the predominant lymphocyte recruited to the lung in CD4-deficient humans and mice during Pneumocystis pneumonia, and we have found that these CD8 T cells are responsible for subsequent lung damage in CD4 T cell-depleted mice. Comparing CD4 T cell-depleted IFN-alpha receptor knockout (KO) mice to wild-type mice, we found that this CD8 T cell recruitment and lung damage is type I IFN (IFN-alphabeta) dependent. However, in both CD4 competent, wild-type and IFN-alpha receptor (IFNAR) KO mice, Pneumocystis infection leads to an eosinophilic granulocyte influx with bronchial epithelial changes as seen in asthma. This response is delayed in IFNAR KO mice, as is pathogen clearance. Although the inflammation is transient in wild-type animals and resolves upon Pneumocystis clearance, it is more severe and persists through day 35 postinfection in IFNAR KO mice, leading to fibrosis. In addition, IFNAR KO, but not wild-type, mice mount a Pneumocystis-specific IgE response, an indicator of allergic sensitization. Thus, in the absence of IFNAR signaling and CD4 T cells, Pneumocystis-mediated lung damage does not occur, whereas in CD4-competent animals, the absence of IFNAR signaling results in an exacerbated Th2 response, asthma-like symptoms, and fibrosis. Therefore, both CD4 T cell- and type I IFN-mediated mechanisms can determine pulmonary complications from Pneumocystis infection.


Assuntos
Interferon-alfa/fisiologia , Interferon beta/fisiologia , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Receptores de Interferon/fisiologia , Transdução de Sinais/imunologia , Alérgenos/imunologia , Animais , Antígenos de Fungos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Imunoglobulina E/biossíntese , Interferon-alfa/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Pneumonia por Pneumocystis/genética , Pneumonia por Pneumocystis/patologia , Eosinofilia Pulmonar/imunologia , Eosinofilia Pulmonar/microbiologia , Eosinofilia Pulmonar/patologia , Receptor de Interferon alfa e beta , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais/genética
19.
J Immunol ; 170(1): 356-64, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12496420

RESUMO

Gene expression profiles were compared in circulating bovine GD3.5+ (CD8-) and GD3.5- (predominantly CD8+) gammadelta T cells using serial analysis of gene expression (SAGE). Approximately 20,000 SAGE tags were generated from each library. A comparison of the two libraries demonstrated 297 and 173 tags representing genes with 5-fold differential expression in GD3.5+ and GD3.5- gammadelta T cells, respectively. Consistent with their localization into sites of inflammation, GD3.5+ gammadelta T cells appeared transcriptionally and translationally more active than GD3.5- gammadelta cells. GD3.5- gammadelta T cells demonstrated higher expression of the cell proliferation inhibitor BAP 37, which was associated with their less activated gene expression phenotype. The immune regulatory and apoptosis-inducing molecule, galectin-1, was identified as a highly abundant molecule and was higher in GD3.5+gammadelta T cells. Surface molecules attributed to myeloid cells, such as CD14, CD68, and scavenger receptor-1, were identified in both populations. Furthermore, expression of B lymphocyte-induced maturation protein, a master regulator of B cell and myeloid cell differentiation, was identified by SAGE analysis and was confirmed at the RNA level to be selectively expressed in gammadelta T cells vs alphabeta T cells. These results provide new insights into the inherent differences between circulating gammadelta T cell subsets.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/imunologia , Imunofenotipagem , Receptores de Antígenos de Linfócitos T gama-delta/biossíntese , Receptores de Antígenos de Linfócitos T gama-delta/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Antígenos CD8/biossíntese , Bovinos , Separação Celular/métodos , Citometria de Fluxo/métodos , Regulação da Expressão Gênica/genética , Biblioteca Gênica , Ativação Linfocitária/genética , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Biossíntese de Proteínas/imunologia , Processamento de Proteína Pós-Traducional , Receptores de Antígenos de Linfócitos T gama-delta/sangue , Transcrição Gênica/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA