Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 4(2): 102208, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37086410

RESUMO

Preserving structurally intact tissue through proper tissue fixation and cryopreservation minimizes tissue autolysis, desiccation, and enzymatic degradation. In this protocol, we describe the use of an optimized fresh freezing technique that cryopreserves the tissue and favors the retention of the natural protein structure of antigens. We also detail the use of cold, low-concentration paraformaldehyde (PFA) fixation to enhance tissue morphology and detection of fluorescent proteins, such as GFP and TdTomato, in tissues of genetically engineered mice. For complete details on the use and execution of this protocol, please refer to Chen et al. (2022)1 and El-Naccache et al. (2022).2.

2.
J Immunol ; 209(11): 2160-2171, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36426972

RESUMO

More than 2 billion people worldwide are infected with helminths. Thus, it is possible for individuals to experience concomitant infection with helminth and intracellular microbes. Although the helminth-induced type 2 response can suppress type 1 proinflammatory responses required for the immunity against intracellular pathogens in the context of a coinfection, conflicting evidence suggest that helminth infection can enhance antimicrobial immunity. Using a coinfection model with the intestinal helminth Heligmosomoides polygyrus followed by infection with Toxoplasma gondii in Mus Musculus, we showed that the complex and dynamic effect of helminth infection is highly suppressive during the innate phase (days 0-3) of T. gondii infection and less stringent during the acute phase (d10). Helminth coinfection had a strong suppressive effect on the neutrophil, monocytic, and early IFN-γ/IL-12 responses. The IFN-γ response was later restored by compensatory production from T cells despite decreased effector differentiation of T. gondii-specific CD8 T cells. In accordance with the attenuated IFN-γ response, parasite loads were elevated during the acute phase (d10) of T. gondii infection but were transiently controlled by the compensatory T cell response. Unexpectedly, 40% of helminth-coinfected mice exhibited a sustained weight loss phenotype during the postacute phase (d14-18) that was not associated with T. gondii outgrowth, indicating that coinfection led to decreased disease tolerance during T. gondii infection. Our work uncovers the dynamic nature of the helminth immunomodulatory effects on concomitant infections or immune responses and unveils a loss of disease tolerance phenotype triggered by coinfection with intestinal helminth.


Assuntos
Coinfecção , Nematospiroides dubius , Toxoplasma , Toxoplasmose , Animais , Camundongos , Tolerância Imunológica
3.
Cell Rep ; 40(5): 111150, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35926464

RESUMO

Intestinal nematode parasites can cross the epithelial barrier, causing tissue damage and release of danger-associated molecular patterns (DAMPs) that may promote host protective type 2 immunity. We investigate whether adenosine binding to the A2B adenosine receptor (A2BAR) on intestinal epithelial cells (IECs) plays an important role. Specific blockade of IEC A2BAR inhibits the host protective memory response to the enteric helminth, Heligmosomoides polygyrus bakeri (Hpb), including disruption of granuloma development at the host-parasite interface. Memory T cell development is blocked during the primary response, and transcriptional analyses reveal profound impairment of IEC activation. Extracellular ATP is visualized 24 h after inoculation and is shown in CD39-deficient mice to be critical for the adenosine production mediating the initiation of type 2 immunity. Our studies indicate a potent adenosine-mediated IEC pathway that, along with the tuft cell circuit, is critical for the activation of type 2 immunity.


Assuntos
Adenosina , Receptor A2B de Adenosina , Adenosina/metabolismo , Trifosfato de Adenosina , Animais , Células Epiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2B de Adenosina/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35994660

RESUMO

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Assuntos
Interleucina-33 , Placenta , Placentação , Útero , Animais , Decídua/irrigação sanguínea , Decídua/citologia , Decídua/crescimento & desenvolvimento , Decídua/imunologia , Feminino , Feto/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/deficiência , Interleucina-33/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Placenta/imunologia , Placenta/metabolismo , Gravidez , Útero/irrigação sanguínea , Útero/crescimento & desenvolvimento , Útero/imunologia , Útero/metabolismo
5.
Cell Rep ; 38(2): 110215, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021079

RESUMO

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Assuntos
Pulmão/imunologia , Macrófagos Alveolares/imunologia , Infecções por Strongylida/imunologia , Animais , Arginase/metabolismo , Diferenciação Celular , Citocinas , Feminino , Pulmão/parasitologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nippostrongylus , Infecções por Strongylida/parasitologia
6.
Trends Immunol ; 42(2): 151-164, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33386241

RESUMO

Type 2 immune responses are typically associated with protection against helminth infections and also with harmful inflammation in response to allergens. Recent advances have revealed that type 2 immunity also contributes to sterile inflammation, cancer, and microbial infections. However, the early events that initiate type 2 immune responses remain poorly defined. New insights reveal major contributions from danger-associated molecular patterns (DAMPs) in the initiation of type 2 immune responses. In this review, we examine the molecules released by the host and pathogens and the role they play in mediating the initiation of mammalian innate type 2 immune responses under a variety of conditions.


Assuntos
Helmintíase , Imunidade Inata , Alarminas , Alérgenos , Animais , Humanos , Inflamação
7.
Cell Host Microbe ; 27(2): 165-168, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32053785

RESUMO

Recent studies show that neutrophils mediate both tissue damage and host protection in response to multicellular parasites. In this issue of Cell Host & Microbe, Bouchery et al. demonstrate the importance of neutrophil extracellular traps in helminth damage after primary infections.


Assuntos
Armadilhas Extracelulares , Ancylostomatoidea , Animais , Desoxirribonucleases , Neutrófilos
8.
Semin Cell Dev Biol ; 88: 163-172, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29501617

RESUMO

Defensins have been long recognized as natural antimicrobial peptides, but they also possess diverse and versatile immune functions. Defensins can both induce inflammation and suppress inflammatory responses by acting on specific cells through distinct mechanisms. Defensins can also modulate the immune response by forming a complex with cellular molecules including proteins, nucleic acids, and carbohydrates. The mechanisms of defensin-mediated immune modulation appear to be cell-type and context specific. Because the levels of human defensins are often altered in response to infection or disease states, suggesting their clinical relevance, this review summarizes the complex immune functions of human defensins and their underlying mechanisms of action, which have implications for the development of new therapeutics.


Assuntos
Imunidade Inata , Interleucinas/imunologia , Receptores Toll-Like/imunologia , alfa-Defensinas/imunologia , beta-Defensinas/imunologia , Sequência de Aminoácidos , Animais , Carcinogênese/genética , Carcinogênese/imunologia , Carcinogênese/patologia , Células Epiteliais/imunologia , Regulação da Expressão Gênica , Humanos , Interleucinas/genética , Receptores CCR6/genética , Receptores CCR6/imunologia , Transdução de Sinais , Receptores Toll-Like/genética , alfa-Defensinas/genética , beta-Defensinas/genética
9.
Cell Rep ; 25(10): 2775-2783.e3, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517865

RESUMO

Emphysema results in destruction of alveolar walls and enlargement of lung airspaces and has been shown to develop during helminth infections through IL-4R-independent mechanisms. We examined whether interleukin 17A (IL-17A) may instead modulate development of emphysematous pathology in mice infected with the helminth parasite Nippostrongylus brasiliensis. We found that transient elevations in IL-17A shortly after helminth infection triggered subsequent emphysema that destroyed alveolar structures. Furthermore, lung B cells, activated through IL-4R signaling, inhibited early onset of emphysematous pathology. IL-10 and other regulatory cytokines typically associated with B regulatory cell function did not play a major role in this response. Instead, at early stages of the response, B cells produced high levels of the tissue-protective protein, Resistin-like molecule α (RELMα), which then downregulated IL-17A expression. These studies show that transient elevations in IL-17A trigger emphysema and reveal a helminth-induced immune regulatory mechanism that controls IL-17A and the severity of emphysema.


Assuntos
Linfócitos B/metabolismo , Enfisema/imunologia , Enfisema/parasitologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-17/metabolismo , Nippostrongylus/fisiologia , Infecções por Strongylida/parasitologia , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/imunologia , Animais , Anticorpos/farmacologia , Regulação para Baixo , Imunidade/efeitos dos fármacos , Pulmão/imunologia , Pulmão/parasitologia , Pulmão/patologia , Camundongos Endogâmicos BALB C , Fenótipo , Receptores de Interleucina-4/metabolismo , Transdução de Sinais
10.
Cell Host Microbe ; 22(5): 717, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29120747
11.
Cell Host Microbe ; 22(4): 429-431, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29024637

RESUMO

The determinants of helminth resistance are not well understood. In this issue of Cell Host & Microbe, Entwistle et al. (2017) provide intriguing evidence that a phospholipase A2 (Pla2gb1) produced by epithelial cells can impair larval development in helminths, providing a novel mechanism contributing to intestinal nematode resistance.


Assuntos
Fosfolipases A2 do Grupo IB
12.
PLoS Pathog ; 12(7): e1005801, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27463802

RESUMO

Studies have suggested that Epithelial-Mesenchymal Transition (EMT) and transformation is an important step in progression to cancer. Par3 (partitioning-defective protein) is a crucial factor in regulating epithelial cell polarity. However, the mechanism by which the latency associated nuclear antigen (LANA) encoded by Kaposi's Sarcoma associated herpesvirus (KSHV) regulates Par3 and EMTs markers (Epithelial-Mesenchymal Transition) during viral-mediated B-cell oncogenesis has not been fully explored. Moreover, several studies have demonstrated a crucial role for EMT markers during B-cell malignancies. In this study, we demonstrate that Par3 is significantly up-regulated in KSHV-infected primary B-cells. Further, Par3 interacted with LANA in KSHV positive and LANA expressing cells which led to translocation of Par3 from the cell periphery to a predominantly nuclear signal. Par3 knockdown led to reduced cell proliferation and increased apoptotic induction. Levels of SNAIL was elevated, and E-cadherin was reduced in the presence of LANA or Par3. Interestingly, KSHV infection in primary B-cells led to enhancement of SNAIL and down-regulation of E-cadherin in a temporal manner. Importantly, knockdown of SNAIL, a major EMT regulator, in KSHV cells resulted in reduced expression of LANA, Par3, and enhanced E-cadherin. Also, SNAIL bound to the promoter region of p21 and can regulate its activity. Further a SNAIL inhibitor diminished NF-kB signaling through upregulation of Caspase3 in KSHV positive cells in vitro. This was also supported by upregulation of SNAIL and Par3 in BC-3 transplanted NOD-SCID mice which has potential as a therapeutic target for KSHV-associated B-cell lymphomas.


Assuntos
Linfócitos B/virologia , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Viral/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Infecções por Herpesviridae/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos Virais/metabolismo , Western Blotting , Feminino , Imunofluorescência , Regulação da Expressão Gênica/fisiologia , Herpesvirus Humano 8 , Humanos , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
13.
Oncotarget ; 7(14): 18116-34, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26908453

RESUMO

Epstein-Barr virus (EBV), a gamma herpes virus is associated with B-cell malignancies. EBNA-3C is critical for in vitro primary B-cell transformation. Interestingly, the N terminal domain of EBNA3C which contains residues 130-159, interacts with various cellular proteins, such as p53, Mdm2, CyclinD1/Cdk6 complex, and E2F1. In the current reverse genetics study, we deleted the residues 130-159 aa within EBNA3C open reading frame (ORF) by BACmid recombinant engineering methodology. Our experiments demonstrated that deletion of the 130-159 aa showed a reduction in cell proliferation. Also, this recombinant virus showed with higher infectivity of human peripheral blood mononuclear cells (PBMCs) compared to wild type EBV. PBMCs- infected with recombinant EBV deleted for 130-159 residues have differential expression patterns for the p53/Mdm2, CyclinD1/Cdk6 and pRb/E2F1 pathways compared to wild type EBV-infected PBMCs. PBMCs infected with recombinant virus showed increased apoptotic cell death which further resulted in activation of polymerase 1 (PARP1), an important contributor to apoptotic signaling. Interestingly, cells infected with this recombinant virus showed a dramatic decrease in chromosomal instability, indicated by the presence of increased multinucleation and micronucleation. In addition infection with recombinant virus have increased cells in G0/G1 phase and decreased cells in S-G2M phase when compared to wild type infected cells. Thus, these differences in signaling activities due to 29 amino acid residues of EBNA3C is of particular significance in deregulation of cell proliferation in EBV-infected cells.


Assuntos
Apoptose/genética , Proliferação de Células/genética , Ciclina D1/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Herpesvirus Humano 4/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linfócitos B/metabolismo , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Transformação Celular Viral , Fator de Transcrição E2F1/metabolismo , Ativação Enzimática , Infecções por Vírus Epstein-Barr , Antígenos Nucleares do Vírus Epstein-Barr/genética , Células HEK293 , Humanos , Leucócitos Mononucleares/virologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Deleção de Sequência/genética
14.
Oncotarget ; 6(8): 5788-803, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25691063

RESUMO

In multicellular organisms p53 maintains genomic integrity through activation of DNA repair, and apoptosis. EBNA3C can down regulate p53 transcriptional activity. Aurora kinase (AK) B phosphorylates p53, which leads to degradation of p53. Aberrant expression of AK-B is a hallmark of numerous human cancers. Therefore changes in the activities of p53 due to AK-B and EBNA3C expression is important for understanding EBV-mediated cell transformation. Here we show that the activities of p53 and its homolog p73 are dysregulated in EBV infected primary cells which can contribute to increased cell transformation. Further, we showed that the ETS-1 binding site is crucial for EBNA3C-mediated up-regulation of AK-B transcription. Further, we determined the Ser 215 residue of p53 is critical for functional regulation by AK-B and EBNA3C and that the kinase domain of AK-B which includes amino acid residues 106, 111 and 205 was important for p53 regulation. AK-B with a mutation at residue 207 was functionally similar to wild type AK-B in terms of its kinase activities and knockdown of AK-B led to enhanced p73 expression independent of p53. This study explores an additional mechanism by which p53 is regulated by AK-B and EBNA3C contributing to EBV-induced B-cell transformation.


Assuntos
Aurora Quinase B/biossíntese , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Proteína Supressora de Tumor p53/genética , Antígenos Virais , Aurora Quinase B/genética , Linhagem Celular , Proliferação de Células/fisiologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Indução Enzimática , Antígenos Nucleares do Vírus Epstein-Barr/genética , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Fosforilação , Transfecção , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Proteínas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA