Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
bioRxiv ; 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37292721

RESUMO

The majority of the world population carry the gastric pathogen Helicobacter pylori. Fortunately, most individuals experience only low-grade or no symptoms, but in many cases the chronic inflammatory infection develops into severe gastric disease, including duodenal ulcer disease and gastric cancer. Here we report on a protective mechanism where H. pylori attachment and accompanying chronic mucosal inflammation can be reduced by antibodies that are present in a vast majority of H. pylori carriers. These antibodies block binding of the H. pylori attachment protein BabA by mimicking BabA's binding to the ABO blood group glycans in the gastric mucosa. However, many individuals demonstrate low titers of BabA blocking antibodies, which is associated with an increased risk for duodenal ulceration, suggesting a role for these antibodies in preventing gastric disease.

2.
FEBS Lett ; 595(16): 2160-2168, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34216493

RESUMO

The persistence of Helicobacter pylori in the human gastric mucosa implies that the immune response fails to clear the infection. We found that H. pylori compromises the antigen presentation ability of macrophages, because of the decline of the presenting molecules HLA-II. Here, we reveal that the main bacterial factor responsible for this effect is ADP-heptose, an intermediate metabolite in the biosynthetic pathway of lipopolysaccharide (LPS) that elicits a pro-inflammatory response in gastric epithelial cells. In macrophages, it upregulates the expression of miR146b which, in turn, would downmodulate CIITA, the master regulator for HLA-II genes. Hence, H. pylori, utilizing ADP-heptose, exploits a specific arm of macrophage response to establish its survival niche in the face of the immune defense elicited in the gastric mucosa.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Helicobacter pylori/fisiologia , Heptoses/farmacologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Macrófagos/efeitos dos fármacos , Helicobacter pylori/metabolismo , Heptoses/química , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas Nucleares/metabolismo , Transativadores/metabolismo
3.
Gastroenterology ; 159(1): 169-182.e8, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32169428

RESUMO

BACKGROUND & AIMS: Helicobacter pylori induces strong inflammatory responses that are directed at clearing the infection, but if not controlled, these responses can be harmful to the host. We investigated the immune-regulatory effects of the innate immune molecule, nucleotide-binding oligomerization domain-like receptors (NLR) family CARD domain-containing 5 (NLRC5), in patients and mice with Helicobacter infection. METHODS: We obtained gastric biopsies from 30 patients in Australia. We performed studies with mice that lack NLRC5 in the myeloid linage (Nlrc5møKO) and mice without Nlrc5 gene disruption (controls). Some mice were gavaged with H pylori SS1 or Helicobacter felis; 3 months later, stomachs, spleens, and sera were collected, along with macrophages derived from bone marrow. Human and mouse gastric tissues and mouse macrophages were analyzed by histology, immunohistochemistry, immunoblots, and quantitative polymerase chain reaction. THP-1 cells (human macrophages, controls) and NLRC5-/- THP-1 cells (generated by CRISPR-Cas9 gene editing) were incubated with Helicobacter and gene expression and production of cytokines were analyzed. RESULTS: Levels of NLRC5 messenger RNA were significantly increased in gastric tissues from patients with H pylori infection, compared with patients without infection (P < .01), and correlated with gastritis severity (P < .05). H pylori bacteria induced significantly higher levels of chemokine and cytokine production by NLRC5-/- THP-1 macrophages than by control THP-1 cells (P < .05). After 3 months of infection with H felis, Nlrc5mø-KO mice developed gastric hyperplasia (P < .0001), splenomegaly (P < .0001), and increased serum antibody titers (P < .01), whereas control mice did not. Nlrc5mø-KO mice with chronic H felis infection had increased numbers of gastric B-cell follicles expressing CD19 (P < .0001); these follicles had features of mucosa-associated lymphoid tissue lymphoma. We identified B-cell-activating factor as a protein that promoted B-cell hyperproliferation in Nlrc5mø-KO mice. CONCLUSIONS: NLRC5 is a negative regulator of gastric inflammation and mucosal lymphoid formation in response to Helicobacter infection. Aberrant NLRC5 signaling in macrophages can promote B-cell lymphomagenesis during chronic Helicobacter infection.


Assuntos
Infecções por Helicobacter/complicações , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfoma de Zona Marginal Tipo Células B/imunologia , Neoplasias Gástricas/imunologia , Animais , Linfócitos B/imunologia , Biópsia , Proliferação de Células , Modelos Animais de Doenças , Feminino , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Regulação Neoplásica da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter felis/imunologia , Helicobacter pylori/imunologia , Humanos , Hiperplasia/imunologia , Hiperplasia/microbiologia , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Tecido Linfoide/patologia , Linfoma de Zona Marginal Tipo Células B/microbiologia , Linfoma de Zona Marginal Tipo Células B/patologia , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais/imunologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Células THP-1
4.
Am J Obstet Gynecol ; 221(5): 457-469, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31288009

RESUMO

Research on the placenta as the interface between the mother and the fetus has been undertaken for some 150 years, and in 2 subsequent reviews, we attempted to summarize the situation. In the first part, we described the discovery of unique physiological modifications of the uteroplacental spiral arteries, enabling them to cope with a major increase in blood flow necessary to ensure proper growth of the fetus. These consist of an invasion of the arterial walls by trophoblast and a progressive disappearance of its normal structure. Researchers then turned to the pathophysiology of the placental bed and in particular to its maternal vascular tree. This yielded vital information for a better understanding of the so-called great obstetrical syndromes (preeclampsia, fetal growth restriction, premature labor and delivery, placenta accreta). Systematic morphological investigations of the uteroplacental vasculature showed that preeclampsia is associated with decreased or failed transformation of spiral arteries and the persistence of endothelial and smooth muscle cells in segments of their myometrial portion. Here we report on recent functional investigations of the placental bed, including in situ biophysical studies of uteroplacental blood flow and vascular resistance, and manipulation of uteroplacental perfusion. These new methodologies have provided a novel way of identifying pregnancies in which remodeling is impaired. In animals it is now possible to manipulate uteroplacental blood flow, leading to an enhancement of fetal growth; this opens the way to trials in abnormal human pregnancies. In this second part, we explored a new, extremely important area of research that deals with the role of specific subsets of leukocytes and macrophages in the placental bed. The human first-trimester decidua is rich in leukocytes called uterine natural killer cells. Both macrophages and uterine natural killer cells increase in number from the secretory endometrium to early pregnancy and play a critical role in mediating the process of spiral artery transformation by inducing initial structural changes. It seems therefore that vascular remodeling of spiral arteries is initiated independently of trophoblast invasion. Dysregulation of the immune system may lead to reproductive failure or pregnancy complications, and in this respect, recent studies have advanced our understanding of the mechanisms regulating immunological tolerance during pregnancy, with several mechanisms being proposed for the development of tolerance to the semiallogeneic fetus. In particular, these include several strategies by which the trophoblast avoids maternal recognition. Finally, an important new dimension is being explored: the likelihood that pregnancy syndromes and impaired uteroplacental vascular remodeling may be linked to future maternal and even the child's cardiovascular disease risk. The functional evidence underlying these observations will be discussed.


Assuntos
Placenta/citologia , Placenta/imunologia , Placentação , Doenças Cardiovasculares/etiologia , Decídua/citologia , Feminino , História do Século XX , História do Século XXI , Humanos , Tolerância Imunológica , Células Matadoras Naturais/metabolismo , Leucócitos/metabolismo , Macrófagos/metabolismo , Placenta/irrigação sanguínea , Gravidez , Fluxo Sanguíneo Regional , Risco , Células Estromais/metabolismo , Remodelação Vascular , Resistência Vascular
5.
Haematologica ; 104(10): 2040-2052, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30819907

RESUMO

The Shc family adaptor p66Shc acts as a negative regulator of proliferative and survival signals triggered by the B-cell receptor and, by enhancing the production of reactive oxygen species, promotes oxidative stress-dependent apoptosis. Additionally, p66Shc controls the expression and function of chemokine receptors that regulate lymphocyte traffic. Chronic lymphocytic leukemia cells have a p66Shc expression defect which contributes to their extended survival and correlates with poor prognosis. We analyzed the impact of p66Shc ablation on disease severity and progression in the Eµ-TCL1 mouse model of chronic lymphocytic leukemia. We showed that Eµ-TCL1/p66Shc-/- mice developed an aggressive disease that had an earlier onset, occurred at a higher incidence and led to earlier death compared to that in Eµ-TCL1 mice. Eµ-TCL1/p66Shc-/- mice displayed substantial leukemic cell accumulation in both nodal and extranodal sites. The target organ selectivity correlated with upregulation of chemokine receptors whose ligands are expressed therein. This also applied to chronic lymphocytic leukemia cells, where chemokine receptor expression and extent of organ infiltration were found to correlate inversely with these cells' level of p66Shc expression. p66Shc expression declined with disease progression in Eµ-TCL1 mice and could be restored by treatment with the Bruton tyrosine kinase inhibitor ibrutinib. Our results highlight p66Shc deficiency as an important factor in the progression and severity of chronic lymphocytic leukemia and underscore p66Shc expression as a relevant therapeutic target.


Assuntos
Carcinogênese/metabolismo , Leucemia Linfocítica Crônica de Células B/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Receptores de Quimiocinas/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/deficiência , Animais , Carcinogênese/genética , Carcinogênese/patologia , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Receptores de Quimiocinas/genética , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/metabolismo
6.
Clin Immunol ; 201: 15-19, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30742970

RESUMO

CD25 deficiency is a very rare autosomal recessive disorder that shows a clinical phenotype highly overlapping IPEX syndrome with an increased susceptibility to viral, bacterial, and fungal infections. It is due to mutations in the IL2Rα gene that codes for the α subunit of the IL2 receptor complex. Here we report the characterization of a novel IL2Rα gene mutation leading to a severe protein conformational alteration that abrogates its cell surface expression in a child presenting with early-onset IPEX-like disorder. Cytofluorimetric analysis revealed the total absence of CD25 cell surface expression and addressed IL2Rα molecular investigation. The early clinical and molecular diagnosis of CD25 deficiency in this patient promptly led to hematopoietic stem cell transplantation (HSCT), allowing complete resolution of the symptoms and definitive cure of the disease.


Assuntos
Síndromes de Imunodeficiência/genética , Subunidade alfa de Receptor de Interleucina-2/deficiência , Citocinas/imunologia , Humanos , Síndromes de Imunodeficiência/diagnóstico , Síndromes de Imunodeficiência/imunologia , Lactente , Subunidade alfa de Receptor de Interleucina-2/química , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Mutação , Conformação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/imunologia
7.
Cell Microbiol ; 21(5): e13006, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30646431

RESUMO

Helicobacter pylori (HP) is a Gram-negative bacterium that chronically infects the stomach of more than 50% of human population and represents a major cause of gastric cancer, gastric lymphoma, gastric autoimmunity, and peptic ulcer. It still remains to be elucidated, which HP virulence factors are important in the development of gastric disorders. Here, we analysed the role of the HP protein HP1454 in the host-pathogen interaction. We found that a significant proportion of T cells isolated from HP patients with chronic gastritis and gastric adenocarcinoma proliferated in response to HP1454. Moreover, we demonstrated in vivo that HP1454 protein drives Th1/Th17 inflammatory responses. We further analysed the in vitro response of human T cells exposed either to an HP wild-type strain or to a strain with a deletion of the hp1454 gene, and we revealed that HP1454 triggers the T-cell antigen receptor-dependent signalling and lymphocyte proliferation, as well as the CXCL12-dependent cell adhesion and migration. Our study findings prove that HP1454 is a crucial bacterial factor that exerts its proinflammatory activity by directly modulating the T-cell response. The relevance of these results can be appreciated by considering that compelling evidence suggest that chronic gastric inflammation, a condition that paves the way to HP-associated diseases, is dependent on T cells.


Assuntos
Adenocarcinoma/imunologia , Gastrite/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/metabolismo , Lipoproteínas/imunologia , Neoplasias Gástricas/imunologia , Linfócitos T/imunologia , Adenocarcinoma/microbiologia , Idoso , Adesão Celular/imunologia , Diferenciação Celular/imunologia , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Movimento Celular/imunologia , Feminino , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/ultraestrutura , Gastrite/microbiologia , Regulação da Expressão Gênica/imunologia , Helicobacter pylori/genética , Helicobacter pylori/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Neoplasias Gástricas/microbiologia , Células Th1/imunologia , Células Th17/imunologia
8.
Oncogene ; 37(11): 1534-1550, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29326436

RESUMO

Neoplastic cell traffic abnormalities are central to the pathogenesis of chronic lymphocytic leukemia (CLL). Enhanced CXC chemokine receptor-4 (CXCR4) and chemokine receptor-7 (CCR7) recycling contributes to the elevated surface levels of these receptors on CLL cells. Here we have addressed the role of p66Shc, a member of the Shc family of protein adaptors the expression of which is defective in CLL cells, in CXCR4/CCR7 recycling. p66Shc reconstitution in CLL cells reduced CXCR4/CCR7 recycling, lowering their surface levels and attenuating B-cell chemotaxis, due to their accumulation in Rab5+ endosomes as serine-phosphoproteins bound to ß-arrestin. This results from the ability of p66Shc to inhibit Ca2+ and PP2B-dependent CXCR4/CCR7 dephosphorylation and ß-arrestin release. We also show that ibrutinib, a Btk inhibitor that promotes leukemic cell mobilization from lymphoid organs, reverses the CXCR4/CCR7 recycling abnormalities in CLL cells by increasing p66Shc expression. These results, identifying p66Shc as a regulator of CXCR4/CCR7 recycling in B cells, underscore the relevance of its deficiency to CLL pathogenesis and provide new clues to the mechanisms underlying the therapeutic effects of ibrutinib.


Assuntos
Endossomos/metabolismo , Leucemia Linfocítica Crônica de Células B , Receptores CCR7/metabolismo , Receptores CXCR4/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/genética , beta-Arrestinas/metabolismo , Adenina/análogos & derivados , Adulto , Animais , Estudos de Casos e Controles , Células Cultivadas , Endossomos/efeitos dos fármacos , Endossomos/patologia , Mutação em Linhagem Germinativa , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/metabolismo , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos Knockout , Fosforilação/genética , Piperidinas , Proteólise , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico
9.
J Immunol ; 193(11): 5584-94, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25339679

RESUMO

BAFF is a crucial cytokine that affects the activity of both innate and adaptive immune cells. It promotes the expansion of Th17 cells in autoimmune disorders. With this study, we investigated the BAFF/Th17 responses in Helicobacter pylori-induced gastritis in humans. Our results show that the mucosa from Helicobacter(+) patients with chronic gastritis is enriched in IL-17 and BAFF, whereas the two cytokines are weakly expressed in Helicobacter(-) patients with chronic gastritis; moreover, the expression of both BAFF and IL-17 decreases after bacteria eradication. We demonstrate that BAFF accumulates in macrophages in vivo and that it is produced by monocyte-derived macrophages in vitro, after Helicobacter stimulation. Application of BAFF on monocytes triggers the accumulation of reactive oxygen species that are crucial for the release of pro-Th17 cytokines, such as IL-23, IL-1ß, and TGF-ß. Moreover, BAFF directly promotes the differentiation of Th17 cells. In conclusion, our results support the notion that an axis BAFF/Th17 exists in chronic gastritis of Helicobacter(+) patients and that its presence strictly depends on the bacterium. Moreover, we demonstrated that BAFF is able to drive Th17 responses both indirectly, by creating a pro-Th17 cytokine milieu through the involvement of innate immune cells, and directly, via the differentiation of T cells toward the specific profile. The results obtained in this study are of great interest for Helicobacter-related diseases and the development of novel therapeutic strategies based on the inhibition of the BAFF/IL-17 response.


Assuntos
Fator Ativador de Células B/metabolismo , Gastrite/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Macrófagos/imunologia , Mucosa/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Diferenciação Celular , Células Cultivadas , Doença Crônica , Gastrite/etiologia , Infecções por Helicobacter/complicações , Humanos , Imunidade Inata , Interleucina-17/metabolismo , Macrófagos/microbiologia , Mucosa/microbiologia , Espécies Reativas de Oxigênio/metabolismo
10.
Recent Pat Anticancer Drug Discov ; 8(2): 126-42, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22894642

RESUMO

The response of the body to cancer is not a unique mechanism and has many parallels with inflammation and wound healing. Unresolved inflammation generates a microenvironment favorable for cellular transformation and the growth of cancer cells. Chronic tissue damage triggers a repair response that includes the production of growth factors, cytokines and chemokines. Cytokines and chemokines have a crucial role in cancer-related inflammation with consequent, direct and indirect effects on the proliferative and invasive properties of tumor cells. In view of the multifactorial functions of cytokines and chemokines in tumorigenesis, the elucidation of their roles will further advance our understanding of the patho-physiological processes of tumor development and highlights potential innovative anti-cancer strategies. Despite recent advances, main anti-cancer therapies, namely surgery, radiation therapy and chemotherapy, are limited in their ability to treat minimal and metastatic residual disease. Furthermore, the benefit of conventional therapies is often limited by collateral damage to normal tissues. Immunotherapy is a new avenue of cancer treatment being investigated by researchers and clinicians for different cancer types. The aim of this paper is to analyze the recent patents and scientific reviews on the major cytokine/chemokine pathways involved in cancer immunotherapy and discuss their basic biology, clinical relevance and potential directions for future anti-cancer therapeutic applications.


Assuntos
Quimiocinas/uso terapêutico , Citocinas/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Animais , Quimiocinas/metabolismo , Quimiocinas/fisiologia , Citocinas/metabolismo , Citocinas/fisiologia , Humanos , Imunoterapia/tendências , Neoplasias/imunologia , Patentes como Assunto
11.
Expert Rev Gastroenterol Hepatol ; 6(4): 437-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22928895

RESUMO

Helicobacter pylori represents the major cause of gastric cancer, gastric lymphoma and peptic ulcer diseases. In some cases, the infection persists even after three rounds of treatment. The evaluated article reports on the efficacy of an empirical multicenter, prospective fourth­line rescue study with rifabutin in patients with three consecutive eradication failures. A total of 100 patients (31% peptic ulcer and 69% functional dyspepsia) were included to receive a fourth­line with rifabutin (150 mg twice daily [b.i.d.]), amoxicillin (1 g b.i.d.) and a proton­pump inhibitor (standard dose b.i.d.) for 10 days. The end point was H. pylori eradication, determined by (13)C-urea breath test 4-8 weeks after therapy. H. pylori eradication was achieved in approximately 50% of patients. Adverse events (mainly metallic taste, nausea and diarrhea) were reported in 30 patients. Thus, a fourth-line rifabutin-based rescue therapy constitutes a valid strategy after multiple previous H. pylori eradication failures with key antibiotics, such as clarithromycin, metronidazole, tetracycline and levofloxacin.

12.
Clin Dev Immunol ; 2012: 690571, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22693525

RESUMO

Gastric cancer is the second cause of cancer-related deaths worldwide. Helicobacter pylori is the major risk factor for gastric cancer. As for any type of cancer, T cells are crucial for recognition and elimination of gastric tumor cells. Unfortunately T cells, instead of protecting from the onset of cancer, can contribute to oncogenesis. Herein we review the different types, "friend or foe", of T-cell response in gastric cancer.


Assuntos
Neoplasias Gástricas/imunologia , Linfócitos T/imunologia , Animais , Humanos , Neoplasias Gástricas/metabolismo , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
13.
Clin Dev Immunol ; 2011: 320571, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110523

RESUMO

Gastrointestinal oncology is one of the foremost causes of death: the gastric cancer accounts for 10.4% of cancer deaths worldwide, the pancreatic cancer for 6%, and finally, the colorectal cancer for 9% of all cancer-related deaths. For all these gastrointestinal cancers, surgical tumor resection remains the primary curative treatment, but the overall 5-year survival rate remains poor, ranging between 20-25%; the addition of combined modality strategies (pre- or postoperative chemoradiotherapy or perioperative chemotherapy) results in 5-year survival rates of only 30-35%. Therefore, many investigators believe that the potential for making significant progress lies on understanding and exploiting the molecular biology of gastrointestinal tumors to investigate new therapeutic strategies such as specific immunotherapy. In this paper we will focus on recent knowledge concerning the role of T cells and the use of T adoptive immunotherapy in the treatment of gastrointestinal cancers.


Assuntos
Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/terapia , Imunoterapia Adotiva , Oncologia , Linfócitos T/imunologia , Animais , Terapia Combinada , Neoplasias Gastrointestinais/patologia , Humanos , Oncologia/tendências , Linfócitos T/transplante
14.
J Exp Med ; 208(6): 1317-30, 2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21576384

RESUMO

The Bordetella pertussis adenylate cyclase toxin (CyaA) assists infection by potently suppressing the host immune response. Although CyaA effectively targets T lymphocytes, its putative receptor on these cells is unknown. Here, we show that CyaA binds to T cells via the ß2 integrin LFA-1 in its active conformation. CyaA clusters with LFA-1 at the immune synapse (IS), from which it induces the premature disengagement of LFA-1 concomitant with the dissipation of talin, which tethers the integrin to the underlying actin cytoskeleton. The CyaA-induced redistribution of LFA-1 was cAMP- and protein kinase A (PKA)-dependent. These results not only identify LFA-1 as a CyaA receptor on T cells but unveil a novel mechanism of immunosuppression whereby the toxin parasitizes its interaction with LFA-1 to inhibit signaling at the IS through the local production of cAMP. The data also provide novel insights into the role of cAMP/PKA signaling in controlling the dynamics of the IS.


Assuntos
Toxina Adenilato Ciclase/metabolismo , Bordetella pertussis/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Linfócitos T/microbiologia , Linfócitos B/citologia , Antígeno CD11a/biossíntese , Complexo CD3/imunologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Células Dendríticas/citologia , Citometria de Fluxo/métodos , Humanos , Sistema Imunitário , Células Jurkat , Microscopia de Fluorescência/métodos , Linfócitos T/imunologia
15.
Blood ; 117(24): 6612-6, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21527528

RESUMO

Lymphoid hyperplasia of gastric mucosa associated with Helicobacter pylori (HP) infection represents a preneoplastic condition of the mucosa associated lymphoid tissue (MALT), which may evolve to a B-cell lymphoma. While it is well established that the initial neoplastic proliferation of B cells is antigen-driven and dependent on the helper activity of HP-specific T cells, it needs to be elucidated which cytokine or soluble factor(s) promote B-cell activation and lymphomagenesis. Herein, we originally report that gastric MALT lymphoma express high levels of a proliferation inducing ligand (APRIL), a novel cytokine crucial in sustaining B-cell proliferation. By immunohistochemistry, we demonstrate that APRIL is produced almost exclusively by gastric lymphoma-infiltrating macrophages located in close proximity to neoplastic B cells. We also show that macrophages produce APRIL on direct stimulation with both HP and HP-specific T cells. Collectively, our results represent the first evidence for an involvement of APRIL in gastric MALT lymphoma development in HP-infected patients.


Assuntos
Mucosa Gástrica/metabolismo , Linfoma de Zona Marginal Tipo Células B/metabolismo , Macrófagos/metabolismo , Neoplasias Gástricas/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Doença Crônica , Mucosa Gástrica/imunologia , Mucosa Gástrica/patologia , Gastrite/imunologia , Gastrite/metabolismo , Gastrite/patologia , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/complicações , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori/fisiologia , Humanos , Linfoma de Zona Marginal Tipo Células B/etiologia , Linfoma de Zona Marginal Tipo Células B/patologia , Macrófagos/patologia , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Lesões Pré-Cancerosas/imunologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/patologia
17.
J Biomed Biotechnol ; 2011: 437348, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22253528

RESUMO

Gastric cancer (GC) is the fourth most common cancer and the second most frequent cause of cancer-related deaths, accounting for 10.4% of cancer deaths worldwide. Despite the improvements, estimated cure rates for patients with advanced stages remain poor, and in the metastatic setting, chemotherapy is the mainstay of palliative therapy and results in objective response rates (ORRs) of only 20-40% and median overall survivals (OS) of 8-10 months. Therefore, many investigators believe that the potential for making significant progress lies in understanding and exploiting the molecular biology of these tumors to investigate new therapeutic strategies to combat GC, such as specific immunotherapy. In this paper, we analyze the different approaches used for immune-based (especially dendritic and T cells) therapies to gastric cancer treatment and discuss the results obtained in preclinical models as in clinical trials.


Assuntos
Adenocarcinoma/terapia , Anticorpos Monoclonais/imunologia , Carcinoma de Células Escamosas/terapia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Gástricas/terapia , Adenocarcinoma/imunologia , Anticorpos Monoclonais/uso terapêutico , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Carcinoma de Células Escamosas/imunologia , Células Dendríticas/metabolismo , Humanos , Imunoterapia , Células Matadoras Naturais/metabolismo , Terapia de Alvo Molecular/métodos , Neoplasias Gástricas/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Expert Rev Anti Infect Ther ; 8(8): 887-92, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20695744

RESUMO

Helicobacter pylori is the cause of peptic ulcer, gastric cancer and gastric lymphoma. Diagnosis of H. pylori infection can be made using invasive and noninvasive tests. Invasive tests based on endoscopy, such as histology, are recommended when a gastric malignancy is suspected. Alternatively, noninvasive tests, such as the urea breath test and stool tests are useful for H. pylori diagnosis and follow-up. Triple therapy with either amoxicillin or metronidazole, clarithromycin and proton pump inhibitor given twice daily for 7-14 days is the recommended first-line treatment, after having checked the individual clarithromycin antimicrobial susceptibility. A triple therapy with levofloxacin, amoxicillin and proton pump inhibitor for 10-14 days should be used as second-line treatment, where the strains are susceptible to fluoroquinolone. Alternatively, bismuth-based quadruple therapy is recommended.


Assuntos
Antibacterianos/uso terapêutico , Antiulcerosos/uso terapêutico , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori/efeitos dos fármacos , Quimioterapia Combinada , Diretrizes para o Planejamento em Saúde , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Humanos , Úlcera Péptica/tratamento farmacológico , Úlcera Péptica/etiologia , Úlcera Péptica/microbiologia , Inibidores da Bomba de Prótons/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/microbiologia
20.
J Biomed Biotechnol ; 2010: 104918, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20617132

RESUMO

Helicobacter pylori infection is the major cause of gastroduodenal pathologies, but only a minority of infected patients develop gastric B-cell lymphoma, gastric autoimmunity, or other life threatening diseases, as gastric cancer or peptic ulcer. The type of host immune response against H. pylori, particularly the cytolytic effector functions of T cells, is crucial for the outcome of the infection. T cells are potentially able to kill a target via different mechanisms, such as perforins or Fas-Fas ligand interaction. In H. pylori-infected patients with gastric autoimmunity cytolytic T cells, that cross-recognize different epitopes of H. pylori proteins and H(+)K(+)-ATPase autoantigen, infiltrate the gastric mucosa and lead to gastric atrophy via long-lasting activation of Fas ligand-mediated appotosis and perforin-induced cytotoxicity. On the other hand, gastric T cells from MALT lymphoma exhibit defective perforin- and Fas-Fas ligand-mediated killing of B cells, with consequent abnormal help for B-cell proliferation, suggesting that deregulated and exhaustive H. pylori-induced T cell-dependent B-cell activation can support both the onset and the promotion of low-grade B-cell lymphoma.


Assuntos
Doenças Autoimunes , Helicobacter pylori , Linfoma de Zona Marginal Tipo Células B , Neoplasias Gástricas , Linfócitos T Citotóxicos , Animais , Modelos Animais de Doenças , Humanos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA