Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Cell Commun Signal ; 22(1): 129, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38360687

RESUMO

BACKGROUND: Extracellular vesicles (EVs), including microvesicles, hold promise for the management of bladder urothelial carcinoma (BLCA), particularly because of their utility in identifying therapeutic targets and their diagnostic potential using easily accessible urine samples. Among the transmembrane glycoproteins highly enriched in cancer-derived EVs, tissue factor (TF) and CD147 have been implicated in promoting tumor progression. In this in vitro study, we explored a novel approach to impede cancer cell migration and metastasis by simultaneously targeting these molecules on urothelial cancer-derived EVs. METHODS: Cell culture supernatants from invasive and non-invasive bladder cancer cell lines and urine samples from patients with BLCA were collected. Large, microvesicle-like EVs were isolated using sequential centrifugation and characterized by electron microscopy, nanoparticle tracking analysis, and flow cytometry. The impact of urinary or cell supernatant-derived EVs on cellular phenotypes was evaluated using cell-based assays following combined treatment with a specific CD147 inhibitor alone or in combination with a tissue factor pathway inhibitor (TFPI), an endogenous anticoagulant protein that can be released by low-molecular-weight heparins. RESULTS: We observed that EVs obtained from the urine samples of patients with muscle-invasive BLCA and from the aggressive bladder cancer cell line J82 exhibited higher TF activity and CD147 expression levels than did their non-invasive counterparts. The shedding of GFP-tagged CD147 into isolated vesicles demonstrated that the vesicles originated from plasma cell membranes. EVs originating from invasive cancer cells were found to trigger migration, secretion of matrix metalloproteinases (MMPs), and invasion. The same induction of MMP activity was replicated using EVs obtained from urine samples of patients with invasive BLCA. EVs derived from cancer cell clones overexpressing TF and CD147 were produced in higher quantities and exhibited a higher invasive potential than those from control cancer cells. TFPI interfered with the effect when used in conjunction with the CD147 inhibitor, further suppressing homotypic EV-induced migration, MMP production, and invasion. CONCLUSIONS: Our findings suggest that combining a CD147 inhibitor with low molecular weight heparins to induce TFPI release may be a promising therapeutic approach for urothelial cancer management. This combination can potentially suppress the tumor-promoting actions of cancer-derived microvesicle-like EVs, including collective matrix invasion.


Small particles or vesicles released by cancer cells into their surroundings have the potential to stimulate the spread and growth of cancer cells. In this study, we focused on two specific molecules presented by these cancer cell-derived vesicles that could play a role in promoting the dissemination of cancer cells: a protein related to blood clotting and a protein on the cell surface.We found that large vesicles from bladder cancer cells that have the ability to spread had higher levels of these proteins than vesicles from nonspreading cancer cells. We also found that the former could make cancer cells move about more, produce more of a substance that helps cancer cells spread, and invade other tissues.To counteract the cancer-promoting actions of these vesicles, we examined the impact of combining a naturally occurring anticlotting protein that can be released by medications derived from heparin with an inhibitor targeting the cancer cell surface protein. We found that this combination stopped the vesicles from helping cancer cells move about more, produce more of the spreading substance, and invade other tissues.This approach of simultaneously targeting the two protein molecules present on cancer cell-derived vesicles might be a new way to treat bladder cancer.


Assuntos
Basigina , Carcinoma de Células de Transição , Vesículas Extracelulares , Lipoproteínas , Neoplasias da Bexiga Urinária , Humanos , Carcinoma de Células de Transição/tratamento farmacológico , Linhagem Celular Tumoral , Vesículas Extracelulares/efeitos dos fármacos , Lipoproteínas/metabolismo , Neoplasias da Bexiga Urinária/tratamento farmacológico , Basigina/antagonistas & inibidores
2.
Pediatr Blood Cancer ; : e30435, 2023 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-37246348

RESUMO

We analyzed changes in laboratory parameters, including blood counts, liver enzymes, inflammation and coagulation markers, and cytokines, from 70 blinatumomab-treated pediatric patients (NCT01471782). Overall, trends were consistent in responders and nonresponders. Platelets and lymphocytes peaked on day (D) 10 in cycle 1 and returned to baseline on D42 and D29, respectively. Neutrophils peaked on D2 and returned to baseline on D42. Alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and bilirubin peaked on D17, reversing to baseline on D29; total protein levels were constant. These findings indicate that blinatumomab-induced changes in laboratory parameters were transient, reversible, and not requiring treatment interruptions in responders and nonresponders.

3.
Pediatr Blood Cancer ; 68(12): e29323, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34519430

RESUMO

Blinatumomab is a BiTE® (bispecific T-cell engager) immuno-oncology therapy, which has demonstrated significant activity in patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia (R/R B-ALL); however, a subset of patients relapse. Monitoring expression of cluster of differentiation (CD)19 in relapsed patients is critical to inform sequencing of subsequent therapies. The expression of CD19 in 59 pediatric patients with R/R B-ALL was analyzed on the day of diagnosis of R/R B-ALL and on days 15 and 29 of cycle 1 of blinatumomab. Most patients treated with one cycle of blinatumomab retained expression of CD19, and would therefore be eligible for subsequent anti-CD19 CAR T-cell therapy.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Doença Aguda , Antígenos CD19/metabolismo , Antineoplásicos/efeitos adversos , Criança , Humanos , Linfoma de Células B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico
4.
J Fish Dis ; 44(12): 2031-2041, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34424537

RESUMO

Salmon pancreas disease virus (SPDV) has been affecting the salmon farming industry for over 30 years, but despite the substantial amount of studies, there are still a number of recognized knowledge gaps, for example in the transmission of the virus. In this work, an ultrastructural morphological approach was used to describe observations after infection by SPDV of an ex vivo cardiac model generated from Atlantic salmon embryos. The observations in this study and those available on previous ultrastructural work on SPDV are compared and contrasted with the current knowledge on terrestrial mammalian and insect alphaviral replication cycles, which is deeper than that of SPDV both morphologically and mechanistically. Despite their limitations, morphological descriptions remain an excellent way to generate novel hypotheses, and this has been the aim of this work. This study has used a target host, ex vivo model and resulted in some previously undescribed features, including filopodial membrane projections, cytoplasmic stress granules or putative intracytoplasmic budding. The latter suggests a new hypothesis that warrants further mechanistic research: SPDV in salmon may have retained the capacity for non-cytolytic (persistent) infections by intracellular budding, similar to that noted in arthropod vectors of other alphaviruses. In the notable absence of a known intermediate host for SPDV, the presence of this pattern suggests that both cytopathic and persistent infections may coexist in the same host. It is our hope that the ultrastructural comparison presented here stimulates new research that brings the knowledge on SPDV replication cycle up to a similar level to that of terrestrial alphaviruses.


Assuntos
Infecções por Alphavirus/veterinária , Alphavirus/fisiologia , Replicação Viral/fisiologia , Alphavirus/ultraestrutura , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/virologia , Animais , Doenças dos Peixes/virologia , Interações Hospedeiro-Patógeno , Microscopia Eletrônica , Salmo salar , Técnicas de Cultura de Tecidos
5.
Immunol Rev ; 270(1): 193-208, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26864113

RESUMO

Bispecific T-cell engager (BiTE(®)) antibody constructs represent a novel immunotherapy that bridges cytotoxic T cells to tumor cells, thereby inducing target cell-dependent polyclonal T-cell activation and proliferation, and leading to apoptosis of bound tumor cells. Anti-CD19 BiTE(®) blinatumomab has demonstrated clinical activity in Philadelphia chromosome (Ph)-negative relapsed or refractory (r/r) acute lymphoblastic leukemia (ALL) eventually resulting in conditional approval by the U.S. Food and Drug Administration in 2014. This drug is currently further developed in pediatric and Ph(+) r/r, as well as in minimal residual disease-positive ALL, and might also offer clinical benefit for patients with non-Hodgkin's lymphoma, especially for those with aggressive forms like diffuse large B-cell lymphoma. Another BiTE(®) antibody construct in hemato-oncology designated AMG 330 targets CD33 on acute myeloid leukemia blast cells. After showing promising ex vivo activity, this drug candidate has recently entered phase 1 clinical development, and has further indicated potential for combination with checkpoint inhibitors. In solid tumor indications, three BiTE(®) antibody constructs have been tested in phase 1 studies so far: anti-EpCAM BiTE(®) AMG 110, anti-CEA BiTE(®) MEDI-565/AMG 211, and anti-PSMA BiTE(®) BAY2010112/AMG 212. Pertinent questions comprise how to maximize BiTE(®) penetration and T-cell infiltration of the tumor while simultaneously minimizing any adverse events, which is currently explored by a continuous intravenous infusion approach. Thus, BiTE(®) antibody constructs will hopefully provide new treatment options for patients in several indications with high unmet medical need.


Assuntos
Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/uso terapêutico , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Subpopulações de Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/farmacologia , Antígenos de Neoplasias/imunologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Humanos , Ativação Linfocitária/imunologia , Neoplasias/diagnóstico , Neoplasias/mortalidade , Engenharia de Proteínas , Subpopulações de Linfócitos T/metabolismo , Resultado do Tratamento
6.
J Oral Pathol Med ; 48(10): 943-950, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31400171

RESUMO

BACKGROUND: Osteoblast adhesion is a crucial step in osseointegration of dental implants and can be influenced by modification of implant surface or the addition of bioactive agents. Bisphosphonates affect bone turnover, attenuating bone healing in implants patients. PRP and PRF are sources of growth factors involved in osteoblast adhesion, improving subsequent bone healing. The aim of the study was to investigate the impacts of PRP and PRF on adhesion of bisphosphonate-pretreated osteoblasts on titanium implant surfaces using the cell-count wash assay, the MTT-assay as well as real-time-cell analyser assay and scanning electronic microscopy. METHODS: Titanium implants were colonised for 24 hours with osteoblasts and zolendronic acid, PRP or PRF in different combinations. Afterwards, primary osteoblast adhesion was evaluated by counting the number of attached cells using a wash-assay cell analysis. Scanning electronic microscopy was performed and evaluated semi-quantitatively to assess the influence of the different groups on the ultrastructural cell morphology, such as cell size and shape as well as length and number of filopodia. RESULTS: Zoledronic acid led to a decrease of osteoblast adherence onto implant surface. This effect was reversed by adding PRP or PRF. Scanning electronic microscopy showed that both PRP and PRF increased number and length of filopodia in adherent osteoblasts. CONCLUSIONS: Zoledronic acid decreased osteoblast adhesion on implant surfaces, and PRF as well as PRP increased primary adhesion of zoledronic acid-treated osteoblasts on implant surfaces in vitro. Therefore, PRP and PRF may improve initial bone apposition and primary healing of dental implants in patients with bisphosphonate treatment.


Assuntos
Adesão Celular , Implantes Dentários , Difosfonatos/farmacologia , Osteoblastos/citologia , Fibrina Rica em Plaquetas , Plasma Rico em Plaquetas , Titânio , Células Cultivadas , Humanos , Ácido Zoledrônico
7.
Cytokine ; 112: 95-101, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29885991

RESUMO

Interferon-γ (IFN-γ) is a central mediator of host immune responses including T-cell differentiation and activation of macrophages for the control of bacterial pathogens. Anti-bacterial mechanisms of IFN-γ against the obligate intracellular bacteria Chlamydiatrachomatis in epithelial cells have been intensively investigated in the past, focusing on cellular tryptophan depletion by an IFN-γ induced expression of the indoleamine 2, 3-deoxygenase (IDO). In this study, we could show that IFN-γ treatment caused a significant reduction of the host cell glycolysis that was accompanied by a reduction of glucose transporter-1 (GLUT1) and hypoxia inducible factor-1α (HIF-1α) expression. Furthermore, C. trachomatis induced enhancement of glycolytic and mitochondrial activation were significantly suppressed by IFN-γ treatment. We could further show that glucose starvation, as observed under IFN-γ treatment, was associated with an attenuated antimicrobial efficacy of doxycycline (DOX) against C. trachomatis. In conclusions, anti-chlamydial activity of IFN-γ goes beyond tryptophan depletion including interference with cellular energy metabolism resulting reduced progeny, but also impaired antimicrobial susceptibility of C. trachomatis.


Assuntos
Infecções por Chlamydia/metabolismo , Interferon gama/metabolismo , Anti-Infecciosos/farmacologia , Linhagem Celular Tumoral , Infecções por Chlamydia/tratamento farmacológico , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/efeitos dos fármacos , Doxiciclina/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Glucose/metabolismo , Glicólise/fisiologia , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Triptofano/metabolismo
8.
Fish Shellfish Immunol ; 72: 181-186, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29102629

RESUMO

In vitro fish based models have been extensively applied in human biomedical research but, paradoxically, less frequently in the research of fish health issues. Farmed Atlantic salmon can suffer from several viral conditions affecting the heart. Therefore, species-specific, cardiac in vitro models may represent a useful tool to help further understanding and management of these diseases. The mechanisms underlying genotype based resistance are complex and usually rely on a combined effect of elements from both the innate and adaptive immune response, which are further complicated by external environmental factors. Here we propose that Salmon Cardiac Primary Cultures (SCPCs) are a useful tool to investigate these mechanisms as the basis for genotypic differences between Atlantic salmon families in susceptibility to cardiotropic viral disease. Using SCPCs produced from two different commercially available Atlantic salmon embryonated ova (Atlantic Ova IPN sensitive" (S) and "Atlantic QTL-innOva® IPN/PD" (R)), the influence of host genotype on the viral load and mx expression following Salmon Pancreas Disease Virus infection was assessed over a 15 day period. Both R and S SCPCs groups were successfully infected. A measurable difference between groups of viral nsP1 and host antiviral mx gene expression was observed (i.e. a later, but larger onset of mx expression in the R group). Mx expression peaks were followed by a decrease in viral nsP1 in both groups. Additionally, ultrastructural examination of infected SCPCs allowed the description of degenerative changes at the individual cell level. The SCPC model presents some advantages, over current fish cell culture monolayers and in vivo material, such as the presence of different cell components normally present in the target organ, as well as the removal of a layer of functional complexity (acquired immunity), making it possible to focus on tissue specific, early innate immune mechanisms. These preliminary results highlight the importance of considering genetic origin when selecting the fish source for the production of SCPCs, as well as their usefulness as screening tools for assessment of genotypic differences in disease resistance.


Assuntos
Infecções por Alphavirus/veterinária , Alphavirus/fisiologia , Salmo salar/imunologia , Carga Viral , Proteínas Virais/genética , Alphavirus/genética , Alphavirus/ultraestrutura , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Células Cultivadas , Doenças dos Peixes/patologia , Doenças dos Peixes/virologia , Genótipo , Cinética , Óvulo/virologia , Salmo salar/genética
9.
Blood ; 126(24): 2578-84, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26480933

RESUMO

This long-term follow-up analysis evaluated overall survival (OS) and relapse-free survival (RFS) in a phase 2 study of the bispecific T-cell engager antibody construct blinatumomab in 36 adults with relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL). In the primary analysis, 25 (69%) patients with relapsed/refractory ALL achieved complete remission with full (CR) or partial (CRh) hematologic recovery of peripheral blood counts within the first 2 cycles. Twenty-five patients (69%) had a minimal residual disease (MRD) response (<10(-4) blasts), including 22 CR/CRh responders, 2 patients with hypocellular bone marrow, and 1 patient with normocellular bone marrow but low peripheral counts. Ten of the 36 patients (28%) were long-term survivors (OS ≥30 months). Median OS was 13.0 months (median follow-up, 32.6 months). MRD response was associated with significantly longer OS (Mantel-Byar P = .009). All 10 long-term survivors had an MRD response. Median RFS was 8.8 months (median follow-up, 28.9 months). A plateau for RFS was reached after ∼18 months. Six of the 10 long-term survivors remained relapse-free, including 4 who received allogeneic stem cell transplantation (allo-SCT) as consolidation for blinatumomab and 2 who received 3 additional cycles of blinatumomab instead of allo-SCT. Three long-term survivors had neurologic events or cytokine release syndrome, resulting in temporary blinatumomab discontinuation; all restarted blinatumomab successfully. Long-term survivors had more pronounced T-cell expansion than patients with OS <30 months.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos/uso terapêutico , Imunossupressores/uso terapêutico , Terapia de Alvo Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Terapia de Salvação , Subpopulações de Linfócitos T/patologia , Adolescente , Adulto , Idoso , Aloenxertos , Anticorpos Biespecíficos/efeitos adversos , Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Medula Óssea/patologia , Ensaios Clínicos Fase II como Assunto/estatística & dados numéricos , Citocinas/metabolismo , Intervalo Livre de Doença , Feminino , Seguimentos , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunossupressores/efeitos adversos , Estimativa de Kaplan-Meier , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto/estatística & dados numéricos , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Recidiva , Indução de Remissão , Subpopulações de Linfócitos T/efeitos dos fármacos , Adulto Jovem
10.
Exp Dermatol ; 26(12): 1199-1206, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28940860

RESUMO

Previous reports have demonstrated that cell-derived nanoparticles (CDNPs) composed of bovine or porcine protein complexes exerted therapeutic effects against viral infections and cancer in mice and humans. Based on these observations, we asked whether CDNPs would improve inflammatory skin disorders. To address this, we utilized two distinct mouse models of cutaneous inflammation: the autoimmune skin-blistering disease epidermolysis bullosa acquisita (EBA) as an example of an autoantibody-induced cutaneous inflammation, and Leishmania major (L. major) infection as an example of a pathogen-induced cutaneous inflammation. In both models, we observed that CDNPs increased mRNA expression of the Th2 cytokine IL-4. Clinically, CDNPs decreased inflammation due to EBA and increased L. major-specific IgG1 levels without major effects on infected skin lesions. In addition, CDNPs supported the growth of keratinocytes in human skin cultures. In vitro studies revealed that CDNPs were taken up predominantly by macrophages, leading to a shift towards the expression of anti-inflammatory cytokine genes. Altogether, our data demonstrate that treatment with porcine CDNPs may be a new therapeutic option for the control of autoimmune-mediated inflammatory skin disorders.


Assuntos
Micropartículas Derivadas de Células/transplante , Epidermólise Bolhosa Adquirida/terapia , Leishmaniose Cutânea/terapia , Reepitelização , Células Th2/fisiologia , Adulto , Animais , Diferenciação Celular , Epidermólise Bolhosa Adquirida/imunologia , Feminino , Humanos , Interleucina-4/metabolismo , Leishmania major , Leishmaniose Cutânea/imunologia , Linfonodos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Suínos , Adulto Jovem
11.
Basic Res Cardiol ; 111(1): 6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26659360

RESUMO

Cardiomyopathy is one of the most common causes of chronic heart failure worldwide. Mutations in the gene encoding nexilin (NEXN) occur in patients with both hypertrophic and dilated cardiomyopathy (DCM); however, little is known about the pathophysiological mechanisms and relevance of NEXN to these disorders. Here, we evaluated the functional role of NEXN using a constitutive Nexn knock-out (KO) mouse model. Heterozygous (Het) mice were inter-crossed to produce wild-type (WT), Het, and homozygous KO mice. At birth, 32, 46, and 22 % of the mice were WT, Het, and KO, respectively, which is close to the expected Mendelian ratio. After postnatal day 6, the survival of the Nexn KO mice decreased dramatically and all of the animals died by day 8. Phenotypic characterizations of the WT and KO mice were performed at postnatal days 1, 2, 4, and 6. At birth, the relative heart weights of the WT and KO mice were similar; however, at day 4, the relative heart weight of the KO group was 2.3-fold higher than of the WT group. In addition, the KO mice developed rapidly progressive cardiomyopathy with left ventricular dilation and wall thinning and decreased cardiac function. At day 6, the KO mice developed a fulminant DCM phenotype characterized by dilated ventricular chambers and systolic dysfunction. At this stage, collagen deposits and some elastin deposits were observed within the left ventricle cavity, which resembles the features of endomyocardial fibroelastosis (EFE). Overall, these results further emphasize the role of NEXN in DCM and suggest a novel role in EFE.


Assuntos
Cardiomiopatias/metabolismo , Fibroelastose Endocárdica/metabolismo , Proteínas dos Microfilamentos/deficiência , Animais , Western Blotting , Cardiomiopatias/patologia , Modelos Animais de Doenças , Ecocardiografia , Fibroelastose Endocárdica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase
12.
Cell Microbiol ; 17(7): 923-34, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25588955

RESUMO

Direct interaction of Chlamydiae with the endoplasmic reticulum (ER) is essential in intracellular productive infection. However, little is known about the interplay between Chlamydiae and the ER under cellular stress conditions that are observed in interferon gamma (IFN-γ) induced chlamydial persistent infection. ER stress responses are centrally regulated by the unfolded protein response (UPR) under the control of the ER chaperone BiP/GRP78 to maintain cellular homeostasis. In this study, we could show that the ER directly contacted with productive and IFN-γ-induced persistent inclusions of Chlamydia pneumoniae (Cpn). BiP/GRP78 induction was observed in the early phase but not in the late phase of IFN-γ-induced persistent infection. Enhanced BiP/GRP78 expression in the early phase of IFN-γ-induced persistent Cpn infection was accompanied by phosphorylation of the eukaryotic initiation factor-2α (eIF2α) and down-regulation of the vesicle-associated membrane protein-associated protein B. Loss of BiP/GRP78 function resulted in enhanced phosphorylation of eIF2α and increased host cell apoptosis. In contrast, enhanced BiP/GRP78 expression in IFN-γ-induced persistent Cpn infection attenuated phosphorylation of eIF2α upon an exogenous ER stress inducer. In conclusion, ER-related BiP/GRP78 plays a key role to restore cells from stress conditions that are observed in the early phase of IFN-γ-induced persistent infection.


Assuntos
Infecções por Chlamydophila/imunologia , Chlamydophila pneumoniae/imunologia , Proteínas de Choque Térmico/metabolismo , Interações Hospedeiro-Patógeno , Interferon gama/metabolismo , Chaperona BiP do Retículo Endoplasmático , Células Hep G2 , Hepatócitos/imunologia , Hepatócitos/microbiologia , Humanos
13.
J Immunol ; 193(4): 1954-65, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25024378

RESUMO

Canonical neutrophil antimicrobial effector mechanisms, such as degranulation, production of reactive oxygen species, and release of neutrophil extracellular traps (NETs), can result in severe pathology. Activation of neutrophils through immune complexes (ICs) plays a central role in the pathogenesis of many autoimmune inflammatory diseases. In this study, we report that immobilized ICs (iICs), which are hallmarks of several autoimmune diseases, induce the release of NETs from primary human neutrophils. The iIC-induced NET formation was found to require production of reactive oxygen species by NADPH oxidase and myeloperoxidase and to be mediated by FcγRIIIb. Blocking of the ß2 integrin macrophage-1 Ag but not lymphocyte function-associated Ag-1 abolished iIC-induced NET formation. This suggests that FcγRIIIb signals in association with macrophage-1 Ag. As intracellular signaling pathways involved in iIC-induced NET formation we identified the tyrosine kinase Src/Syk pathway, which downstream regulates the PI3K/Akt, p38 MAPK, and ERK1/2 pathways. To our knowledge, the present study shows for the first time that iICs induce NET formation. Thus, we conclude that NETs contribute to pathology in autoimmune inflammatory disorders associated with surface-bound ICs.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Antígeno de Macrófago 1/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Receptores de IgG/imunologia , Aminopirina/farmacologia , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Doenças Autoimunes/imunologia , Butadienos/farmacologia , Antígeno CD11a/metabolismo , Antígenos CD18/metabolismo , Degranulação Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Proteínas Ligadas por GPI/imunologia , Humanos , Imidazóis/farmacologia , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno de Macrófago 1/metabolismo , Mesalamina/farmacologia , Nitrilas/farmacologia , Oniocompostos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Piridinas/farmacologia , Pirimidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de IgG/antagonistas & inibidores , Quinase Syk , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/imunologia
14.
Blood ; 119(26): 6226-33, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22592608

RESUMO

T cell-engaging CD19/CD3-bispecific BiTE Ab blinatumomab has shown an 80% complete molecular response rate and prolonged leukemia-free survival in patients with minimal residual B-lineage acute lymphoblastic leukemia (MRD(+) B-ALL). Here, we report that lymphocytes in all patients of a phase 2 study responded to continuous infusion of blinatumomab in a strikingly similar fashion. After start of infusion, B-cell counts dropped to < 1 B cell/µL within an average of 2 days and remained essentially undetectable for the entire treatment period. By contrast, T-cell counts in all patients declined to a nadir within < 1 day and recovered to baseline within a few days. T cells then expanded and on average more than doubled over baseline within 2-3 weeks under continued infusion of blinatumomab. A significant percentage of reappearing CD8(+) and CD4(+) T cells newly expressed activation marker CD69. Shortly after start of infusion, a transient release of cytokines dominated by IL-10, IL-6, and IFN-γ was observed, which no longer occurred on start of a second treatment cycle. The response of lymphocytes in leukemic patients to continuous infusion of blinatumomab helps to better understand the mode of action of this and other globally T cell-engaging Abs. The trial is registered with www.clinicaltrials.gov identifier NCT00560794.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Antígenos CD19/imunologia , Complexo CD3/imunologia , Sistema Imunitário/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Adulto , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/farmacocinética , Soro Antilinfocitário/administração & dosagem , Soro Antilinfocitário/efeitos adversos , Soro Antilinfocitário/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/farmacocinética , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Bombas de Infusão , Ativação Linfocitária/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Resultado do Tratamento
15.
Blood ; 120(26): 5185-7, 2012 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-23024237

RESUMO

Persistence or recurrence of minimal residual disease (MRD) after chemotherapy results in clinical relapse in patients with acute lymphoblastic leukemia (ALL). In a phase 2 trial of B-lineage ALL patients with persistent or relapsed MRD, a T cell-engaging bispecific Ab construct induced an 80% MRD response rate. In the present study, we show that after a median follow-up of 33 months, the hematologic relapse-free survival of the entire evaluable study cohort of 20 patients was 61% (Kaplan-Meier estimate). The hema-tologic relapse-free survival rate of a subgroup of 9 patients who received allogeneic hematopoietic stem cell transplantation after blinatumomab treatment was 65% (Kaplan-Meier estimate). Of the subgroup of 6 Philadelphia chromosome-negative MRD responders with no further therapy after blinatumomab, 4 are in ongoing hematologic and molecular remission. We conclude that blinatumomab can induce long-lasting complete remission in B-lineage ALL patients with persistent or recurrent MRD. The original study and this follow-up study are registered at www.clinicaltrials.gov as NCT00198991 and NCT00198978, respectively.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Adulto , Antineoplásicos/uso terapêutico , Terapia Combinada , Intervalo Livre de Doença , Seguimentos , Transplante de Células-Tronco Hematopoéticas , Humanos , Estimativa de Kaplan-Meier , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidade , Recidiva , Análise de Sobrevida , Transplante Homólogo
16.
Antimicrob Agents Chemother ; 57(6): 2828-30, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23478971

RESUMO

Gamma interferon (IFN-γ)-mediated host responses play a central role in resolving genital Chlamydia trachomatis infections but may also result in persistence of the pathogen, which shows reduced susceptibility to antimicrobials. The antichlamydial function of IFN-γ is oxygen dependent, and the efficacy of antimicrobials against C. trachomatis is reduced in a low-oxygen environment. In this study, we show that the antichlamydial efficacies of azithromycin and doxycycline differ in IFN-γ-treated cells under hypoxia.


Assuntos
Antibacterianos/farmacologia , Azitromicina/farmacologia , Chlamydia trachomatis/efeitos dos fármacos , Doxiciclina/farmacologia , Interferon gama/farmacologia , Oxigênio/farmacologia , Antibacterianos/uso terapêutico , Azitromicina/uso terapêutico , Hipóxia Celular , Infecções por Chlamydia/tratamento farmacológico , Infecções por Chlamydia/microbiologia , Doxiciclina/uso terapêutico , Células HeLa/efeitos dos fármacos , Células HeLa/microbiologia , Humanos , Interferon gama/uso terapêutico
17.
FASEB J ; 26(1): 29-39, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21908716

RESUMO

Leishmania pathogenesis is primarily studied using the disease-inducing promastigote stage of Leishmania major. Despite many efforts, all attempts so far have failed to culture the disease-relevant multiplying amastigote stage of L. major. Here, we established a stably growing axenic L. major amastigote culture system that was characterized genetically, morphologically, and by stage-specific DsRed protein expression. We found parasite stage-specific disease development in resistant C57BL/6 mice. Human neutrophils, as first host cells for promastigotes, do not take up amastigotes. In human macrophages, we observed an amastigote-specific complement receptor 3-mediated, endocytotic entry mechanism, whereas promastigotes are taken up by complement receptor 1-mediated phagocytosis. Promastigote infection of macrophages induced the inflammatory mediators TNF, CCL3, and CCL4, whereas amastigote infection was silent and resulted in significantly increased parasite numbers: from 7.1 ± 1.4 (after 3 h) to 20.1 ± 7.9 parasites/cell (after 96 h). Our study identifies Leishmania stage-specific disease development, host cell preference, entry mechanism, and immune evasion. Since the amastigote stage is the disease-propagating form found in the infected mammalian host, the newly developed L. major axenic cultures will serve as an important tool in better understanding the amastigote-driven immune response in leishmaniasis.


Assuntos
Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/parasitologia , Macrófagos/parasitologia , Neutrófilos/parasitologia , Animais , Cultura Axênica/métodos , Endocitose/imunologia , Feminino , Expressão Gênica/fisiologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Leishmania major/genética , Leishmania major/crescimento & desenvolvimento , Macrófagos/imunologia , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Neutrófilos/imunologia , Neutrófilos/ultraestrutura , Fagocitose/imunologia
18.
Mediators Inflamm ; 2013: 710239, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24381411

RESUMO

Neutrophil extracellular traps (NETs) have been suggested to play a pathophysiological role in several autoimmune diseases. Since NET-formation in response to several biological and chemical stimuli is mostly ROS dependent, in theory any substance that inhibits or scavenges ROS could prevent ROS-dependent NET release. Therefore, in the present comprehensive study, several antioxidative substances were assessed for their capacity to inhibit NET formation of primary human neutrophils in vitro. We could show that the flavonoids (-)-epicatechin, (+)-catechin hydrate, and rutin trihydrate as well as vitamin C and the pharmacological substances N-acetyl-L-cysteine and 5-aminosalicylic acid inhibited PMA induced ROS production and NET formation. Therefore, a broad spectrum of antioxidative substances that reduce ROS production of primary human neutrophils also inhibits ROS-dependent NET formation. It is tempting to speculate that such antioxidants can have beneficial therapeutic effects in diseases associated with ROS-dependent NET formation.


Assuntos
Antioxidantes/farmacologia , Flavonoides/farmacologia , Mesalamina/farmacologia , Neutrófilos/efeitos dos fármacos , Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Ácido Ascórbico/farmacologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Humanos , NADPH Oxidases/antagonistas & inibidores , Neutrófilos/imunologia , Peroxidase/metabolismo , Fagocitose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
19.
Proc Natl Acad Sci U S A ; 107(45): 19502-7, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974954

RESUMO

IFN-γ has an important role in the adaptive immune response against intracellular pathogens. In urogenital tract (UGT) infections with the obligate intracellular pathogen Chlamydia trachomatis, IFN-γ-mediated control of chlamydial growth implies the JAK-STAT signaling cascades and subsequent induction of the indoleamine 2,3-dioxygenase (IDO). As oxygen concentrations in the UGT are low under physiological conditions (O(2) < 5%) and further decrease during an inflammatory process, we wondered whether antibacterial properties of IFN-γ are maintained under hypoxic conditions. Using primary cells that were isolated from human fallopian tubes and an ex vivo human fallopian tube model (HFTM), we found that even high IFN-γ concentrations (200 units/mL) were not sufficient to limit growth of C. trachomatis under hypoxia. Reduced antibacterial activity of IFN-γ under hypoxia was restricted to the urogenital serovars D and L(2), but was not observed with the ocular serovar A. Impaired effectiveness of IFN-γ on chlamydial growth under hypoxia was accompanied by reduced phosphorylation of Stat-1 on Tyr701 and diminished IDO activity. This study shows that IFN-γ effector functions on intracellular C. trachomatis depend on the environmental oxygen supply, which could explain inadequate bacterial clearance and subsequent chronic infections eventually occurring in the UGT of women.


Assuntos
Infecções por Chlamydia/imunologia , Tubas Uterinas/imunologia , Tubas Uterinas/microbiologia , Hipóxia/microbiologia , Interferon gama/imunologia , Infecções Urinárias/imunologia , Imunidade Adaptativa , Infecções por Chlamydia/etiologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/imunologia , Tubas Uterinas/citologia , Feminino , Humanos , Hipóxia/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Infecções Urinárias/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA