Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(20)2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34681717

RESUMO

The immunosuppressive character of head and neck cancers may explain the relatively low response rates to antibody therapy targeting a tumor antigen, such as cetuximab, and anti-PD-1 checkpoint inhibition. Immunostimulatory agents that overcome tumor-derived inhibitory signals could augment therapeutic efficacy, thereby enhancing tumor elimination and improving patient survival. Here, we demonstrate that cetuximab treatment combined with immunostimulatory agonists for Toll-like receptor (TLR) 2 induces profound immune responses. Natural killer (NK) cells, isolated from healthy individuals or patients with head and neck cancer, harbored enhanced cytotoxic capacity and increased tumor-killing potential in vitro. Additionally, combination treatment increased the release of several pro-inflammatory cytokines and chemokines by NK cells. Tumor-bearing mice that received cetuximab and the TLR2 ligand Pam3CSK4 showed increased infiltration of immune cells into the tumors compared to mice that received cetuximab monotherapy, resulting in a significant delay in tumor growth or even complete tumor regression. Moreover, combination treatment resulted in improved overall survival in vivo. In conclusion, combining tumor-targeting antibody-based immunotherapy with TLR stimulation represents a promising treatment strategy to improve the clinical outcomes of cancer patients. This treatment could well be applied together with other therapeutic strategies such as anti-PD-(L)1 checkpoint inhibition to further overcome immunosuppression.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Neoplasias de Cabeça e Pescoço/terapia , Células Matadoras Naturais/imunologia , Receptor 2 Toll-Like/agonistas , Animais , Linhagem Celular Tumoral , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Citocinas/metabolismo , Quimioterapia Combinada , Feminino , Humanos , Imunoterapia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipopeptídeos/farmacologia , Lipopeptídeos/uso terapêutico , Camundongos , Camundongos Nus , Receptores de IgG/agonistas , Receptores de IgG/metabolismo , Receptor 2 Toll-Like/metabolismo , Transplante Heterólogo
2.
Nat Commun ; 10(1): 4206, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31562320

RESUMO

Human immunoglobulin G (IgG) agonistic antibodies targeting costimulatory immunoreceptors represent promising cancer immunotherapies yet to be developed. Whether, and how, human IgG hinge and Fc impact on their agonistic functions have been disputed. Here, we show that different natural human IgGs confer divergent agonistic anti-CD40 immunostimulatory and antitumour activities in FcγR-humanized mice, including inactive IgG3 and superior IgG2. This divergence is primarily due to their CH1-hinges despite all human IgGs requiring Fc-FcγR binding for optimal agonistic activities. Unexpectedly, biophysical flexibility of these CH1-hinges inversely correlates with, and can modulate, their agonistic potency. Furthermore, IgG Fcs optimized for selective FcγR binding synergize with and still require IgG hinge, selected for rigidity, to confer improved anti-CD40 immunostimulatory and antitumour activities. These findings highlight the importance of both hinge rigidity and selective FcγR binding in antibody agonistic function, and the need for newer strategies to modulate antibody agonism for improved clinical application.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD40/imunologia , Imunoglobulina G/imunologia , Neoplasias/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoterapia/métodos , Camundongos Knockout , Camundongos Transgênicos , Neoplasias/metabolismo , Neoplasias/terapia , Ligação Proteica , Receptores de IgG/agonistas , Receptores de IgG/genética
3.
Hum Antibodies ; 25(3-4): 87-109, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28085016

RESUMO

Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Receptores de IgG/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Celular/efeitos dos fármacos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/agonistas , Receptores de IgG/genética , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia
4.
J Biol Chem ; 288(19): 13455-66, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23532848

RESUMO

BACKGROUND: Fn14 is a therapeutic target in various diseases. RESULTS: Anti-Fn14 antibodies activate the alternative NFκB pathway but not other Fn14-related activities induced by soluble or membrane-bound TWEAK. FcγR-bound anti-Fn14 antibodies, however, activate the full spectrum of Fn14-associated activities. CONCLUSION: Anti-Fn14 antibodies elicit agonistic activities differing from those of the natural Fn14 ligand TWEAK. SIGNIFICANCE: These findings influence the rationale of designing Fn14-targeted therapies. The Fn14-specific monoclonal antibodies PDL192 and P4A8, which are under consideration in clinical trials, showed no agonistic activity with respect to IL8 production and cell death induction. However, oligomerization with protein G or binding to Fcγ receptors converted both anti-Fn14 antibodies into potent agonists. TNF-like weak inducer of apoptosis (TWEAK), the ligand of Fn14, occurs naturally in two forms with partly different signaling capabilities, as a membrane-bound ligand and as a soluble trimeric molecule. Although membrane TWEAK strongly triggers all Fn14-associated pathways, soluble TWEAK predominately triggers the alternative nuclear factor κB (NFκB) pathway and enhances TNF-induced cell death but has only a poor effect on the classical NFκB pathway and chemokine production. Thus, the oligomerized and FcγR-bound anti-Fn14 mAbs mimicked the activity of membrane TWEAK. Notably, both anti-Fn14 antibodies significantly triggered p100 processing, the hallmark of the alternative NFκB pathway, and therefore resembled soluble TWEAK. In contrast to the latter, however, the anti-Fn14s showed no effect on TNF receptor 1-induced cell death and P4A8 even blocked the corresponding TWEAK response. Thus, we showed that Fn14 antibodies display an alternative NFκB pathway-specific agonistic activity but fail to phenocopy other activities of soluble TWEAK, whereas oligomerized or FcγR-bound Fn14 antibodies fully mimic the activity of membrane TWEAK. In view of the trivalent nature of the TWEAK-Fn14 interaction, this suggests that the alternative NFκB pathway is uniquely responsive already to Fn14 dimerization enabling antibodies to elicit an unnatural response pattern distinct from that of the naturally occurring Fn14 ligands.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais/farmacologia , Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais/efeitos dos fármacos , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados/química , Proteínas de Bactérias/química , Linhagem Celular Tumoral , Citocina TWEAK , Endonucleases , Células HEK293 , Humanos , Interleucina-8/biossíntese , Macaca fascicularis , Camundongos , Mutagênese Sítio-Dirigida , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica , Multimerização Proteica , Receptores de IgG/agonistas , Receptores de IgG/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor de TWEAK , Inibidores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/fisiologia , Fatores de Necrose Tumoral/fisiologia
5.
Leuk Res ; 37(4): 440-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23259986

RESUMO

Rituximab (RTX, anti-CD20 antibody) combined with chemotherapy is currently standard treatment for chronic lymphocytic leukemia (CLL). Serum level of IL-8 is a prognostic factor for CLL that correlates with disease stage. We investigated whether endogenous IL-8 affects RTX or Obinutuzumab (GA101) B-leukemic depletion mediated by natural killers (NK). Using whole peripheral blood lymphocytes from untreated CLL patients, RTX, but most significantly GA101, were effective in B-cell depletion and NK activation. IL-8 inhibition completely inhibited B-cell depletion by RTX and reduced GA101-induced B-cell depletion. Altogether results underline IL-8 as an endogenous NK co-activator and confirm GA101 therapeutic potential for CLL treatment.


Assuntos
Antígenos CD20/imunologia , Linfócitos B/imunologia , Interleucina-8/fisiologia , Células Matadoras Naturais/imunologia , Leucemia Linfocítica Crônica de Células B/imunologia , Depleção Linfocítica , Receptores de IgG/agonistas , Anticorpos Monoclonais/imunologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos
6.
J Thromb Haemost ; 10(6): 1133-41, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22489915

RESUMO

BACKGROUND: Ligation of the platelet-specific collagen receptor, GPVI/FcRγ, causes rapid, transient disulfide-dependent homodimerization, and the production of intracellular reactive oxygen species (ROS) generated by the NADPH oxidase, linked to GPVI via TRAF4. OBJECTIVES: The aim of this study was to evaluate the role of early signaling events in ROS generation following engagement of either GPVI/FcRγ or a second immunoreceptor tyrosine-based activation motif (ITAM)-containing receptor on platelets, FcγRIIa. METHODS AND RESULTS: Using an H(2) DCF-DA-based flow cytometric assay to measure intracellular ROS, we show that treatment of platelets with either the GPVI agonists, collagen-related peptide (CRP) or convulxin (Cvx), or the FcγRIIa agonist 14A2, increased intraplatelet ROS; other platelet agonists such as ADP and TRAP did not. Basal ROS in platelet-rich plasma from 14 healthy donors displayed little inter-individual variability. CRP, Cvx or 14A2 induced an initial burst of ROS within 2 min followed by additional ROS reaching a plateau after 15-20 min. The Syk inhibitor BAY61-3606, which blocks ITAM-dependent signaling, had no effect on the initial ROS burst, but completely inhibited the second phase. CONCLUSIONS: Together, these results show for the first time that ROS generation downstream of GPVI or FcγRIIa consists of two distinct phases: an initial Syk-independent burst followed by additional Syk-dependent generation.


Assuntos
Plaquetas/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de IgG/metabolismo , Transdução de Sinais , Motivos de Aminoácidos , Plaquetas/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Venenos de Crotalídeos/farmacologia , Ativação Enzimática , Feminino , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Lectinas Tipo C , Masculino , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Peptídeos/farmacologia , Glicoproteínas da Membrana de Plaquetas/agonistas , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Receptores de IgG/agonistas , Transdução de Sinais/efeitos dos fármacos , Quinase Syk , Fatores de Tempo
7.
J Immunol ; 183(10): 6808-18, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19846865

RESUMO

A major challenge for inducing antitumor immune responses with native or modified tumor/self-Ags in tumor-bearing hosts relates to achieving efficient uptake and processing by dendritic cells (DCs) to activate immune effector cells and limit the generation of regulatory T cell activity. We analyzed the ability of therapeutic DC vaccines expressing a CD166 cross-reactive mimotope of the GD2 ganglioside, 47-LDA, to selectively expand adoptively transferred, tumor-specific T cells in NXS2 neuroblastoma tumor-bearing syngeneic mice. Before the adoptive cell transfer and DC vaccination, the tumor-bearing mice were lymphodepleted by nonmyeloablative total body irradiation or a myeloablative regimen that required bone marrow transplantation. The 47-LDA mimotope was presented to DCs either as a linear polypeptide in conjunction with universal Th epitopes or as a fusion protein with the murine IgG2a Fc fragment (47-LDA-Fcgamma2a) to deliver the antigenic cassette to the activating Fcgamma receptors. We demonstrate that immunization of adoptively transferred T cells in tumor-bearing mice with the 47-LDA mimotope expressed in the context of the activating Fc fusion protein induced higher levels of antitumor immune responses and protection than the 47-LDA polypeptide-DC vaccine. The antitumor efficacy of the therapeutic 47-LDA-Fcgamma2a-DC vaccine was comparable to that achieved by a virotherapy-associated cancer vaccine using a recombinant oncolytic vaccinia virus expressing the 47-LDA-Fcgamma2a fusion protein. The latter treatment, however, did not require total body irradiation or adoptive cell transfer and resulted in induction of antitumor immune responses in the setting of established tolerance, paving the way for testing novel anticancer treatment strategies.


Assuntos
Molécula de Adesão de Leucócito Ativado/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Neuroblastoma/terapia , Molécula de Adesão de Leucócito Ativado/metabolismo , Transferência Adotiva , Animais , Epitopos/imunologia , Feminino , Gangliosídeos/imunologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos A , Neuroblastoma/imunologia , Terapia Viral Oncolítica , Receptores de IgG/agonistas , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/imunologia , Irradiação Corporal Total
8.
J Immunol ; 179(12): 8350-6, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18056380

RESUMO

PGE2 has important inhibitory effects on the macrophage host defense functions of phagocytosis and killing, yet the molecular mechanisms involved remain to be fully elucidated. PGE2 causes an elevation of cAMP in alveolar macrophages (AMs), which in turn activates the cAMP effector targets, protein kinase A and the exchange protein activated by cAMP (Epac)-1. We now report that FcgammaR-induced PI3K/Akt and ERK-1/2 activation are inhibited by PGE2 in AMs. By specifically inhibiting the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in AMs, we attenuated the inhibitory effects of both PGE2 and a specific Epac-1 agonist (8-pCPT-2'-O-Me-cAMP) on FcgammaR-mediated phagocytosis and Akt/ERK-1/2 activation; PTEN inhibition also decreased PGE2-induced suppression of bacterial killing by AMs. Moreover, PGE2 and the Epac-1 agonist induced an increase in PTEN lipid phosphatase activity, and this was associated with decreased tyrosine phosphorylation on PTEN-a mechanism known to regulate PTEN activity. Using a pharmacological approach, we demonstrated a role for Src homology 2-containing protein tyrosine phosphatase-1 in the PGE2-induced tyrosine dephosphorylation of PTEN. Collectively, these data reveal that PGE2, via Epac-1 activation, enhances SHP-1 activity, resulting in increased PTEN activity. We suggest that this mechanism contributes to the ability of PGE2 to inhibit PI3K-dependent innate immune signaling in primary macrophages.


Assuntos
Dinoprostona/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , PTEN Fosfo-Hidrolase/metabolismo , Fagocitose , Animais , Dinoprostona/farmacologia , Feminino , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Klebsiella pneumoniae/imunologia , Macrófagos Alveolares/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptores de IgG/agonistas , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/fisiologia , Tirosina/metabolismo
9.
Mol Cell Endocrinol ; 279(1-2): 16-25, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17923257

RESUMO

Estrogen can significantly influence CD16 expression and alter monocytic cytokine release upon CD16 receptor activation. However, the function of the estrogen receptor (ER) alpha and beta in this response is unclear. To test whether estrogen binds ERalpha and/or ERbeta to affect CD16 expression, monocytic cells were treated with and without physiological levels of 17beta-estradiol and various doses of the ERalpha and ERbeta antagonist fulvestrant followed by measurement of CD16 transcript levels. To determine how estrogen induced changes in TNF-alpha and IL-1beta release due to CD16 activation we quantitated the amount of cytokines after treatment with estrogen, fulvestrant and antibodies that specifically bind and activate the CD16 receptor. Interaction of ERalpha and the CD16 promoter was then determined by chromatin immunoprecipitation. Furthermore, specific promoter elements utilized by estrogen to control CD16 expression were mutated and expression from a luciferase reporter quantitated after transfection. Using the luciferase reporter construct containing a wild type CD16 promoter, the role of ERalpha and ERbeta in the estrogen response was tested by treating transfected monocytes with an ERalpha specific agonist or an ERbeta specific agonist and measuring expression. Our results show that CD16 transcript levels significantly decreased in monocytic cells due to estrogen and that the observed decrease in message was blocked by the antagonist fulvestrant. Estrogen reduced CD16 expression and decreased TNF-alpha and IL-1beta release upon CD16 activation but the administration of fulvestrant blocked this decrease. ERalpha was found to interact with a region 5' of the CD16 gene in the presence of estrogen, and site-directed mutational analysis of this region indicated the necessity for an estrogen response element in modulating estrogen effects on CD16 expression. Moreover, both an ERalpha and an ERbeta agonist reduced expression of the CD16 reporter construct suggesting both receptors can play a role in CD16 regulation. In conclusion, CD16 expression can be altered by the activity of ERalpha or ERbeta and our results also show that ERalpha can associate with a region within the CD16 promoter that is important in production of transcript.


Assuntos
Estradiol/fisiologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Monócitos/metabolismo , Receptores de IgG/metabolismo , Anticorpos/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Monócitos/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores de IgG/agonistas , Receptores de IgG/genética , Fator de Necrose Tumoral alfa/metabolismo
10.
J Immunol ; 178(7): 4033-8, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17371957

RESUMO

The recently described TL1A/DR3 ligand/receptor pair mediates strong costimulation of Th1 cells. Activation of T and NK cells induces DR3 expression, permitting soluble recombinant TL1A to increase IFN-gamma production and proliferation of these cells. Gut T cells and macrophages express TL1A, especially in Crohn's disease (CD), and there is a strong association between CD and tl1a single nucleotide polymorphisms. Murine studies implicate TL1A in gut inflammation. To determine whether professional T cell-activating cells can express TL1A, fresh blood monocytes and monocyte-derived dendritic cells were stimulated with various activating ligands, including TLR agonists, IFN-gamma, and immune complexes. FcgammaR stimulation strongly induced TL1A mRNA in both cell types, which correlated with the detection of TL1A on the cell surface and in cell culture medium. TLR agonists capable of inducing IL-6 and TNF-alpha in monocytes and dendritic cells did not induce surface nor soluble TL1A. Furthermore, we demonstrate that TL1A production in monocytes leads to enhancement of T cell responses. The induction of TL1A on APCs via specific pathway stimulation suggests a role for TL1A in Th1 responses to pathogens, and in CD.


Assuntos
Células Dendríticas/imunologia , Monócitos/imunologia , Receptores de IgG/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Membrana Celular/química , Membrana Celular/imunologia , Células Cultivadas , Células Dendríticas/química , Humanos , Interferon gama/metabolismo , Ligantes , Monócitos/química , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/agonistas , Receptores de IgG/agonistas , Transdução de Sinais , Linfócitos T/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/análise , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
11.
J Immunol ; 178(6): 3702-12, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17339468

RESUMO

Pneumocystis carinii (PC) pneumonia is a leading opportunistic infection found among HIV-infected individuals worldwide. Although CD4(+) T cell deficiency clearly correlates with susceptibility to PC pneumonia, murine models of disease indicate that PC-directed Abs may prevent infection and/or inhibit growth of existing PC within the lungs. Recognition of PC by alveolar macrophages involves the beta-glucan receptor Dectin-1 and macrophage effector function against PC is enhanced by Abs derived from PC-vaccinated hosts. We developed a fusion protein consisting of the extracellular domain of Dectin-1 linked to the Fc portion of murine IgG1, which we hypothesized would enhance host recognition and opsonic phagocytosis of PC. The recombinant protein, Dectin-Fc, is dimeric and the Ag recognition site identifies beta-1,3 glucan linkages specifically and with high affinity (K(D) = 2.03 x 10(-7) M). Dectin-Fc enhances RAW264.7 macrophage recognition of the beta-glucan containing particulate zymosan in an FcgammaRII- and FcgammaRIII-dependent manner and preopsonization of PC organisms with Dectin-Fc increased alveolar and peritoneal macrophage-dependent killing of PC. SCID mice treated with a replication incompetent adenoviral vector expressing Dectin-Fc had attenuated growth of PC within the lungs, overall decreased PC lung burden, and diminished correlates of PC-related lung damage relative to SCID mice receiving a control vector. These findings demonstrate that targeting PC beta-glucan with Dectin-Fc enhances host recognition and clearance of PC in the absence of B and T cells, and suggest that FcgammaR-based targeting of PC, via cell wall carbohydrate recognition, may promote resistance against PC pneumonia in the immunodeficient host.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/imunologia , Hospedeiro Imunocomprometido/imunologia , Regiões Constantes de Imunoglobulina/farmacologia , Proteínas de Membrana/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Pneumocystis carinii/imunologia , Pneumonia por Pneumocystis/imunologia , Infecções Oportunistas Relacionadas com a AIDS/prevenção & controle , Adenoviridae , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/genética , Modelos Animais de Doenças , Humanos , Regiões Constantes de Imunoglobulina/genética , Lectinas Tipo C , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos Alveolares/imunologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos SCID , Proteínas do Tecido Nervoso/genética , Pneumonia por Pneumocystis/terapia , Receptores de IgG/agonistas , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , beta-Glucanas/imunologia
12.
J Immunol ; 177(9): 6291-300, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17056559

RESUMO

We previously reported that the stimulation of monocyte-derived macrophages (MDM) by plate-bound i.v. Igs inhibits HIV-1 replication. In this study, we show that IgG immune complexes also suppress HIV-1 replication in MDMs and that activating receptors for the Fc portion of IgG-FcgammaRI, FcgammaRIIA, and FcgammaRIII-are responsible for the inhibition. MDM stimulation through FcgammaRs induces activation signals and the secretion of HIV-1 modulatory cytokines, such as M-CSF, TNF-alpha, and macrophage-derived chemokine. However, none of these cytokines contribute to HIV-1 suppression. HIV-1 entry and postintegration steps of viral replication are not affected, whereas reduced levels of reverse transcription products and of integrated proviruses, as determined by real-time PCR analysis, account for the suppression of HIV-1 gene expression in FcgammaR-activated MDMs. We found that FcgammaR-dependent activation of MDMs also inhibits the replication of HIV-2, SIVmac, and SIVagm, suggesting a common control mechanism for primate immunodeficiency lentiviruses in activated macrophages.


Assuntos
HIV-1/fisiologia , Macrófagos/imunologia , Macrófagos/virologia , Receptores de IgG/agonistas , Replicação Viral , Animais , Células Cultivadas , DNA Complementar/análise , DNA Viral/análise , Humanos , Lentivirus/fisiologia , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Primatas/virologia , Provírus/isolamento & purificação , Receptores de IgG/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA