Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(11): eadj6406, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38489355

RESUMEN

There is a compelling need to find drugs active against Mycobacterium tuberculosis (Mtb). 4'-Phosphopantetheinyl transferase (PptT) is an essential enzyme in Mtb that has attracted interest as a potential drug target. We optimized a PptT assay, used it to screen 422,740 compounds, and identified raltitrexed, an antineoplastic antimetabolite, as the most potent PptT inhibitor yet reported. While trying unsuccessfully to improve raltitrexed's ability to kill Mtb and remove its ability to kill human cells, we learned three lessons that may help others developing antibiotics. First, binding of raltitrexed substantially changed the configuration of the PptT active site, complicating molecular modeling of analogs based on the unliganded crystal structure or the structure of cocrystals with inhibitors of another class. Second, minor changes in the raltitrexed molecule changed its target in Mtb from PptT to dihydrofolate reductase (DHFR). Third, the structure-activity relationship for over 800 raltitrexed analogs only became interpretable when we quantified and characterized the compounds' intrabacterial accumulation and transformation.


Asunto(s)
Mycobacterium tuberculosis , Neoplasias , Quinazolinas , Tiofenos , Transferasas (Grupos de Otros Fosfatos Sustitutos) , Humanos , Mycobacterium tuberculosis/metabolismo , Timidilato Sintasa/metabolismo , Proteínas Bacterianas/metabolismo
2.
Antimicrob Agents Chemother ; 68(2): e0104323, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38132181

RESUMEN

Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDTs) offer a novel approach to TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that the inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here, we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO), in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P = 0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 colony-forming units (CFUs), a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P = 0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.


Asunto(s)
Metaloporfirinas , Mycobacterium tuberculosis , Protoporfirinas , Tuberculosis Resistente a Múltiples Medicamentos , Animales , Ratones , Metaloporfirinas/uso terapéutico , Hemo-Oxigenasa 1 , Modelos Animales de Enfermedad , Antituberculosos/farmacología , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Recurrencia
3.
bioRxiv ; 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37609351

RESUMEN

Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDT) offer a novel approach for TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO) in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5 mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P=0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes, and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10 mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 CFU, a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P=0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.

4.
Clin Cancer Res ; 26(22): 5895-5902, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32928795

RESUMEN

PURPOSE: Addition of daratumumab to pomalidomide and low-dose dexamethasone (LoDEX) is a safe and effective combination for relapsed/refractory multiple myeloma treatment. We sought to better understand immune combinational benefit of pomalidomide and daratumumab with LoDEX. PATIENTS AND METHODS: Immunophenotypic changes were analyzed in peripheral blood from longitudinal sampling of patients treated with this triplet regimen from cohort B of the CC4047-MM-014 phase II trial (NCT01946477). RESULTS: Consistent with the daratumumab mechanism, treatment led to decreased natural killer (NK) and B cells. In contrast, pronounced increases occurred in activated and proliferating NK and T cells, appreciably in CD8+ T cells, along with reduction in naïve and expansion of effector memory compartments. Timing of T-cell changes correlated with pomalidomide dosing schedule. Enhanced activation/differentiation did not result in increased exhausted T-cell phenotypes or increases in regulatory T cells. Similar immune enhancements were also observed in patients previously refractory to lenalidomide. CONCLUSIONS: These data support a potential mechanism for enhanced immune-mediated cytotoxicity in which daratumumab-mediated NK-cell diminution is partially offset by pomalidomide effects on the remaining NK-cell pool. Furthermore, daratumumab antimyeloma activity and elimination of CD38+ T cells (regulatory/activated) provide a rationale for therapeutic combination with direct tumoricidal activity and immunomodulation of pomalidomide-directed T-cell enhancements. These data highlight enhancements in immune subpopulations for the combination of daratumumab with pomalidomide and potentially with next-generation cereblon-targeting agents.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Dexametasona/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Talidomida/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Inmunomodulación/efectos de los fármacos , Inmunofenotipificación , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Lenalidomida/administración & dosificación , Masculino , Persona de Mediana Edad , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Talidomida/administración & dosificación
5.
Blood ; 133(11): 1217-1221, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30692124

RESUMEN

Deletions of chromosome 17p (del17p) that span the TP53 gene are associated with poor outcome in multiple myeloma (MM), but the prognostic value of del17p cancer clonal fraction (CCF) remains unclear. We applied uniform cytogenetic assessments in a large cohort of newly diagnosed MM (NDMM) patients carrying varying levels of del17p. Incremental CCF change was associated with shorter survival, and a robust CCF threshold of 0.55 was established in discovery and replication data sets. After stratification on the 0.55-CCF threshold, high-risk patients had statistically significantly poorer outcomes compared with low-risk patients (median progression-free survival [PFS] and overall survival [OS], 14 and 32 vs 23.1 and 76.2 months, respectively). Analyses of a third data set comprising whole-exome sequencing data from NDMM patients identified presence of TP53 deletions/mutations as a necessary requirement for high-risk stratification in addition to exceeding the del17p CCF threshold. Meta-analysis conducted across 3 data sets confirmed the robustness of the CCF threshold for PFS and OS. Our analyses demonstrate the feasibility of fluorescence in situ hybridization- and sequencing-based methods to identify TP53 deletions, estimate CCF, and establish that both CCF threshold of 0.55 and presence of TP53 deletion are necessary to identify del17p-carrying NDMM patients with poor prognosis.


Asunto(s)
Biomarcadores de Tumor/genética , Deleción Cromosómica , Cromosomas Humanos Par 17/genética , Evolución Clonal , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Proteína p53 Supresora de Tumor/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mieloma Múltiple/patología , Mutación , Pronóstico , Tasa de Supervivencia
6.
Artículo en Inglés | MEDLINE | ID: mdl-29133571

RESUMEN

The recommended management of inhalational anthrax, a high-priority bioterrorist threat, includes antibiotics and antitoxins. Obiltoxaximab, a chimeric monoclonal antibody against anthrax protective antigen (PA), is licensed under the U.S. Food and Drug Administration's (FDA's) Animal Rule for the treatment of inhalational anthrax. Because of spore latency, disease reemergence after treatment cessation is a concern, and there is a need to understand the development of endogenous protective immune responses following antitoxin-containing anthrax treatment regimens. Here, acquired protective immunity was examined in New Zealand White (NZW) rabbits challenged with a targeted lethal dose of Bacillus anthracis spores and treated with antibiotics, obiltoxaximab, or a combination of both. Survivors of the primary challenge were rechallenged 9 months later and monitored for survival. Survival rates after primary and rechallenge for controls and animals treated with obiltoxaximab, levofloxacin, or a combination of both were 0, 65, 100, and 95%, and 0, 100, 95, and 89%, respectively. All surviving immune animals had circulating antibodies to PA and serum toxin-neutralizing titers prior to rechallenge. Following rechallenge, systemic bacteremia and toxemia were not detected in most animals, and the levels of circulating anti-PA IgG titers increased starting at 5 days postrechallenge. We conclude that treatment with obiltoxaximab, alone or combined with antibiotics, significantly improves the survival of rabbits that received a lethal inhalation B. anthracis spore challenge dose and does not interfere with the development of immunity. Survivors of primary challenge are protected against reexposure, have rare incidents of systemic bacteremia and toxemia, and have evidence of an anamnestic response.


Asunto(s)
Carbunco , Antibacterianos , Anticuerpos Monoclonales , Antitoxinas , Bacillus anthracis , Levofloxacino , Infecciones del Sistema Respiratorio , Esporas Bacterianas , Animales , Femenino , Masculino , Conejos , Carbunco/inmunología , Carbunco/microbiología , Carbunco/mortalidad , Carbunco/prevención & control , Antibacterianos/farmacología , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Monoclonales/farmacología , Antígenos Bacterianos/sangre , Antígenos Bacterianos/inmunología , Antitoxinas/farmacología , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/sangre , Toxinas Bacterianas/inmunología , Quimioterapia Combinada , Inmunización Pasiva/métodos , Inmunoglobulina G/biosíntesis , Memoria Inmunológica/efectos de los fármacos , Levofloxacino/farmacología , Distribución Aleatoria , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/mortalidad , Infecciones del Sistema Respiratorio/prevención & control , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/inmunología , Esporas Bacterianas/patogenicidad , Análisis de Supervivencia
7.
Antimicrob Agents Chemother ; 60(10): 5796-805, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431219

RESUMEN

The Centers for Disease Control and Prevention recommend adjunctive antitoxins when systemic anthrax is suspected. Obiltoxaximab, a monoclonal antibody against protective antigen (PA), is approved for treatment of inhalational anthrax in combination with antibiotics and for prophylaxis when alternative therapies are not available. The impact of toxin neutralization with obiltoxaximab during pre- and postexposure prophylaxis was explored, and efficacy results that supported the prophylaxis indication are presented here. New Zealand White rabbits and cynomolgus macaques received obiltoxaximab as a single intramuscular or intravenous dose of 2 to 16 mg/kg of body weight at various times relative to Bacillus anthracis aerosol spore challenge. The primary endpoint was survival, and effect of treatment timing was explored. In rabbits, obiltoxaximab administration 9 h postchallenge singly or combined with a 5-day levofloxacin regimen protected 89% to 100% of animals compared to 33% with levofloxacin monotherapy. In cynomolgus macaques, a single intramuscular dose of 16 mg/kg obiltoxaximab led to 100% survival when given 1 to 3 days preexposure and 83% to 100% survival when given 18 to 24 h postexposure and prior to systemic bacteremia onset. Obiltoxaximab administration after bacteremia onset resulted in lower (25% to 50%) survival rates reflective of treatment setting. Prophylactic administration of obiltoxaximab before spore challenge or to spore-challenged animals before systemic bacterial dissemination is efficacious in promoting survival, ameliorating toxemia, and inhibiting bacterial spread to the periphery.


Asunto(s)
Carbunco/mortalidad , Carbunco/prevención & control , Anticuerpos Monoclonales/farmacología , Antitoxinas/farmacología , Bacillus anthracis/patogenicidad , Infecciones del Sistema Respiratorio/mortalidad , Infecciones del Sistema Respiratorio/prevención & control , Animales , Carbunco/tratamiento farmacológico , Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Antibacterianos/farmacología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Antitoxinas/administración & dosificación , Bacillus anthracis/efectos de los fármacos , Bacteriemia/tratamiento farmacológico , Bacteriemia/microbiología , Modelos Animales de Enfermedad , Femenino , Inyecciones Intramusculares , Inyecciones Intravenosas , Macaca fascicularis , Masculino , Profilaxis Posexposición , Profilaxis Pre-Exposición , Conejos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Tasa de Supervivencia
8.
Antimicrob Agents Chemother ; 60(10): 5787-95, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431222

RESUMEN

Inhalational anthrax has high mortality even with antibiotic treatment, and antitoxins are now recommended as an adjunct to standard antimicrobial regimens. The efficacy of obiltoxaximab, a monoclonal antibody against anthrax protective antigen (PA), was examined in multiple studies conducted in two animal models of inhalational anthrax. A single intravenous bolus of 1 to 32 mg/kg of body weight obiltoxaximab or placebo was administered to New Zealand White rabbits (two studies) and cynomolgus macaques (4 studies) at disease onset (significant body temperature increase or detection of serum PA) following lethal challenge with aerosolized Bacillus anthracis spores. The primary endpoint was survival. The relationship between efficacy and disease severity, defined by pretreatment bacteremia and toxemia levels, was explored. In rabbits, single doses of 1 to 16 mg/kg obiltoxaximab led to 17 to 93% survival. In two studies, survival following 16 mg/kg obiltoxaximab was 93% and 62% compared to 0% and 0% for placebo (P = 0.0010 and P = 0.0013, respectively). Across four macaque studies, survival was 6.3% to 78.6% following 4 to 32 mg/kg obiltoxaximab. In two macaque studies, 16 mg/kg obiltoxaximab reduced toxemia and led to survival rates of 31%, 35%, and 47% versus 0%, 0%, and 6.3% with placebo (P = 0.0085, P = 0.0053, P = 0.0068). Pretreatment bacteremia and toxemia levels inversely correlated with survival. Overall, obiltoxaximab monotherapy neutralized PA and increased survival across the range of disease severity, indicating clinical benefit of toxin neutralization with obiltoxaximab in both early and late stages of inhalational anthrax.


Asunto(s)
Carbunco/tratamiento farmacológico , Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Antitoxinas/farmacología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Animales , Carbunco/etiología , Carbunco/mortalidad , Antibacterianos/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Femenino , Macaca fascicularis , Masculino , Conejos , Infecciones del Sistema Respiratorio/etiología , Infecciones del Sistema Respiratorio/mortalidad , Tasa de Supervivencia , Resultado del Tratamiento
9.
Adv Immunol ; 113: 119-34, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22244581

RESUMEN

Infection of mice with Listeria monocytogenes induces a robust innate inflammatory response that restricts bacterial growth in the liver and spleen prior to the development of protective T cell responses. Ly6C(hi) monocytes contribute to the innate immune response following L. monocytogenes infection and in their absence, mice rapidly succumb to infection. Emigration of Ly6C(hi) monocytes from the bone marrow into the circulation is the first step in their recruitment to sites of L. monocytogenes infection and is triggered by CCL2- and CCL7-mediated stimulation of CCR2 chemokine receptors on monocytes. CCL2 expression by mesenchymal stem cells in the bone marrow, in response to TLR stimulation, drives monocyte emigration from cellular compartments into vascular sinuses of the bone marrow. In addition to TLR ligands, type I interferon-mediated signals can also drive monocyte emigration from the bone marrow during L. monocytogenes infection. Once Ly6C(hi) monocytes enter the bloodstream, trafficking to sites of infection in the liver and spleen is CCR2 independent. In the liver, CD11b on the monocyte and ICAM-1 on the surface of endothelial cells target Ly6C(hi) monocytes to foci of L. monocytogenes infection. At the site of infection, Ly6C(hi) monocytes undergo MyD88-dependent differentiation into TNF and iNOS-producing dendritic cells (TipDCs) and express MHC class II, B7.1, and CD40 on their cell surface. How TipDCs mediate bacterial clearance during early L. monocytogenes infection remains an active area of investigation.


Asunto(s)
Citocinas/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Monocitos/inmunología , Animales , Médula Ósea/inmunología , Diferenciación Celular , Movimiento Celular , Modelos Animales de Enfermedad , Humanos , Listeria monocytogenes/patogenicidad , Ratones , Monocitos/microbiología , Transducción de Señal
10.
Immunity ; 34(4): 590-601, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21458307

RESUMEN

Inflammatory (Ly6C(hi) CCR2+) monocytes provide defense against infections but also contribute to autoimmune diseases and atherosclerosis. Monocytes originate from bone marrow and their entry into the bloodstream requires stimulation of CCR2 chemokine receptor by monocyte chemotactic protein-1 (MCP1). How monocyte emigration from bone marrow is triggered by remote infections remains unclear. We demonstrated that low concentrations of Toll-like receptor (TLR) ligands in the bloodstream drive CCR2-dependent emigration of monocytes from bone marrow. Bone marrow mesenchymal stem cells (MSCs) and their progeny, including CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells, rapidly expressed MCP1 in response to circulating TLR ligands or bacterial infection and induced monocyte trafficking into the bloodstream. Targeted deletion of MCP1 from MSCs impaired monocyte emigration from bone marrow. Our findings suggest that bone marrow MSCs and CAR cells respond to circulating microbial molecules and regulate bloodstream monocyte frequencies by secreting MCP1 in proximity to bone marrow vascular sinuses.


Asunto(s)
Médula Ósea/inmunología , Movimiento Celular , Células Madre Mesenquimatosas/inmunología , Monocitos/citología , Monocitos/inmunología , Receptores Toll-Like/inmunología , Animales , Ligandos , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/inmunología
11.
J Immunol ; 183(4): 2678-87, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19635902

RESUMEN

Aspergillus fumigatus is an environmental fungus that causes life-threatening infections in neutropenic patients. In the absence of intact innate immunity, inhaled A. fumigatus spores (conidia) germinate in the lung, forming hyphae that invade blood vessels and disseminate to other tissues. Although macrophages and neutrophils are postulated to provide defense against invasive fungal infection, animal models and human studies suggest that circulating monocytes also contribute to antifungal immunity. Although human monocyte subsets, defined as either CD14(+)CD16(-) or CD14(+)CD16(+), have been extensively characterized, their respective roles during fungal infection remain undefined. We isolated CD14(+)CD16(-) and CD14(+)CD16(+) monocytes from healthy allogeneic hematopoietic stem cell transplantation donors and compared their ability to phagocytose and inhibit A. fumigatus conidia. Both monocyte subsets efficiently phagocytose conidia, but only CD14(+)CD16(-) monocytes inhibit conidial germination yet secrete little TNF. In contrast CD14(+)CD16(+) do not inhibit conidial germination and secrete large amounts of TNF. Although CD14(+)CD16(-) and CD14(+)CD16(+) monocytes differ in their response to dormant conidia, responses are similar if conidia are already germinated at the time of monocyte uptake. Our study demonstrates that functional CD14(+)CD16(-) and CD14(+)CD16(+) monocytes can be isolated from allogeneic hematopoietic stem cell transplantation donors and that these subsets differ in their response to A. fumigatus conidia.


Asunto(s)
Aspergillus fumigatus/inmunología , Monocitos/inmunología , Monocitos/microbiología , Esporas Fúngicas/inmunología , Antígenos CD34/biosíntesis , Aspergillus fumigatus/crecimiento & desarrollo , Células Cultivadas , Regulación hacia Abajo/inmunología , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Humanos , Receptores de Lipopolisacáridos/biosíntesis , Monocitos/citología , Receptores de IgG/biosíntesis , Receptores de IgG/metabolismo , Esporas Fúngicas/crecimiento & desarrollo , Regulación hacia Arriba/inmunología
12.
J Immunol ; 183(3): 1900-10, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19596996

RESUMEN

CCR2-mediated recruitment of Ly6C(high) monocytes is essential for defense against a range of microbial pathogens. Although our understanding of monocyte trafficking to inflammatory sites is increasing, how innate immune inflammation influences monocyte development and maturation during microbial infection remains undefined. Herein, we demonstrate that infection with the intracellular bacterial pathogen Listeria monocytogenes specifically and selectively promotes monopoiesis. Systemic infection with virulent L. monocytogenes induces marked proliferation of bone marrow monocyte precursors and results in depletion of myeloid progenitors. Proliferation of monocyte precursors correlates with the intensity of systemic infection and is unaffected by the density of monocytes in the bone marrow. Although MyD88/Trif-mediated signaling is not required for early emigration of the mature monocyte population from the bone marrow, replenishment of monocyte populations depends on MyD88/Trif. Our studies demonstrate that TLR-mediated signals play an essential role in the maintenance of monocyte homeostasis during systemic bacterial infection.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Infecciones Bacterianas/inmunología , Proliferación Celular , Células Precursoras de Monocitos y Macrófagos/citología , Monocitos/citología , Factor 88 de Diferenciación Mieloide/fisiología , Animales , Células de la Médula Ósea/citología , Linaje de la Célula , Quimiotaxis de Leucocito , Homeostasis , Listeria monocytogenes , Listeriosis/inmunología , Ratones , Ratones Noqueados , Monocitos/inmunología , Factor 88 de Diferenciación Mieloide/deficiencia , Receptores Toll-Like/metabolismo
14.
J Immunol ; 180(10): 6846-53, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18453605

RESUMEN

Chemokine receptor-mediated recruitment of inflammatory cells is essential for innate immune defense against microbial infection. Recruitment of Ly6C(high) inflammatory monocytes from bone marrow to sites of microbial infection is dependent on CCR2, a chemokine receptor that responds to MCP-1 and MCP-3. Although CCR2(-/-) mice are markedly more susceptible to Listeria monocytogenes infection than are wild-type mice, MCP-1(-/-) mice have an intermediate phenotype, suggesting that other CCR2 ligands contribute to antimicrobial defense. Herein, we show that L. monocytogenes infection rapidly induces MCP-3 in tissue culture macrophages and in serum, spleen, liver, and kidney following in vivo infection. Only cytosol invasive L. monocytogenes induce MCP-3, suggesting that cytosolic innate immune detection mechanisms trigger chemokine production. MCP-3(-/-) mice clear bacteria less effectively from the spleen than do wild-type mice, a defect that correlates with diminished inflammatory monocyte recruitment. MCP-3(-/-) mice have significantly fewer Ly6C(high) monocytes in the spleen and bloodstream, and increased monocyte numbers in bone marrow. MCP-3(-/-) mice, like MCP-1(-/-) mice, have fewer TNF- and inducible NO synthase-producing dendritic cells (Tip-DCs) in the spleen following L. monocytogenes infection. Our data demonstrate that MCP-3 and MCP-1 provide parallel contributions to CCR2-mediated inflammatory monocyte recruitment and that both chemokines are required for optimal innate immune defense against L. monocytogenes infection.


Asunto(s)
Quimiocina CCL2/inmunología , Quimiocina CCL7/inmunología , Quimiotaxis de Leucocito/inmunología , Listeriosis/inmunología , Monocitos/inmunología , Receptores CCR2/inmunología , Animales , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inflamación/inmunología , Listeria monocytogenes/inmunología , Macrófagos/inmunología , Ratones , Ratones Mutantes , Receptores CCR2/metabolismo
15.
Annu Rev Immunol ; 26: 421-52, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18303997

RESUMEN

Circulating blood monocytes supply peripheral tissues with macrophage and dendritic cell (DC) precursors and, in the setting of infection, also contribute directly to immune defense against microbial pathogens. In humans and mice, monocytes are divided into two major subsets that either specifically traffic into inflamed tissues or, in the absence of overt inflammation, constitutively maintain tissue macrophage/DC populations. Inflammatory monocytes respond rapidly to microbial stimuli by secreting cytokines and antimicrobial factors, express the CCR2 chemokine receptor, and traffic to sites of microbial infection in response to monocyte chemoattractant protein (MCP)-1 (CCL2) secretion. In murine models, CCR2-mediated monocyte recruitment is essential for defense against Listeria monocytogenes, Mycobacterium tuberculosis, Toxoplasma gondii, and Cryptococcus neoformans infection, implicating inflammatory monocytes in defense against bacterial, protozoal, and fungal pathogens. Recent studies indicate that inflammatory monocyte recruitment to sites of infection is complex, involving CCR2-mediated emigration of monocytes from the bone marrow into the bloodstream, followed by trafficking into infected tissues. The in vivo mechanisms that promote chemokine secretion, monocyte differentiation and trafficking, and finally monocyte-mediated microbial killing remain active and important areas of investigation.


Asunto(s)
Infecciones/inmunología , Monocitos/inmunología , Animales , Diferenciación Celular/inmunología , Quimiotaxis/inmunología , Humanos , Inmunidad Celular/inmunología , Inmunidad Mucosa/inmunología , Infecciones/microbiología , Infecciones/fisiopatología , Proteínas Quimioatrayentes de Monocitos/fisiología , Monocitos/citología , Receptores CCR2/fisiología
16.
Immunity ; 26(1): 67-78, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17239631

RESUMEN

Intravenous immune globulin (IVIG) suppresses autoantibody-mediated inflammation by inducing and activating the inhibitory Fc receptor FcgammaRIIb and downstream negative signaling pathways. We investigated the effects of IVIG on cellular responses to interferon-gamma (IFN-gamma), a potent macrophage activator that exacerbates inflammation. Our study showed that IVIG blocked IFN-gamma signaling and IFN-gamma-induced gene expression and suppressed IFN-gamma function in vivo during immune responses to Listeria monocytogenes and in an IFN-gamma-enhanced model of immune thrombocytopenic purpura. The mechanism of inhibition of IFN-gamma signaling was suppression of expression of the IFNGR2 subunit of the IFN-gamma receptor. The inhibitory effect of IVIG was mediated at least in part by soluble immune complexes and was dependent on FcgammaRIII but independent of FcgammaRIIb. These results reveal an unexpected inhibitory role for the activating FcgammaRIII in mediating suppression of IFN-gamma signaling and suggest that inhibition of macrophage responses to IFN-gamma contributes to the anti-inflammatory properties of IVIG.


Asunto(s)
Inmunoglobulinas Intravenosas/farmacología , Interferón gamma/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Receptores de IgG/inmunología , Transducción de Señal/inmunología , Animales , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Inmunoglobulinas Intravenosas/inmunología , Interferón gamma/inmunología , Listeriosis/inmunología , Activación de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Receptores de IgG/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
17.
Nat Immunol ; 7(3): 311-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16462739

RESUMEN

Monocytes recruited to tissues mediate defense against microbes or contribute to inflammatory diseases. Regulation of the number of circulating monocytes thus has implications for disease pathogenesis. However, the mechanisms controlling monocyte emigration from the bone marrow niche where they are generated remain undefined. We demonstrate here that the chemokine receptor CCR2 was required for emigration of Ly6C(hi) monocytes from bone marrow. Ccr2(-/-) mice had fewer circulating Ly6C(hi) monocytes and, after infection with Listeria monocytogenes, accumulated activated monocytes in bone marrow. In blood, Ccr2(-/-) monocytes could traffic to sites of infection, demonstrating that CCR2 is not required for migration from the circulation into tissues. Thus, CCR2-mediated signals in bone marrow determine the frequency of Ly6C(hi) monocytes in the circulation.


Asunto(s)
Infecciones Bacterianas/inmunología , Quimiotaxis de Leucocito/inmunología , Monocitos/inmunología , Receptores de Quimiocina/inmunología , Animales , Antígenos Ly/inmunología , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Diferenciación Celular , Citometría de Flujo , Ratones , Monocitos/citología , Receptores CCR2
18.
Infect Immun ; 72(2): 1057-64, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14742553

RESUMEN

Chemokine receptor 5 (CCR5) binds macrophage inflammatory protein 1alpha (MIP-1alpha), MIP-1beta, RANTES, and members of the monocyte chemotactic protein family and is also a receptor for human immunodeficiency virus (HIV). CCR5 ligands can suppress HIV-1 entry into cells. In humans, homozygous mutations of the ccr5 gene confer resistance to HIV-1 infection. The role of CCR5 in defense against microbial infection is unclear. In this study we examined the innate and adaptive immune responses of CCR5-deficient mice to the intracellular bacterial pathogen Listeria monocytogenes. We found that migration of monocytic cells, formation of L. monocytogenes-containing lesions, and bacterial clearance occurred normally in the spleens and livers of CCR5-deficient animals. Activation of macrophages and dendritic cells during the first 3 days postinfection was normal in the absence of CCR5, as demonstrated by intact expression of inducible nitric oxide synthase (iNOS) and production of the cytokines tumor necrosis factor alpha, gamma interferon, and interleukin-12. Priming of L. monocytogenes-specific CD8 T cells also occured independently of CCR5 expression. Previously immunized, CCR5-deficient animals mounted normal secondary CD8 T-cell responses and cleared bacteria from infected organs similarly to wild-type controls, suggesting that CCR5 is dispensable for migration and activation of memory CD8 T cells. Our data indicate that CCR5-mediated chemotaxis is not required for defense against infection with L. monocytogenes.


Asunto(s)
Toxinas Bacterianas , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptores CCR5/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Movimiento Celular , Células Dendríticas/fisiología , Proteínas de Choque Térmico/inmunología , Proteínas Hemolisinas , Inmunidad Innata , Activación de Linfocitos , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL
19.
Immunity ; 19(6): 891-901, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14670305

RESUMEN

Microbial infections induce chemokine and cytokine cascades that coordinate innate immune defenses. Infection with the intracellular bacterial pathogen Listeria monocytogenes induces CCR2-dependent monocyte recruitment and activation, an essential response for host survival. Herein we show that invasive L. monocytogenes, but not killed or noninvasive bacteria, induce secretion of MCP-1, the requisite chemokine for monocyte recruitment. Induction of MCP-1, but not TNF or IL-12, following L. monocytogenes infection is MyD88 independent. Consistent with these results, MyD88 deficiency does not impair monocyte recruitment to L. monocytogenes infected spleens, but prevents monocyte activation. Our results indicate that distinct microbial signals activate innate immune responses in an ordered, step-wise fashion, providing a mechanism to specify and modulate antimicrobial effector functions.


Asunto(s)
Antígenos de Diferenciación/inmunología , Inmunidad Innata , Listeriosis/inmunología , Receptores Inmunológicos/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Células Dendríticas/inmunología , Macrófagos/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide , Bazo/inmunología
20.
Science ; 301(5641): 1856-7, 2003 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-14512609

RESUMEN

Dendritic cells in the skin, called Langerhans cells, are the obvious candidates for transporting antigen from the epidermis to the lymph nodes and for activating T cell defense. However, as Serbina and Pamer discuss in their Perspective, at least in the case of viral antigens, it seems that other dendritic cell subsets residing in the dermis are the most important players in T cell-mediated defense against viral infection of the skin.


Asunto(s)
Presentación de Antígeno , Células Dendríticas/inmunología , Herpes Simple/inmunología , Células de Langerhans/inmunología , Linfocitos T/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Células Epidérmicas , Epidermis/inmunología , Herpes Genital/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Listeriosis/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Monocitos/citología , Monocitos/inmunología , Simplexvirus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...