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.
Front Immunol ; 9: 2794, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619242

RESUMO

After infection with Trypanosoma cruzi, the etiologic agent of Chagas disease, immunosuppression, and apoptosis of mature lymphocytes contribute to the establishment of the parasite in the host and thereby to persistence and pathology in the chronic stage of infection. In a systemic mouse model of experimental Chagas disease, we have demonstrated a strong depletion of mature B cells in the spleen during the first 2 weeks of infection. Remarkably, the decrease in this cell population commenced already in the bone marrow from infected mice and was a concomitant of an increased apoptosis in pro- and pre-B cell populations. Pro- and pre-B cells in the bone marrow showed a significant reduction accompanied by a functional disturbance of bone marrow-derived stromal cells resulting in diminished levels of IL-7, an essential factor for the development of B cell precursors. Ex vivo, stromal cells isolated from the bone marrow of infected mice had a strikingly impaired capacity to maintain the development of pro- and pre-B cells obtained from uninfected animals. Together, the reduction of an active humoral immune response during acute Chagas disease suggests to be an initial immune evasion mechanism of the parasite to establish persistent infection. Therefore, prevention of B cell depletion by rescuing the stromal cells during this early phase, could give rise to new therapeutic approaches.


Assuntos
Células da Medula Óssea/imunologia , Doença de Chagas/imunologia , Tolerância Imunológica , Trypanosoma cruzi/imunologia , Doença Aguda , Animais , Apoptose/imunologia , Células da Medula Óssea/parasitologia , Células da Medula Óssea/patologia , Doença de Chagas/parasitologia , Doença de Chagas/patologia , Doença Crônica , Interleucina-7/imunologia , Camundongos , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/parasitologia , Células Precursoras de Linfócitos B/patologia , Baço/imunologia , Baço/parasitologia , Baço/patologia , Células Estromais/imunologia , Células Estromais/parasitologia , Células Estromais/patologia
2.
Malar J ; 8: 54, 2009 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-19341445

RESUMO

BACKGROUND: To date all efforts to develop a malaria vaccine have failed, reflecting the still fragmentary knowledge about protective mechanisms against malaria. In order to evaluate if vaccination changes responses of the anti-malaria effectors spleen and liver to blood stage malaria, BALB/c mice succumbing to infection with Plasmodium chabaudi were compared to those surviving after vaccination. METHODS: Mice were vaccinated with host cell plasma membranes isolated from P. chabaudi-infected erythrocytes. Hepatic and splenic capacity to trap particulate material was determined after injection of fluorescent polystyrol beads. Hepatic gene expression was measured using real-time RT-PCR and Northern blotting. RESULTS: Survival of BALB/c mice was raised from 0% to 80% and peak parasitaemia was decreased by about 30% by vaccination. Vaccination boosted particle trapping capacity of the liver during crisis when splenic trapping is minimal due to spleen 'closing'. It also attenuated malaria-induced inflammation, thus diminishing severe damages and hence liver failure. Vaccination increased hepatic IFN-gamma production but mitigated acute phase response. Vaccination has a complex influence on infection-induced changes in expression of hepatic nuclear receptors (CAR, FXR, RXR, and PXR) and of the metabolic enzymes Sult2a and Cyp7a1. Although vaccination decreased CAR mRNA levels and prevented Cyp7a1 suppression by the CAR ligand 1,2-bis [2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) on day 8 p.i., Sult2a-induction by TCPOBOP was restored. CONCLUSION: These data support the view that the liver is an essential effector site for a vaccine against blood stage malaria: vaccination attenuates malaria-induced inflammation thus improving hepatic metabolic activity and particle trapping activity of the liver.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Parasitemia/imunologia , Plasmodium chabaudi/imunologia , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Northern Blotting , DNA de Protozoário/genética , DNA de Protozoário/imunologia , Membrana Eritrocítica/imunologia , Citometria de Fluxo , Hepatite Animal , Fígado/imunologia , Fígado/metabolismo , Malária/imunologia , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Parasitemia/genética , Plasmodium chabaudi/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/imunologia , Vacinação , Vacinas de DNA/genética , Vacinas de DNA/imunologia
3.
Eur J Immunol ; 38(3): 680-94, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18266299

RESUMO

The role of Toll-like receptors (TLR) and MyD88 for immune responses to Mycobacterium tuberculosis (Mtb) infection remains controversial. To address the impact of TLR-mediated pathogen recognition and MyD88-dependent signaling events on anti-mycobacterial host responses, we analyzed the outcome of Mtb infection in TLR2/4/9 triple- and MyD88-deficient mice. After aerosol infection, both TLR2/4/9-deficient and wild-type mice expressed pro-inflammatory cytokines promoting antigen-specific T cells and the production of IFN-gamma to similar extents. Moreover, TLR2/4/9-deficient mice expressed IFN-gamma-dependent inducible nitric oxide synthase and LRG-47 in infected lungs. MyD88-deficient mice expressed pro-inflammatory cytokines and were shown to expand IFN-gamma-producing antigen-specific T cells, albeit in a delayed fashion. Only mice that were deficient for MyD88 rapidly succumbed to unrestrained mycobacterial growth, whereas TLR2/4/9-deficient mice controlled Mtb replication. IFN-gamma-dependent restriction of mycobacterial growth was severely impaired only in Mtb-infected MyD88, but not in TLR2/4/9-deficient bone marrow-derived macrophages. Our results demonstrate that after Mtb infection neither TLR2, -4, and -9, nor MyD88 are required for the induction of adaptive T cell responses. Rather, MyD88, but not TLR2, TLR4 and TLR9, is critical for triggering macrophage effector mechanisms central to anti-mycobacterial defense.


Assuntos
Mycobacterium tuberculosis/imunologia , Fator 88 de Diferenciação Mieloide/fisiologia , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Receptor Toll-Like 9/fisiologia , Tuberculose/imunologia , Estruturas Animais/microbiologia , Animais , Citocinas/genética , Citocinas/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica/efeitos dos fármacos , Imunidade Celular/genética , Imunidade Celular/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , Interferon gama/metabolismo , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/agonistas , Fator 88 de Diferenciação Mieloide/genética , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/genética , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/genética , Tuberculose/genética , Tuberculose/microbiologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
4.
Immunobiology ; 210(2-4): 217-27, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16164029

RESUMO

Produced by macrophages and dendritic cells, interleukin (IL)-12 is composed of a p35 and a p40 subunit and promotes protection against intracellular pathogens through the development of interferon-gamma (IFNgamma) -producing T cells. The p40 subunit is also shared by the dimeric cytokines IL-12p40 homodimer and IL-23. In man, genetic defects in IL-12p40-mediated mechanisms are responsible for the familial occurrence of nontuberculous mycobacterial infections, the most common of which is infection with Mycobacterium avium. To experimentally differentiate the contribution of IL-12p40-containing cytokines in the outcome of M. avium infection, we studied wild-type, p35- and p35/p40 doubly deficient mice in an intravenous infection model which reflects many parameters of the disseminated infection in humans. Our study shows that in contrast to p35/p40 doubly deficient mice, p35-deficient mice mount a transient antibacterially protective response against M. avium although such animals were unable to produce detectable levels of IFNgamma or generate efficient granulomas. In conclusion, our results identify an antibacterial effector mechanism preserved in p35-deficient mice that is absent in mice devoid of p35 and p40. This phenotype probably reflects an IL-12p40-dependent effect on macrophage activation at the level of innate immunity.


Assuntos
Interleucina-12/imunologia , Infecções por Mycobacterium/imunologia , Mycobacterium avium/imunologia , Subunidades Proteicas/imunologia , Animais , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interleucina-12/deficiência , Interleucina-12/genética , Subunidade p40 da Interleucina-12 , Fígado/microbiologia , Fígado/patologia , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Camundongos , Fagócitos/imunologia , Subunidades Proteicas/genética , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia
5.
Infect Immun ; 73(10): 6390-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16177310

RESUMO

It is currently accepted that malaria-parasitized red blood cells (pRBC) are eliminated, like senescent erythrocytes, phagocytically by macrophages in the red pulp of the spleen. Here, however, we show that self-healing Plasmodium chabaudi malaria activates spleen closure in C57BL/6 mice. Confocal laser scanning microscopy revealed that spleen closing was manifested by elimination of entry into the red pulp of 3-microm polystyrol particles, pRBC, and nonparasitized red blood cells but not of bovine serum albumin. This spleen closure did not reflect a reduction in the number of phagocytic cells, as shown by flow cytometry, whereas marginal zone macrophages (MZM) were lost and red pulp macrophages entered the white pulp. Splenic trapping of pBRC was strongly reduced in the absence of MZM and marginal metallophilic macrophages (MMM), as it is in noninfected mice with a disrupted lymphotoxin beta receptor (LTbetaR(-/-)), and it was still significantly reduced when the number of MZM and MMM was diminished, as in tumor necrosis factor alpha-deficient (TNF-alpha(-/-)) mice. Moreover, mice deficient in TNF-alpha, tumor necrosis factor receptor I (TNFRI(-/-)), and LTbetaR exhibited progressive impairment in malaria-induced spleen closing. Treatment of C57BL/6 mice with TNF-alpha induced loss of MZM and spleen closing by about 20%. Our data indicate that TNF/TNFRI signaling is involved in regulating malaria-induced spleen closure, which is maximal during crisis, when parasitemia declines more than 100-fold. Consequently, the vast majority of pRBC cannot be destroyed by the spleen during crisis, suggesting that the known sophisticated sequestration system of Plasmodium parasites did not evolve to avoid splenic clearance.


Assuntos
Eritrócitos/parasitologia , Malária/imunologia , Parasitemia/imunologia , Plasmodium chabaudi , Baço/imunologia , Animais , Receptor beta de Linfotoxina , Macrófagos/imunologia , Malária/sangue , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Parasitemia/parasitologia , Parasitemia/patologia , Fagocitose , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Transdução de Sinais , Baço/irrigação sanguínea , Baço/citologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia
6.
Microbes Infect ; 7(3): 399-409, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15788153

RESUMO

Disrupted signaling through lymphotoxin beta receptor (LTbetaR) results in severe defects of the spleen and even loss of all other secondary lymphoid tissues, making mice susceptible to diverse infectious agents. Surprisingly, however, we find that female LTbetaR-deficient mice are even more resistant to blood stages of Plasmodium chabaudi malaria than wild-type C57BL/6 mice. Higher resistance of LTbetaR-deficient mice correlates with an earlier onset of reticulocytosis, and the period of anemia is shorter. After surviving fulminant parasitemias of about 35%, mice develop long-lasting protective immunity against homologous rechallenge, with both spleen and liver acting as anti-malaria effectors. Testosterone suppresses resistance, i.e. all mice succumb to infections during or shortly after peak parasitemia. At peak parasitemia, testosterone does not essentially affect cellularity and apoptosis in the spleen, but aggravates liver pathology in terms of increased cell swelling, numbers of apoptotic and binucleated cells and reduced serum alkaline phosphatase levels, and conversely, reduces inflammatory lymphocytic infiltrates in the liver. In the spleen, hybridization of cDNA arrays identified only a few testosterone-induced changes in gene expression, in particular upregulation of INFgamma and IFN-regulated genes. By contrast, a much larger number of testosterone-affectable genes was observed in the liver, including genes involved in regulation of the extracellular matrix, in chemokine and cytokine signaling, and in cell cycle control. Collectively, our data suggest that testosterone dysregulates the inflammatory response in spleen and liver during their differentiation to anti-malaria effectors in malaria-resistant female LTbetaR-deficient mice, thus contributing to the testosterone-induced lethal outcome of malaria.


Assuntos
Fígado/efeitos dos fármacos , Malária/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Baço/efeitos dos fármacos , Testosterona/farmacologia , Animais , Feminino , Expressão Gênica/efeitos dos fármacos , Imunidade Inata/genética , Receptor beta de Linfotoxina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parasitemia/imunologia , Plasmodium chabaudi , Receptores do Fator de Necrose Tumoral/genética , Fatores de Tempo
7.
Infect Immun ; 73(1): 436-43, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15618182

RESUMO

Testosterone induces a lethal outcome in otherwise self-healing blood-stage malaria caused by Plasmodium chabaudi. Here, we examine possible testosterone effects on the antimalaria effectors spleen and liver in female C57BL/6 mice. Self-healing malaria activates gating mechanisms in the spleen and liver that lead to a dramatic reduction in trapping activity, as measured by quantifying the uptake of 3-mum-diameter fluorescent polystyrol particles. However, testosterone delays malaria-induced closing of the liver, but not the spleen. Coincidently, testosterone causes an approximately 3- to 28-fold depression of the mRNA levels of nine malaria-responsive genes, out of 299 genes tested, only in the liver and not in the spleen, as shown by cDNA arrays and Northern blotting. Among these are the genes encoding plasminogen activator inhibitor (PAI1) and hydroxysteroid sulfotransferase (STA2). STA2, which detoxifies bile acids, is suppressed 10-fold by malaria and an additional 28-fold by testosterone, suggesting a severe perturbation of bile acid metabolism. PAI1 is protective against malaria, since disruption of the PAI1 gene results in partial loss of the ability to control the course of P. chabaudi infections. Collectively, our data indicate that the liver rather than the spleen is a major target organ for testosterone-mediated suppression of resistance against blood-stage malaria.


Assuntos
Imunossupressores/farmacologia , Fígado/imunologia , Malária/imunologia , Parasitemia/imunologia , Testosterona/farmacologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Baço/imunologia , Sulfotransferases/genética , Sulfotransferases/fisiologia
8.
Immunity ; 20(5): 623-35, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15142530

RESUMO

Macrophage/neutrophil-specific IL-4 receptor alpha-deficient mice (LysM(Cre)IL-4Ralpha(-/flox)) were generated to understand the role of IL-4/IL-13 responsive myeloid cells during Type 2 immune responses. LysM(Cre)IL-4Ralpha(-/flox) mice developed protective immunity against Nippostrongylus brasiliensis accompanied by T(H)2 development and goblet cell hyperplasia. In contrast, LysM(Cre)IL-4Ralpha(-/flox) mice were extremely susceptible to Schistosoma mansoni infection with 100% mortality during acute infection. Mortality was not dependent on neutrophils and occurred in the presence of T(H)2/Type 2 responses, granuloma formation, and egg-induced fibrosis. Death was associated with increased T(H)1 cytokines, hepatic and intestinal histopathology, increased NOS-2 activity, impaired egg expulsion, and sepsis. IL-10 was not able to compensate for the absence of IL-4/IL-13-activated alternative macrophages. Together, this shows that alternative macrophages are essential during schistosomiasis for protection against organ injury through downregulation of egg-induced inflammation.


Assuntos
Interleucina-4/imunologia , Ativação de Macrófagos/imunologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/patologia , Células Th1/imunologia , Animais , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interleucina-13/imunologia , Intestinos/patologia , Fígado/parasitologia , Fígado/patologia , Camundongos , Neutrófilos/imunologia , Receptores de Interleucina-4/deficiência , Receptores de Interleucina-4/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th2/imunologia
9.
Eur J Immunol ; 33(6): 1697-706, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12778488

RESUMO

During priming, naive CD4(+) Th cells differentiate into cells that produce either IFN-gamma or IL-4. Even though the cascade of pathways that induces IL-4-producing Th2 cells has been determined in vitro, the signals promoting Th2 differentiation under physiological conditions remain enigmatic, especially the natural role of the single most important Th2-inducing signal,IL-4. Using Th2 and naive Th cells, each expressing a distinct transgenic TCR, here we show that Th2 cells migrate with the same dynamics as naive Th cells in draining lymph nodes and bind to the same DC, when driven by antigen in complete Freund's adjuvant (CFA). Th2-cell-derived IL-4 deviates CFA-induced Th1 development toward a Th2 phenotype, if both cell populations co-localize in the same T cell area, and are activated simultaneously. Thus, intranodal Th2 cells directly influence Th cell differentiation in vivo, but only under restricted conditions. These findings have implications for the design of cytokine-based therapies and explain the spreading of Th2 responses to multiple aeroallergens in allergic asthma, where naive Th and Th2 cells co-localize in lung-draining lymph nodes.


Assuntos
Células Dendríticas/imunologia , Células Th2/imunologia , Sequência de Aminoácidos , Animais , Comunicação Celular , Diferenciação Celular , Movimento Celular , Adjuvante de Freund , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Memória Imunológica/imunologia , Interferon gama/biossíntese , Interleucina-4/biossíntese , Interleucina-4/fisiologia , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Modelos Imunológicos , Dados de Sequência Molecular , Ovalbumina/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Células Th1/citologia , Células Th2/metabolismo
10.
Eur J Immunol ; 33(1): 70-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12594834

RESUMO

CTL and NK cells are critical for resistance to acute Trypanosoma cruzi infection, but are also implicated in the pathology induced by this intracellular protozoan parasite. Here we explore to what extent the two main cytolytic pathways of CTL and NK cells, i.e. the granule exocytosis and the Fas ligand (FasL)/Fas pathways, are responsible for the elimination of parasites from mouse tissues and control of organ pathology. For this purpose we have employed mouse strains with targeted gene defects in one or more components - including perforin, granzyme A and granzyme B, and Fas - of either of the two cytolytic pathways, and we used the highly pathogenic T. cruzi strain Tulahuen. We show that parasites are effectively cleared from infected tissues independently of the FasL/Fas pathway by the concerted action of perforin and the two granzymes. However, prevention of pathology and early host death is critically dependent in addition on an operational FasL/Fas interaction. Thus, in contrast to C57BL/6 (B6) wild-type mice, mouse strains with deficiencies in either the FasL/Fas or the perforin/granzyme pathway similarly suffer from early death, independently of their differential capacity to control parasite growth; this finding indicates that the two cytolytic pathways control distinct but vital processes during infection with T. cruzi.


Assuntos
Glicoproteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Trypanosoma cruzi/imunologia , Tripanossomíase/imunologia , Tripanossomíase/metabolismo , Receptor fas/metabolismo , Animais , Proteína Ligante Fas , Citometria de Fluxo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Perforina , Proteínas Citotóxicas Formadoras de Poros , Baço/patologia , Tripanossomíase/enzimologia , Fator de Necrose Tumoral alfa/metabolismo
11.
Steroids ; 67(6): 535-8, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11960632

RESUMO

This review summarizes data about non-genomic actions of testosterone on murine malaria, T cells and macrophages produced by our group during the last 15 years. In C57BL/10 mice, testosterone induces a lethal outcome of blood stage infections with Plasmodium chabaudi which normally takes a self-healing course controlled by genes of the H-2 complex and the non-H-2 background. This suppressive effect of testosterone is mediated neither via the classic intracellular androgen receptor (AR) response nor, after conversion of testosterone to estradiol, via the estrogen receptor. Testosterone acts non-genomically, i.e. through surface receptors, on murine T cells and macrophages, which becomes evident as a rapid rise in the intracellular free Ca(2+) concentration ([Ca(2+)](i)). In T cells, this rise reflects predominantly influx of extracellular Ca(2+), while it is predominantly due to release of Ca(2+) from intracellular Ca(2+)-stores in macrophages. The testosterone-induced rise in [Ca(2+)](i) of both macrophages and T cells is not inhibited by the AR-blocker cyproterone, and it is also inducible by the plasma membrane impermeable ligand testosterone-BSA. The surface receptors initiate a transcription-independent signaling pathway of testosterone. Currently, we are trying to isolate testosterone surface receptors and to investigate a possible cross-talk of non-genomic testosterone signaling with other genotropic signaling pathways.


Assuntos
Macrófagos/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Testosterona/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Macrófagos/metabolismo , Malária/induzido quimicamente , Malária/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Androgênicos/deficiência , Receptores Androgênicos/metabolismo , Transdução de Sinais/fisiologia , Linfócitos T/metabolismo
12.
Methods Microbiol ; 32: 183-231, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-38620305

RESUMO

This chapter discusses the management of immunocompromised and infected animals. The microbiological quality of laboratory animals is a direct result of colony management practices and monitoring provides an after-the-fact assessment of the adequacy of those practices. Monitoring is, therefore, of greatest value in connection with the maintenance of animals in isolation systems where vigorous microbiological control is applied. In addition to constructive measures, an appropriate management system is necessary for the prevention of infections, as well as for their detection and control. It is a major task for the management of an animal facility to understand the way micro-organisms might be introduced or spread under the specific conditions given. The management of all animal facilities in an institution is best centralized. This warrants that all information dealing with the purchase of animals, the use of experimental materials and equipment and the performance of animal experiments flows through one office. This reduces the opportunity for the failures of communication. Centralized management can best establish comprehensive monitoring programs to evaluate important risk factors, such as animals and biological materials, before they are introduced into a facility.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...