Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 247
Filtrar
1.
mSphere ; 6(3)2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33952667

RESUMEN

Prior work has shown that parenterally administered anti-CD20 (5D2) inhibits CD4+ T cell priming in response to challenge with Pneumocystis murina and predisposes to pneumonia. In this study, we investigated the effect of subcutaneous anti-CD20 antibody and Pneumocystis infection. In mice with primary infection, anti-CD20 antibody treatment depleted both CD19+ and CD27+ CD19+ cells but not T cells in the lung at days 14 and 28 after Pneumocystis inoculation. Although anti-CD20 antibody treatment impaired fungal clearance at day 14 postinfection, fungal burden in the lungs was substantially reduced at day 28 in both depleted and control mice in the low-dose group. Subcutaneous anti-CD20 antibody treatment did not alter antigen-specific serum immunoglobulin levels in mice compared with control mice, and there were no significant differences in the numbers of lung gamma interferon-positive (IFN-γ+) CD4+, interleukin 4-positive (IL-4+) CD4+, IL-5+ CD4+, and IL-17A+ CD4+ cells between depleted and control mice after infection. In mice with secondary infection, the lung fungal burden was comparable between depleted and control mice 14 days after reinfection. Low-dose subcutaneous anti-CD20 antibody treatment may delay fungal clearance, but it did not impair the ability of the host to clear Pneumocystis infection, irrespective of primary or secondary infection.IMPORTANCE Anti-CD20 antibody therapy is used for both cancer and autoimmune disease but has been shown to be associated with Pneumocystis pneumonia in humans. This study shows that low-dose subcutaneous anti-CD20 can modulate B cell populations without grossly perturbing fungal immunity against Pneumocystis lung infection.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD20/inmunología , Linfocitos B/inmunología , Pulmón/microbiología , Pneumocystis/inmunología , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/terapia , Animales , Linfocitos B/efectos de los fármacos , Inyecciones Subcutáneas , Pulmón/efectos de los fármacos , Pulmón/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Pneumocystis/efectos de los fármacos
3.
Bioorg Med Chem Lett ; 29(15): 1874-1880, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31176699

RESUMEN

Pneumocystis pneumonia (PCP) caused by Pneumocystis jirovecii (pj) can lead to serious health consequences in patients with an immunocompromised system. Trimethoprim (TMP), used as first-line therapy in combination with sulfamethoxazole, is a selective but only moderately potent pj dihydrofolate reductase (pjDHFR) inhibitor, whereas non-clinical pjDHFR inhibitors, such as, piritrexim and trimetrexate are potent but non-selective pjDHFR inhibitors. To meet the clinical needs for a potent and selective pjDHFR inhibitor for PCP treatment, fourteen 6-substituted pyrido[3,2-d]pyrimidines were developed. Comparison of the amino acid residues in the active site of pjDHFR and human DHFR (hDHFR) revealed prominent amino acid differences which could be exploited to structurally design potent and selective pjDHFR inhibitors. Molecular modeling followed by enzyme assays of the compounds revealed 15 as the best compound of the series with an IC50 of 80 nM and 28-fold selectivity for inhibiting pjDHFR over hDHFR. Compound 15 serves as the lead analog for further structural variations to afford more potent and selective pjDHFR inhibitors.


Asunto(s)
Antagonistas del Ácido Fólico/uso terapéutico , Pneumocystis carinii/patogenicidad , Pneumocystis/efectos de los fármacos , Pirimidinas/uso terapéutico , Trimetoprim/uso terapéutico , Antagonistas del Ácido Fólico/farmacología , Humanos , Modelos Moleculares , Pirimidinas/farmacología , Relación Estructura-Actividad , Trimetoprim/farmacología
4.
PLoS One ; 14(6): e0217684, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31170201

RESUMEN

Clara cells are the main airway secretory cells able to regenerate epithelium in the distal airways through transdifferentiating into goblet cells, a process under negative regulation of the Notch pathway. Pneumocystis is a highly prevalent fungus in humans occurring between 2 and 5 months of age, a period when airways are still developing and respiratory morbidity typically increases. Pneumocystis induces mucus hyperproduction in immunocompetent host airways and whether it can stimulate Clara cells is unknown. Markers of Clara cell secretion and Notch1 activation were investigated in lungs of immunocompetent rats at 40, 60, and 80 days of age during Pneumocystis primary infection with and without Valproic acid (VPA), a Notch inducer. The proportion of rats expressing mucin increased in Pneumocystis-infected rats respect to controls at 60 and 80 days of age. Frequency of distal airways Clara cells was maintained while mRNA levels for the mucin-encoding genes Muc5B and Muc5ac in lung homogenates increased 1.9 and 3.9 times at 60 days of infection (P. = 0.1609 and P. = 0.0001, respectively) and protein levels of the Clara cell marker CC10 decreased in the Pneumocystis-infected rats at 60 and 80 days of age (P. = 0.0118 & P. = 0.0388). CC10 and Muc5b co-localized in distal airway epithelium of Pneumocystis-infected rats at day 60. Co-localization of Muc5b and Ki67 as marker of mitosis in distal airways was not observed suggesting that Muc5b production by Clara cells was independent of mitosis. Notch levels remained similar and no transnucleation of activated Notch associated to Pneumocystis infection was detected. Unexpectedly, mucus was greatly increased at day 80 in Pneumocystis-infected rats receiving VPA suggesting that a Notch-independent mechanism was triggered. Overall, data suggests a Clara to goblet cell transdifferentiation mechanism induced by Pneumocystis and independent of Notch.


Asunto(s)
Pulmón/metabolismo , Pulmón/microbiología , Mucina 5AC/biosíntesis , Mucina 5B/biosíntesis , Infecciones por Pneumocystis/metabolismo , Infecciones por Pneumocystis/microbiología , Pneumocystis/patogenicidad , Receptores Notch/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Antígeno Ki-67/metabolismo , Mitosis/efectos de los fármacos , Mucina 5AC/genética , Mucina 5AC/metabolismo , Mucina 5B/genética , Mucina 5B/metabolismo , Pneumocystis/efectos de los fármacos , Infecciones por Pneumocystis/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Uteroglobina/metabolismo , Ácido Valproico/farmacología
5.
Artículo en Inglés | MEDLINE | ID: mdl-30917977

RESUMEN

An urgent need exists for new antifungal compounds to treat fungal infections in immunocompromised patients. The aim of the current study was to investigate the potency of a novel antifungal compound, MYC-053, against the emerging yeast and yeast-like pathogens Candida glabrata, Candida auris, Cryptococcus neoformans, and Pneumocystis species. MYC-053 was equally effective against the susceptible control strains, clinical isolates, and resistant strains, with MICs of 0.125 to 4.0 µg/ml. Notably, unlike other antifungals such as azoles, polyenes, and echinocandins, MYC-053 was effective against Pneumocystis isolates, therefore being the only synthetic antifungal that may potentially be used against Pneumocystis spp., Candida spp., and Cryptococcus spp. MYC-053 was highly effective against preformed 48-h-old C. glabrata and C. neoformans biofilms, with minimal biofilm eradication concentrations equal to 1 to 4 times the MIC. Together, these data indicated that MYC-053 may be developed into a promising antifungal agent for the treatment and prevention of invasive fungal infections caused by yeasts and yeast-like fungi.


Asunto(s)
Antifúngicos/farmacología , Pirimidinas/farmacología , Biopelículas/efectos de los fármacos , Candida/efectos de los fármacos , Candida glabrata/efectos de los fármacos , Cryptococcus neoformans/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Farmacorresistencia Fúngica/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Pneumocystis/efectos de los fármacos
6.
BMC Syst Biol ; 12(1): 77, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30016951

RESUMEN

BACKGROUND: The yeast-like fungi Pneumocystis, resides in lung alveoli and can cause a lethal infection known as Pneumocystis pneumonia (PCP) in hosts with impaired immune systems. Current therapies for PCP, such as trimethoprim-sulfamethoxazole (TMP-SMX), suffer from significant treatment failures and a multitude of serious side effects. Novel therapeutic approaches (i.e. newly developed drugs or novel combinations of available drugs) are needed to treat this potentially lethal opportunistic infection. Quantitative Systems Pharmacological (QSP) models promise to aid in the development of novel therapies by integrating available pharmacokinetic (PK) and pharmacodynamic (PD) knowledge to predict the effects of new treatment regimens. RESULTS: In this work, we constructed and independently validated PK modules of a number of drugs with available pharmacokinetic data. Characterized by simple structures and well constrained parameters, these PK modules could serve as a convenient tool to summarize and predict pharmacokinetic profiles. With the currently accepted hypotheses on the life stages of Pneumocystis, we also constructed a PD module to describe the proliferation, transformation, and death of Pneumocystis. By integrating the PK module and the PD module, the QSP model was constrained with observed levels of asci and trophic forms following treatments with multiple drugs. Furthermore, the temporal dynamics of the QSP model were validated with corresponding data. CONCLUSIONS: We developed and validated a QSP model that integrates available data and promises to facilitate the design of future therapies against PCP.


Asunto(s)
Antifúngicos/farmacología , Antifúngicos/farmacocinética , Modelos Biológicos , Pneumocystis/efectos de los fármacos , Animales , Ratones , Distribución Tisular
7.
Artículo en Inglés | MEDLINE | ID: mdl-29463544

RESUMEN

The echinocandins are a class of antifungal agents that target ß-1,3-d-glucan (BG) biosynthesis. In the ascigerous Pneumocystis species, treatment with these drugs depletes the ascus life cycle stage, which contains BG, but large numbers of forms which do not express BG remain in the infected lungs. In the present study, the gene expression profiles of Pneumocystis murina were compared between infected, untreated mice and mice treated with anidulafungin for 2 weeks to understand the metabolism of the persisting forms. Almost 80 genes were significantly up- or downregulated. Like other fungi exposed to echinocandins, genes associated with sexual replication, cell wall integrity, cell cycle arrest, and stress comprised the strongest upregulated signals in P. murina from the treated mice. The upregulation of the P. murina ß-1,3-d-glucan endohydrolase and endo-1,3-glucanase was notable and may explain the disappearance of the existing asci in the lungs of treated mice since both enzymes can degrade BG. The biochemical measurement of BG in the lungs of treated mice and fluorescence microscopy with an anti-BG antibody supported the loss of BG. Downregulated signals included genes involved in cell replication, genome stability, and ribosomal biogenesis and function and the Pneumocystis-specific genes encoding the major surface glycoproteins (Msg). These studies suggest that P. murina attempted to undergo sexual replication in response to a stressed environment and was halted in any type of proliferative cycle, likely due to a lack of BG. Asci appear to be a required part of the life cycle stage of Pneumocystis, and BG may be needed to facilitate progression through the life cycle via sexual replication.


Asunto(s)
Anidulafungina/farmacología , Antifúngicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Pared Celular/efectos de los fármacos , Pneumocystis/efectos de los fármacos , Pneumocystis/genética , Animales , Proliferación Celular/efectos de los fármacos , Masculino , Ratones , Pneumocystis/patogenicidad , Neumonía por Pneumocystis/tratamiento farmacológico , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/prevención & control
8.
Artículo en Inglés | MEDLINE | ID: mdl-28760906

RESUMEN

The combination of trimethoprim and sulfamethoxazole (TMP-SMX) is the most effective regimen for therapy of Pneumocystis pneumonia (PCP). As many patients with PCP are allergic or do not respond to it, efforts have been devoted to develop alternative therapies for PCP. We have found that the combination of vitamin D3 (VitD3) (300 IU/kg/day) and primaquine (PMQ) (5 mg/kg/day) was as effective as TMP-SMX for therapy of PCP. In this study, we investigated the mechanisms by which vitamin D enhances the efficacy of PMQ. C57BL/6 mice were immunosuppressed by CD4+ cell depletion, infected with Pneumocystismurina for 8 weeks, and then treated for 9 days with the combination of VitD3 and PMQ (VitD3-PMQ) or with TMP-SMX or PMQ to serve as controls. The results showed that vitamin D supplementation increased the number of CD11c+ cells, suppressed the production of proinflammatory cytokines (tumor necrosis factor alpha [TNF-α], gamma interferon [IFN-γ], and interleukin-6 [IL-6]) and inducible nitric oxide synthase (iNOS), and enhanced the expression of genes related to antioxidation (glutathione reductase and glutamate-cysteine ligase modifier subunit), antimicrobial peptides (cathelicidin), and autophagy (ATG5 and beclin-1). These results suggest that the main action of vitamin D is enhancing the ability of the host to defend against Pneumocystis infection.


Asunto(s)
Antibacterianos/uso terapéutico , Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/tratamiento farmacológico , Primaquina/uso terapéutico , Vitamina D/uso terapéutico , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Proteína 5 Relacionada con la Autofagia/biosíntesis , Beclina-1/biosíntesis , Sinergismo Farmacológico , Femenino , Glutamato-Cisteína Ligasa/biosíntesis , Glutatión Reductasa/biosíntesis , Humanos , Interferón gamma/biosíntesis , Interleucina-6/biosíntesis , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Neumonía por Pneumocystis/microbiología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Factor de Necrosis Tumoral alfa/biosíntesis , Catelicidinas
9.
Am J Respir Cell Mol Biol ; 56(2): 213-222, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27632412

RESUMEN

N-acetylglucosamine (GlcNAc) serves as an essential structural sugar on the cell surface of organisms. For example, GlcNAc is a major component of bacterial peptidoglycan, it is an important building block of fungal cell walls, including a major constituent of chitin and mannoproteins, and it is also required for extracellular matrix generation by animal cells. Herein, we provide evidence for a uridine diphospho (UDP)-GlcNAc pathway in Pneumocystis species. Using an in silico search of the Pneumocystis jirovecii and P. murina (Pm) genomic databases, we determined the presence of at least four proteins implicated in the Saccharomyces cerevisiae UDP-GlcNAc biosynthetic pathway. These genes, termed GFA1, GNA1, AGM1, and UDP-GlcNAc pyrophosphorylase (UAP1), were either confirmed to be present in the Pneumocystis genomes by PCR, or, in the case of Pm uap1 (Pmuap1), functionally confirmed by direct enzymatic activity assay. Expression analysis using quantitative PCR of Pneumocystis pneumonia in mice demonstrated abundant expression of the Pm uap1 transcript. A GlcNAc-binding recombinant protein and a novel GlcNAc-binding immune detection method both verified the presence of GlcNAc in P. carinii (Pc) lysates. Studies of Pc cell wall fractions using high-performance gas chromatography/mass spectrometry documented the presence of GlcNAc glycosyl residues. Pc was shown to synthesize GlcNAc in vitro. The competitive UDP-GlcNAc substrate synthetic inhibitor, nikkomycin Z, suppressed incorporation of GlcNAc by Pc preparations. Finally, treatment of rats with Pneumocystis pneumonia using nikkomycin Z significantly reduced organism burdens. Taken together, these data support an important role for GlcNAc generation in the cell surface of Pneumocystis organisms.


Asunto(s)
Acetilglucosamina/biosíntesis , Terapia Molecular Dirigida , Pneumocystis/metabolismo , Aminoglicósidos/farmacología , Animales , Vías Biosintéticas/efectos de los fármacos , Vías Biosintéticas/genética , Western Blotting , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Genes Fúngicos , Lectinas/metabolismo , Ratones , Pneumocystis/efectos de los fármacos , Pneumocystis/genética , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
10.
Antimicrob Agents Chemother ; 60(12): 7115-7127, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27645246

RESUMEN

Only one new class of antifungal drugs has been introduced into clinical practice in the last 30 years, and thus the identification of small molecules with novel mechanisms of action is an important goal of current anti-infective research. Here, we describe the characterization of the spectrum of in vitro activity and in vivo activity of AR-12, a celecoxib derivative which has been tested in a phase I clinical trial as an anticancer agent. AR-12 inhibits fungal acetyl coenzyme A (acetyl-CoA) synthetase in vitro and is fungicidal at concentrations similar to those achieved in human plasma. AR-12 has a broad spectrum of activity, including activity against yeasts (e.g., Candida albicans, non-albicans Candida spp., Cryptococcus neoformans), molds (e.g., Fusarium, Mucor), and dimorphic fungi (Blastomyces, Histoplasma, and Coccidioides) with MICs of 2 to 4 µg/ml. AR-12 is also active against azole- and echinocandin-resistant Candida isolates, and subinhibitory AR-12 concentrations increase the susceptibility of fluconazole- and echinocandin-resistant Candida isolates. Finally, AR-12 also increases the activity of fluconazole in a murine model of cryptococcosis. Taken together, these data indicate that AR-12 represents a promising class of small molecules with broad-spectrum antifungal activity.


Asunto(s)
Antifúngicos/farmacología , Criptococosis/tratamiento farmacológico , Fluconazol/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Candida/efectos de los fármacos , Candida/genética , Caspofungina , Celecoxib/química , Cryptococcus neoformans/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Farmacorresistencia Fúngica/efectos de los fármacos , Sinergismo Farmacológico , Equinocandinas/farmacología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Lipopéptidos/farmacología , Masculino , Ratones Endogámicos , Pruebas de Sensibilidad Microbiana , Pneumocystis/efectos de los fármacos , Pirazoles/química , Saccharomyces cerevisiae/efectos de los fármacos , Sulfonamidas/química
12.
PLoS One ; 8(1): e53479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23308231

RESUMEN

Pneumocystis pneumonia (PcP) develops in immunocompromised patients. Alveolar macrophages play a key role in the recognition, phagocytosis, and degradation of Pneumocystis, but their number is decreased in PcP. Our study of various inflammatory components during PcP found that myeloid-derived suppressor cells (MDSCs) accumulate in the lungs of mice and rats with Pneumocystis pneumonia (PcP). We hypothesized that treatment with all-trans retinoic acid (ATRA), a metabolite of vitamin A, may effectively control Pneumocystis (Pc) infection by inducing MDSCs to differentiate to AMs. In rodent models of PcP, we found that 5 weeks of ATRA treatment reduced the number of MDSCs in the lungs and increased the number of AMs which cleared Pc infection. We also found that ATRA in combination with primaquine was as effective as the combination of trimethoprim and sulfamethaxazole for treatment of PcP and completely eliminated MDSCs and Pc organisms in the lungs in two weeks. No relapse of PcP was seen after three weeks of the ATRA-primaquine combination treatment. Prolonged survival of Pc-infected animals was also achieved by this regimen. This is the very first successful development of a therapeutic regimen for PcP that combines an immune modulator with an antibiotic, enabling the hosts to effectively defend the infection. Results of our study may serve as a model for development of novel therapies for other infections with MDSC accumulation.


Asunto(s)
Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/tratamiento farmacológico , Primaquina/farmacología , Tretinoina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Femenino , Humanos , Huésped Inmunocomprometido , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos BALB C , Pneumocystis/fisiología , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/mortalidad , Ratas , Ratas Sprague-Dawley , Análisis de Supervivencia , Combinación Trimetoprim y Sulfametoxazol/farmacología
13.
Parasite ; 18(1): 3-11, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21395200

RESUMEN

Pneumocystis pneumonia (PcP) remains a significant cause of morbidity and mortality in immunocompromised persons, especially those with human immunodeficiency virus (HIV) infection. Pneumocystis colonization is described increasingly in a wide range of immunocompromised and immunocompetent populations and associations between Pneumocystis colonization and significant pulmonary diseases such as chronic obstructive pulmonary disease (COPD) have emerged. This mini-review summarizes recent advances in our clinical understanding of Pneumocystis and PcP, describes ongoing areas of clinical and translational research, and offers recommendations for future clinical research from researchers participating in the "First centenary of the Pneumocystis discovery".


Asunto(s)
Investigación Biomédica/tendencias , Pneumocystis , Neumonía por Pneumocystis , Investigación Biomédica Traslacional/tendencias , Infecciones Oportunistas Relacionadas con el SIDA/epidemiología , Infecciones Oportunistas Relacionadas con el SIDA/microbiología , Farmacorresistencia Fúngica , Humanos , Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/diagnóstico , Neumonía por Pneumocystis/epidemiología , Neumonía por Pneumocystis/terapia , Neumonía por Pneumocystis/transmisión
14.
Am J Respir Cell Mol Biol ; 44(4): 540-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20558778

RESUMEN

The effect of nitric oxide (NO) on Pneumocystis (Pc) organisms, the role of NO in the defense against infection with Pc, and the production of NO by alveolar macrophages (AMs) during Pneumocystis pneumonia (PCP) were investigated. The results indicate that NO was toxic to Pc organisms and inhibited their proliferation in culture. When the production of NO was inhibited by intraperitoneal injection of rats with the nitric oxide synthase inhibitor L-N(5)-(1-iminoethyl) ornithine, progression of Pc infection in immunocompetent rats was enhanced. Concentrations of NO in bronchoalveolar lavage fluids from immunosuppressed, Pc-infected rats and mice were greatly reduced, compared with those from uninfected animals, and AMs from these animals were defective in NO production. However, inducible nitric oxide synthase (iNOS) mRNA and protein concentrations were high in AMs from Pc-infected rats and mice. Immunoblot analysis showed that iNOS in AMs from Pc-infected rats existed primarily as a monomer, but the homo-dimerization of iNOS monomers was required for the production of NO. When iNOS dimerization cofactors, including calmodulin, were added to macrophage lysates, iNOS dimerization increased, whereas incubation of the same lysates with all cofactors except calmodulin did not rescue iNOS dimer formation. These data suggest that NO is important in the defense against Pc infection, but that the production of NO in AMs during PCP is defective because of the reduced dimerization of iNOS.


Asunto(s)
Macrófagos Alveolares/metabolismo , Óxido Nítrico/biosíntesis , Neumonía por Pneumocystis/metabolismo , Neumonía por Pneumocystis/patología , Animales , Líquido del Lavado Bronquioalveolar/citología , Extractos Celulares , Línea Celular , Proliferación Celular/efectos de los fármacos , Coenzimas/farmacología , Medios de Cultivo/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/microbiología , Ratones , Viabilidad Microbiana/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Ornitina/farmacología , Pneumocystis/citología , Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/enzimología , Multimerización de Proteína/efectos de los fármacos , Ratas , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
15.
Biochim Biophys Acta ; 1797(1): 38-43, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19660431

RESUMEN

Atovaquone is a substituted 2-hydroxy-naphthoquinone used therapeutically against Plasmodium falciparum (malaria) and Pneumocystis pathogens. It acts by inhibiting the cytochrome bc(1) complex via interactions with the Rieske iron-sulfur protein and cytochrome b in the ubiquinol oxidation pocket. As the targeted pathogens have developed resistance to this drug there is an urgent need for new alternatives. To better understand the determinants of inhibitor binding in the ubiquinol oxidation pocket of the bc(1) complex we synthesized a series of hydroxy-naphthoquinones bearing a methyl group on the benzene ring that is predicted to interact with the nuclear encoded Rieske iron-sulfur protein. We have also attempted to overcome the metabolic instability of a potent cytochrome bc(1) complex inhibitor, a 2-hydroxy-naphthoquinone with a branched side chain, by fluorinating the terminal methyl group. We have tested these new 2-hydroxy-naphthoquinones against yeast and bovine cytochrome bc(1) complexes to model the interaction with pathogen and human enzymes and determine parameters that affect efficacy of binding of these inhibitors. We identified a hydroxy-naphthoquinone with a trifluoromethyl function that has potential for development as an anti-fungal and anti-parasitic therapeutic.


Asunto(s)
Complejo III de Transporte de Electrones/química , Naftoquinonas/farmacología , Animales , Antimaláricos/química , Antimaláricos/uso terapéutico , Atovacuona/química , Atovacuona/uso terapéutico , Bovinos , Citocromos b/química , Citocromos b/efectos de los fármacos , Citocromos b/genética , Citocromos b/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Humanos , Cinética , Malaria/tratamiento farmacológico , Naftoquinonas/química , Plasmodium falciparum/efectos de los fármacos , Pneumocystis/efectos de los fármacos
16.
Bioorg Med Chem Lett ; 19(20): 5884-6, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19736009

RESUMEN

A series of alkanediamide-linked bisbenzamidines was synthesized and tested in vitro against a drug-sensitive strain of Trypanosoma brucei brucei, a drug-resistant strain of Trypanosoma brucei rhodesiense and Pneumocystiscarinii. Bisbenzamidines linked with longer alkanediamide chains were potent inhibitors of both strains of T. brucei. However, bisbenzamidines linked with shorter alkanediamide chains were the most potent compounds against P. carinii. N,N'-Bis[4-(aminoiminomethyl)phenyl] hexanediamide, 4 displayed potent inhibition (IC50=2-3 nM) against T. brucei and P. carinii, and was non-cytotoxic in the A549 human lung carcinoma cell line. The inhibitory bioactivity was significantly reduced when the amidine groups in 4 were moved from the para to the meta positions or replaced with amides.


Asunto(s)
Amidinas/síntesis química , Anilidas/síntesis química , Antiprotozoarios/síntesis química , Benzamidinas/síntesis química , Diamida/química , Amidinas/química , Amidinas/farmacología , Anilidas/química , Anilidas/farmacología , Animales , Antiprotozoarios/química , Antiprotozoarios/toxicidad , Benzamidinas/química , Benzamidinas/toxicidad , Línea Celular Tumoral , Humanos , Pneumocystis/efectos de los fármacos , Relación Estructura-Actividad , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei rhodesiense/efectos de los fármacos
17.
Curr Med Chem ; 16(20): 2514-30, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19601796

RESUMEN

Pneumocystis remains an important cause of fatal pneumonia (PCP) in HIV patients and other immunocompromised hosts. Preclinical drug discovery for agents active against PCP has been hindered in large part by the lack of a continuous in vitro growth system. Since approval in 1978, the combination of the folic acid synthesis inhibitor combination trimethoprim-sulfamethoxazole has been the primary agent for prophylaxis and therapy. Short term in vitro assays using cell monolayer-based and cell free systems in combination with in vivo studies in rodent models of infection have been the mainstay of candidate screening methods. These systems and their applications are reviewed here. Most strategies have focused on testing compounds already in clinical use, such as dapsone or atovaquone, for activity against Pneumocystis alone or in combination, and as parent compounds for chemical derivation, such as pentamidine and its analogues. Other successes from the bench include primaquine-clindamycin for moderate pneumonia and the family of Beta-glucan synthase inhibitors, which hold promise for clinical use against PCP. Despite the significant obstacles for drug discovery, progress in identifying novel agents has been made with current systems and the promise of future new targets is expected with the annotation of the Pneumocystis genome.


Asunto(s)
Antifúngicos/uso terapéutico , Descubrimiento de Drogas , Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/tratamiento farmacológico , Infecciones Oportunistas Relacionadas con el SIDA/tratamiento farmacológico , Infecciones Oportunistas Relacionadas con el SIDA/microbiología , Animales , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Humanos , Estadios del Ciclo de Vida , Pneumocystis/crecimiento & desarrollo , Neumonía por Pneumocystis/microbiología
18.
Transpl Infect Dis ; 11(4): 290-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19497072

RESUMEN

Mycophenolate mofetil (MMF) is one of the most frequently used immunosuppressive drugs in solid organ transplant recipients. MMF is an inhibitor of inosine-5'-monophosphate, and is able to preferentially inhibit B-cell and T-cell function. The immunosuppressive abilities of MMF have made it one of the most successful anti-rejection drugs in transplant patients, but patients also appear to have increased susceptibility to infections, specifically cytomegalovirus and BK virus. Despite its association with an increased risk of infection, MMF has also exhibited antimicrobial activity against pathogens including hepatitis C, Pneumocystis jirovecii, and human immunodeficiency virus. A thorough understanding of the functions of MMF on the immune system and interaction with infectious pathogens could be helpful in implementing preventative strategies against opportunistic infections in transplant patients.


Asunto(s)
Antifúngicos/farmacología , Antivirales/farmacología , Linfocitos B/efectos de los fármacos , Inmunosupresores/uso terapéutico , Ácido Micofenólico/análogos & derivados , Infecciones Oportunistas/prevención & control , Trasplante de Órganos/efectos adversos , Linfocitos T/efectos de los fármacos , Animales , Linfocitos B/inmunología , Ensayos Clínicos como Asunto , Rechazo de Injerto/prevención & control , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Ratones , Ácido Micofenólico/administración & dosificación , Ácido Micofenólico/farmacología , Ácido Micofenólico/uso terapéutico , Infecciones Oportunistas/microbiología , Infecciones Oportunistas/virología , Pneumocystis/efectos de los fármacos , Ratas , Linfocitos T/inmunología , Virus/efectos de los fármacos
20.
Nat Rev Microbiol ; 5(4): 298-308, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17363968

RESUMEN

The fungal infection Pneumocystis pneumonia is the most prevalent opportunistic infection in patients with AIDS. Although the analysis of this opportunistic fungal pathogen has been hindered by the inability to isolate it in pure culture, the use of molecular techniques and genomic analysis have brought insights into its complex cell biology. Analysis of the intricate relationship between Pneumocystis and the host lung during infection has revealed that the attachment of Pneumocystis to the alveolar epithelium promotes the transition of the organism from the trophic to the cyst form. It also revealed that Pneumocystis infection elicits the production of inflammatory mediators, culminating in lung injury and impaired gas exchange. Here we discuss these and other recent findings relating to the biology and pathogenesis of this intractable fungus.


Asunto(s)
Pneumocystis/fisiología , Neumonía por Pneumocystis , Animales , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Antígenos Fúngicos/análisis , Antígenos de Superficie/análisis , Ciclo Celular , Citocinas/metabolismo , Farmacorresistencia Fúngica , Genoma Fúngico , Humanos , Pulmón/microbiología , Pulmón/patología , Pneumocystis/citología , Pneumocystis/efectos de los fármacos , Pneumocystis/patogenicidad , Neumonía por Pneumocystis/tratamiento farmacológico , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/patología , Alveolos Pulmonares/microbiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...