Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Med Chem ; 67(5): 3935-3958, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38365209

RESUMO

As SARS-CoV-2 continues to circulate, antiviral treatments are needed to complement vaccines. The virus's main protease, 3CLPro, is an attractive drug target in part because it recognizes a unique cleavage site, which features a glutamine residue at the P1 position and is not utilized by human proteases. Herein, we report the invention of MK-7845, a novel reversible covalent 3CLPro inhibitor. While most covalent inhibitors of SARS-CoV-2 3CLPro reported to date contain an amide as a Gln mimic at P1, MK-7845 bears a difluorobutyl substituent at this position. SAR analysis and X-ray crystallographic studies indicate that this group interacts with His163, the same residue that forms a hydrogen bond with the amide substituents typically found at P1. In addition to promising in vivo efficacy and an acceptable projected human dose with unboosted pharmacokinetics, MK-7845 exhibits favorable properties for both solubility and absorption that may be attributable to the unusual difluorobutyl substituent.


Assuntos
COVID-19 , Glutamina , Humanos , Glutamina/química , SARS-CoV-2 , Cisteína Endopeptidases/química , Invenções , Inibidores de Proteases/farmacologia , Amidas , Antivirais/farmacologia , Antivirais/química
2.
Hum Vaccin Immunother ; 20(1): 2303226, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38251677

RESUMO

Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosae, with secretory form (sIgA) being dominant and uniquely stable. sIgA is challenging to produce recombinantly but is naturally found in human milk, which could be considered a global resource for this biologic, justifying its development as a mucosal therapeutic. Presently, SARS-CoV-2 was utilized as a model mucosal pathogen, and methods were developed to efficiently extract human milk sIgA from donors who were naïve to SARS-CoV-2 or had recovered from infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA in their milk (pooled to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1% or greater were all associated with sIgA. Western blot demonstrated that all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher Spike binding (mean endpoint of 0.87 versus 5.87). LCTG-002 was capable of blocking the Spike receptor-binding domain - angiotensin-converting enzyme 2 (ACE2) interaction with significantly greater potency compared to control (mean LCTG-002 IC50 154ug/mL versus 50% inhibition not achieved for control), and exhibited significant neutralization activity against Spike-pseudotyped virus infection (mean LCTG-002 IC50 49.8ug/mL versus 114.5ug/mL for control). LCTG-002 was tested for its capacity to reduce viral lung burden in K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 significantly reduced SARS-CoV-2 titers compared to control when administered at 0.25 mg/day or 1 mg/day, with a maximum TCID50 reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure and efficacious in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Camundongos , Animais , Leite Humano , Imunoglobulina A Secretora , Modelos Animais de Doenças , Imunoglobulina A , Camundongos Transgênicos , Antivirais
3.
bioRxiv ; 2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37693438

RESUMO

Immunoglobulin A (IgA) is the most abundant antibody (Ab) in human mucosal compartments including the respiratory tract, with the secretory form of IgA (sIgA) being dominant and uniquely stable in these environments. sIgA is naturally found in human milk, which could be considered a global resource for this biologic, justifying the development of human milk sIgA as a dedicated airway therapeutic for respiratory infections such as SARS-CoV-2. In the present study, methods were therefore developed to efficiently extract human milk sIgA from donors who were either immunologically naïve to SARS-CoV-2 (pooled as a control IgA) or had recovered from a PCR-confirmed SARS-CoV-2 infection that elicited high-titer anti-SARS-CoV-2 Spike sIgA Abs in their milk (pooled together to make LCTG-002). Mass spectrometry determined that proteins with a relative abundance of 1.0% or greater were all associated with sIgA. None of the proteins exhibited statistically significant differences between batches. Western blot demonstrated all batches consisted predominantly of sIgA. Compared to control IgA, LCTG-002 demonstrated significantly higher binding to Spike, and was also capable of blocking the Spike - ACE2 interaction in vitro with 6.3x greater potency compared to control IgA (58% inhibition at ∼240ug/mL). LCTG-002 was then tested in vivo for its capacity to reduce viral burden in the lungs of K18+hACE2 transgenic mice inoculated with SARS-CoV-2. LCTG-002 was demonstrated to significantly reduce SARS-CoV-2 titers in the lungs compared to control IgA when administered at either 250ug/day or 1 mg/day, as measured by TCID50, plaque forming units (PFU), and qRT-PCR, with a maximum reduction of 4.9 logs. This innovative study demonstrates that LCTG-002 is highly pure, efficacious, and well tolerated in vivo, supporting further development of milk-derived, polyclonal sIgA therapeutics against SARS-CoV-2 and other mucosal infections.

4.
ACS Med Chem Lett ; 13(3): 417-427, 2022 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-35300094

RESUMO

Mycobacterium abscessus causes difficult-to-cure pulmonary infections. The bacterium is resistant to most anti-infective agents, including first line antituberculosis (anti-TB) drugs. MMV688844 (844) is a piperidine-4-carboxamide (P4C) with bactericidal properties against M. abscessus. We recently identified DNA gyrase as the molecular target of 844. Here, we present in silico docking and genetic evidence suggesting that P4Cs display a similar binding mode to DNA gyrase as gepotidacin. Gepotidacin is a member of the Novel Bacterial Topoisomerase Inhibitors (NBTIs), a new class of nonfluoroquinolone DNA gyrase poisons. Thus, our work suggests that P4Cs present a novel structural subclass of NBTI. We describe structure-activity relationship studies of 844 leading to analogues showing increased antibacterial activity. Selected derivatives were tested for their inhibitory activity against recombinant M. abscessus DNA gyrase. Further optimization of the lead structures led to improved stability in mouse plasma and increased oral bioavailability.

5.
Antimicrob Agents Chemother ; 66(3): e0221221, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35099272

RESUMO

Nontuberculous mycobacterial pulmonary disease (NTM-PD) is a potentially fatal infectious disease requiring long treatment duration with multiple antibiotics and against which there is no reliable cure. Among the factors that have hampered the development of adequate drug regimens is the lack of an animal model that reproduces the NTM lung pathology required for studying antibiotic penetration and efficacy. Given the documented similarities between tuberculosis and NTM immunopathology in patients, we first determined that the rabbit model of active tuberculosis reproduces key features of human NTM-PD and provides an acceptable surrogate model to study lesion penetration. We focused on clarithromycin, a macrolide and pillar of NTM-PD treatment, and explored the underlying causes of the disconnect between its favorable potency and pharmacokinetics and inconsistent clinical outcome. To quantify pharmacokinetic-pharmacodynamic target attainment at the site of disease, we developed a translational model describing clarithromycin distribution from plasma to lung lesions, including the spatial quantitation of clarithromycin and azithromycin in mycobacterial lesions of two patients on long-term macrolide therapy. Through clinical simulations, we visualized the coverage of clarithromycin in plasma and four disease compartments, revealing heterogeneous bacteriostatic and bactericidal target attainment depending on the compartment and the corresponding potency against nontuberculous mycobacteria in clinically relevant assays. Overall, clarithromycin's favorable tissue penetration and lack of bactericidal activity indicated that its clinical activity is limited by pharmacodynamic, rather than pharmacokinetic, factors. Our results pave the way toward the simulation of lesion pharmacokinetic-pharmacodynamic coverage by multidrug combinations to enable the prioritization of promising regimens for clinical trials.


Assuntos
Pneumopatias , Infecções por Mycobacterium não Tuberculosas , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Humanos , Pneumopatias/tratamento farmacológico , Pneumopatias/microbiologia , Macrolídeos/farmacologia , Macrolídeos/uso terapêutico , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Micobactérias não Tuberculosas , Coelhos
6.
ACS Infect Dis ; 7(8): 2492-2507, 2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34279904

RESUMO

SQ109 is a drug candidate for the treatment of tuberculosis (TB). It is thought to target primarily the protein MmpL3 in Mycobacterium tuberculosis, but it also inhibits the growth of some other bacteria. SQ109 is metabolized by the liver, and it has been proposed that some of its metabolites might be responsible for its activity against TB. Here, we synthesized six potential P450 metabolites of SQ109 and used these as well as 10 other likely metabolites as standards in a mass spectrometry study of M. tuberculosis-infected rabbits treated with SQ109, in addition to testing all 16 putative metabolites for antibacterial activity. We found that there were just two major metabolites in lung tissue: a hydroxy-adamantyl analog of SQ109 and N'-adamantylethylenediamine. Neither of these, or the other potential metabolites tested, inhibited the growth of M. tuberculosis or of M. smegmatis, Bacillus subtilis, or E. coli, making it unlikely that an SQ109 metabolite contributes to its antibacterial activity. In the rabbit TB model, it is thus the gradual accumulation of nonmetabolized SQ109 in tissues to therapeutic levels that leads to good efficacy. Our results also provide new insights into how SQ109 binds to its target MmpL3, based on our mass spectroscopy results which indicate that the charge in SQ109 is primarily localized on the geranyl nitrogen, explaining the very short distance to a key Asp found in the X-ray structure of SQ109 bound to MmpL3.


Assuntos
Mycobacterium tuberculosis , Preparações Farmacêuticas , Tuberculose , Animais , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Escherichia coli , Coelhos , Tuberculose/tratamento farmacológico
7.
Antimicrob Agents Chemother ; 65(8): e0067621, 2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-34001512

RESUMO

New, more-effective drugs for the treatment of lung disease caused by nontuberculous mycobacteria (NTM) are needed. Among NTM opportunistic pathogens, Mycobacterium abscessus is the most difficult to cure and intrinsically multidrug resistant. In a whole-cell screen of a compound collection active against Mycobacterium tuberculosis, we previously identified the piperidine-4-carboxamide (P4C) MMV688844 (844) as a hit against M. abscessus. Here, we identified a more potent analog of 844 and showed that both the parent and improved analog retain activity against strains representing all three subspecies of the M. abscessus complex. Furthermore, P4Cs showed bactericidal and antibiofilm activity. Spontaneous resistance against the P4Cs emerged at a frequency of 10-8/CFU and mapped to gyrA and gyrB encoding the subunits of DNA gyrase. Biochemical studies with recombinant M. abscessus DNA gyrase showed that P4Cs inhibit the wild-type enzyme but not the P4C-resistant mutant. P4C-resistant strains showed limited cross-resistance to the fluoroquinolone moxifloxacin, which is in clinical use for the treatment of macrolide-resistant M. abscessus disease, and no cross-resistance to the benzimidazole SPR719, a novel DNA gyrase inhibitor in clinical development for the treatment of mycobacterial diseases. Analyses of P4Cs in recA promoter-based DNA damage reporter strains showed induction of recA promoter activity in the wild type but not in the P4C-resistant mutant background. This indicates that P4Cs, similar to fluoroquinolones, cause DNA gyrase-mediated DNA damage. Together, our results show that P4Cs present a novel class of mycobacterial DNA gyrase inhibitors with attractive antimicrobial activities against the M. abscessus complex.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Antibacterianos/farmacologia , DNA Girase/genética , Humanos , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Mycobacterium abscessus/genética , Micobactérias não Tuberculosas , Piperidinas/farmacologia
8.
Front Microbiol ; 11: 359, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194537

RESUMO

Persistence of infection despite extensive chemotherapy with antibiotics displaying low MICs is a hallmark of lung disease caused by Mycobacterium abscessus (Mab). Thus, the classical MIC assay is a poor predictor of clinical outcome. Discovery of more efficacious antibiotics requires more predictive in vitro potency assays. As a mycobacterium, Mab is an obligate aerobe and a chemo-organo-heterotroph - it requires oxygen and organic carbon sources for growth. However, bacteria growing in patients can encounter micro-environmental conditions that are different from aerated nutrient-rich broth used to grow planktonic cultures for MIC assays. These in vivo conditions may include oxygen and nutrient limitation which should arrest growth. Furthermore, Mab was shown to grow as biofilms in vivo. Here, we show Mab Bamboo, a clinical isolate we use for Mab drug discovery, can survive oxygen deprivation and nutrient starvation for extended periods of time in non-replicating states and developed an in vitro model where the bacterium grows as biofilm. Using these culture models, we show that non-replicating or biofilm-growing bacteria display tolerance to clinically used anti-Mab antibiotics, consistent with the observed persistence of infection in patients. To demonstrate the utility of the developed "persister" assays for drug discovery, we determined the effect of novel agents targeting membrane functions against these physiological forms of the bacterium and find that these compounds show "anti-persister" activity. In conclusion, we developed in vitro "persister" assays to fill an assay gap in Mab drug discovery compound progression and to enable identification of novel lead compounds showing "anti-persister" activity.

9.
Tuberculosis (Edinb) ; 101: 44-48, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27865396

RESUMO

Tuberculosis (TB) remains an important cause of mortality and morbidity. The TB vaccine, BCG, is not fully protective against the adult form of the disease and is unable to prevent its transmission although it is still useful against severe childhood TB. Hence, the search for new vaccines is of great interest. In a previous study, we have shown that proteoliposomes obtained from Mycobacterium smegmatis (PLMs) induced cross reactive humoral and cellular response against Mycobacterium tuberculosis (Mtb) antigens. With the objective to evaluate the protective capability of PLMs, a murine model of progressive pulmonary TB was used. Animals immunized with PLMs with and without alum (PLMs/PLMsAL respectively) showed protection compared to non-immunized animals. Mice immunized with PLMsAL induced similar protection as that of BCG. Animals immunized with BCG, PLMs and PLMsAL showed a significant decrease in tissue damage (percentage of pneumonic area/lung) compared to non-immunized animals, with a more prominent effect in BCG vaccinated mice. The protective effect of the administration of PLMs in mice supports its future evaluation as experimental vaccine candidate against Mtb.


Assuntos
Mycobacterium smegmatis/imunologia , Proteolipídeos/imunologia , Vacinas contra a Tuberculose , Tuberculose Pulmonar/prevenção & controle , Adjuvantes Imunológicos , Compostos de Alúmen , Animais , Vacina BCG , Carga Bacteriana , Modelos Animais de Doenças , Progressão da Doença , Masculino , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/isolamento & purificação , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Pneumonia Bacteriana/prevenção & controle , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
10.
Hum Vaccin Immunother ; 11(3): 657-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25671612

RESUMO

Tuberculosis (TB) is one of the most important causes of mortality and morbidity due to infectious diseases. BCG, the vaccine in use, is not fully protective against TB. In a previous study, we have shown that proteoliposomes (outer membrane extracts), obtained from BCG (PLBCG) were able to induce humoral immune responses against Mycobacterium tuberculosis (Mtb) antigens. With the objective to evaluate the protective capability of PLBCG alone or as a booster with BCG, a murine model of progressive pulmonary TB was used. Animals immunized with PLBCG adjuvanted with alum (PLBCG-Al) showed similar protection to that conferred by BCG. The group immunized with PLBCG-Al as a booster to BCG gave superior protection than BCG as evidenced by a reduction of bacterial load in lungs 2 months after infection with Mtb. Animals immunized with BCG, PLBCG-Al and this formulation as a booster of BCG, showed a significant decrease of tissue damage (percentage of pneumonic area/lung) compared with non-immunized animals. These results demonstrate that immunization with PLBCG-Al alone or as a booster to BCG induce appropriate protection against challenge with Mtb in mice and support the future evaluation of PLBCG as a promising vaccine candidate against Mtb.


Assuntos
Mycobacterium bovis/imunologia , Proteolipídeos/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Carga Bacteriana , Modelos Animais de Doenças , Pulmão/microbiologia , Masculino , Camundongos Endogâmicos BALB C , Mycobacterium bovis/química , Mycobacterium tuberculosis/isolamento & purificação , Proteolipídeos/administração & dosagem , Proteolipídeos/isolamento & purificação , Tuberculose/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/isolamento & purificação
11.
Biomed Res Int ; 2014: 273129, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25548767

RESUMO

A more effective vaccine against tuberculosis (TB) is urgently needed. Based on its high genetic homology with Mycobacterium tuberculosis (Mtb), the nonpathogenic mycobacteria, Mycobacterium smegmatis (Ms), could be an attractive source of potential antigens to be included in such a vaccine. We evaluated the capability of lipid-based preparations obtained from Ms to provide a protective response in Balb/c mice after challenge with Mtb H37Rv strain. The intratracheal model of progressive pulmonary TB was used to assess the level of protection in terms of bacterial load as well as the pathological changes in the lungs of immunized Balb/c mice following challenge with Mtb. Mice immunized with the lipid-based preparation from Ms either adjuvanted with Alum (LMs-AL) or nonadjuvanted (LMs) showed significant reductions in bacterial load (P < 0.01) compared to the negative control group (animals immunized with phosphate buffered saline (PBS)). Both lipid formulations showed the same level of protection as Bacille Calmette and Guerin (BCG). Regarding the pathologic changes in the lungs, mice immunized with both lipid formulations showed less pneumonic area when compared with the PBS group (P < 0.01) and showed similar results compared with the BCG group. These findings suggest the potential of LMs as a promising vaccine candidate against TB.


Assuntos
Imunização , Mycobacterium smegmatis/imunologia , Tuberculose Pulmonar/prevenção & controle , Vacinação , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/uso terapêutico , Vacina BCG/administração & dosagem , Vacina BCG/imunologia , Modelos Animais de Doenças , Humanos , Lipídeos/administração & dosagem , Lipídeos/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia
12.
Malays J Med Sci ; 21(3): 31-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-25246833

RESUMO

BACKGROUND: Humoral and cellular immune responses are associated with protection against extracellular and intracellular pathogens, respectively. In the present study, we evaluated the effect of receiving human secretory immunoglobulin A (hsIgA) on the histopathology of the lungs of mice challenged with virulent Mycobacterium tuberculosis. METHODS: The hsIgA was purified from human colostrum and administered to Balb/c mice by the intranasal route prior to infection with M. tuberculosis or in a pre-incubated formulation with mycobacteria, with the principal aim to study its effect on qualitative pulmonary histopathology. RESULTS: The intranasal administration of hsIgA and the pre-incubation of mycobacteria with this preparation was associated with the presence of organised granulomas with signs of immune activation and histological features related to efficient disease control. This effect was highly evident during the late stage of infection (60 days), as demonstrated by numerous organised granulomas with numerous activated macrophages in the lungs of treated mice. CONCLUSION: The administration of hsIgA to mice before intratracheal infection with M. tuberculosis or the pre-incubation of the bacteria with the antibody formulation induced the formation of well-organised granulomas and inflammatory lesions in lungs compared with non-treated animals which correlates with the protective effect already demonstrated by these antibody formulations.

13.
BMC Immunol ; 14 Suppl 1: S3, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23458564

RESUMO

BACKGROUND: Immunoglobulin A is the most abundant isotype in secretions from mucosal surfaces of the gastrointestinal, respiratory and genitourinary tracts and in external secretions such as colostrum, breast milk, tears and saliva. The high concentration of human secretory IgA (hsIgA) in human colostrum strongly suggests that it should play an important role in the passive immune protection against gastrointestinal and respiratory infections. MATERIALS AND METHODS: Human secretory IgA was purified from colostrum. The reactivity of hsIgA against mycobacterial antigens and its protective capacity against mycobacterial infection was evaluated. RESULTS: The passive administration of hsIgA reduces the pneumonic area before challenge with M. tuberculosis. The intratracheal administration of M. tuberculosis preincubated with hsIgA to mice greatly reduced the bacterial load in the lungs and diminished lung tissue injury. CONCLUSIONS: HsIgA purified from colostrum protects against M. tuberculosis infection in an experimental mouse model.


Assuntos
Colostro/imunologia , Imunização Passiva , Imunoglobulina A Secretora/administração & dosagem , Imunoglobulina A Secretora/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/prevenção & controle , Animais , Antígenos de Bactérias/imunologia , Carga Bacteriana , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/fisiologia , Tuberculose Pulmonar/imunologia
14.
BMC Immunol ; 14 Suppl 1: S5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23458635

RESUMO

Recombinant Mycobacterium bovis bacille Calmette-Guèrin (rBCG) expressing three T cell epitopes of Mycobacterium tuberculosis (MTB) Ag85B antigen (P1, P2, P3) fused to the Mtb8.4 protein (rBCG018) or a combination of these antigens fused to B cell epitopes from ESAT-6, CFP-10 and MTP40 proteins (rBCG032) were used to immunize Balb/c mice. Total IgG responses were determined against Mtb8.4 antigen and ESAT-6 and CFP-10 B cell epitopes after immunization with rBCG032. Mice immunized with rBCG032 showed a significant increase in IgG1 and IgG2a antibodies against ESAT-6 and MTP40 (P1) B cell epitopes and IgG3 against both P1 and P2 B cell epitopes of MPT40. Splenocytes from mice immunized with rBCG018 proliferated against Ag85B P2 and P3 T cell epitopes and Mtb8.4 protein whereas those from mice-immunized with rBCG032 responded against all Ag85B epitopes and the ESAT-6 B cell epitope. CD4⁺ and CD8⁺ lymphocytes from mice immunized with rBCG018 produced primarily Th1 type cytokines in response to the T cell epitopes. Similar pattern of recognition against the T cell epitopes were obtained with rBCG032 with the additional recognition of ESAT-6, CFP-10 and one of the MTP40 B cell epitopes with the same pattern of cytokines. This study demonstrates that rBCG constructs expressing either T or T and B cell epitopes of MTB induced appropriate immunogenicity against MTB.


Assuntos
Antígenos de Bactérias/imunologia , Vacina BCG/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Mycobacterium tuberculosis/imunologia , Aciltransferases/imunologia , Aciltransferases/metabolismo , Adjuvantes Imunológicos , Animais , Antígenos de Bactérias/metabolismo , Vacina BCG/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Epitopos de Linfócito B/biossíntese , Epitopos de Linfócito T/biossíntese , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Fosfolipases Tipo C/imunologia , Fosfolipases Tipo C/metabolismo , Vacinação , Vacinas Sintéticas/imunologia
15.
Vaccimonitor ; 21(1)ene.-abr. 2012.
Artigo em Espanhol | CUMED | ID: cum-56647

RESUMO

La IgA secretora humana (IgAsh) es estructural y funcionalmente liberada por el ambiente mucosal, con la capacidad de neutralizar antígenos, participa en la aglutinación y exclusión de estos y previene la adherencia de patógenos a las superficies del epitelio mucosal. En este estudio se evaluó la biodistribución de la IgAsh purificada de calostro, después de su administración por vía intranasal, en el modelo de ratón Balb/c y se determinaron los niveles de esta inmunoglobulina en diferentes fluidos biológicos mediante ELISA. Los resultados mostraron la presencia del anticuerpo en saliva de los animales del grupo que recibió la IgAsh en todos los intervalos de tiempo estudiados. En las muestras de lavado tráqueo-bronquial se obtuvo solo la presencia del anticuerpo a las 2 y 3 h posteriores a la inoculación. A partir de los resultados obtenidos consideramos interesante en el futuro utilizar este modelo experimental para evaluar el papel protector de esta inmunoglobulina como candidato terapéutico frente a la infección por Mycobacterium tuberculosis(AU)


The human secretory IgA (IgAsh) is structural and functionally released by the mucosal environment, with the ability to neutralize antigens and participating in promoting the binding and exclusion of them and preventing the adhesion of pathogens to mucosal epithelial surfaces. In this study, the biodistribution of hsIgA purified from colostrum was evaluated, after being administered intranasally to BALB/c mice model and the levels of this immunoglobulin in several biological fluids were determined by ELISA. Results showed the presence of antibody in saliva samples from animals that received the IgAsh, which was significantly higher than levels found in samples from non-treated animals, at all time intervals. In samples of tracheobronchial lavage, high antibody levels were only obtained two and three hours after the inoculation of the animals of the group that received the IgAsh, compared to the non-treated group. According to the results obtained, we consider interesting to evaluate the protective role of this immunoglobulin as a therapeutic candidate against the infection by M. tuberculosis in a near future(AU)


Assuntos
Animais , Camundongos , Imunoglobulina A
16.
Vaccine ; 29(37): 6236-41, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21736914

RESUMO

Proteoliposomes (PL) obtained from Mycobacterium smegmatis (Ms) were evaluated for their capacity to elicit cross-reactive responses against Mycobacterium tuberculosis (Mtb) antigens in BALB/c mice. Animals immunized with PL adjuvanted with alum (PL-AL) or Freund's Incomplete Adjuvant (PL-IFA) showed significant IgG responses against the PL as well as total Ms lipids. Both groups of animals also showed significant IgG responses against BCG, but only animals immunized with PL-AL produced significant IgG responses against soluble cell wall proteins (SCWP) or whole cell lysate (WCL) of Mtb. Significant DTH responses against WCL were observed in both groups of animals after 24 h, but only PL-AL-immunized mice showed significant DTH responses after 48 h and 72 h. PL-Ms are capable of eliciting cross-reactive humoral and cellular responses against Mtb antigens and thus may be a potential vaccine strategy against tuberculosis.


Assuntos
Parede Celular/imunologia , Mycobacterium smegmatis/imunologia , Mycobacterium tuberculosis/imunologia , Proteolipídeos/imunologia , Adjuvantes Imunológicos , Compostos de Alúmen/farmacologia , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias , Reações Cruzadas , Feminino , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium bovis/imunologia , Tuberculose/imunologia , Tuberculose/prevenção & controle , Vacinas contra a Tuberculose
17.
Vaccimonitor ; 20(1)ene.-abr. 2011.
Artigo em Espanhol | CUMED | ID: cum-47089

RESUMO

Mycobacterium tuberculosis es el agente causal de la tuberculosis, una de las enfermedades infecciosas más letales en el mundo. La única vacuna disponible para su control es el BCG, sin embargo, falla en la protección contra la tuberculosis pulmonar, siendo esta la forma más frecuente y responsable de la diseminación. La identificación de factores de virulencia del microorganismo causal pudiera ayudar en el desarrollo de un nuevo candidato vacunal que sea capaz de neutralizar la acción de esos determinantes patogénicos. El empleo de diferentes modelos animales ha permitido reproducir las etapas de la enfermedad, así como medir o cuantificar la virulencia de las distintas cepas circulantes de Mycobacterium tuberculosis. Las mutaciones génicas y otras técnicas de biología molecular han posibilitado dilucidar los genes específicos involucrados en la virulencia de este microorganismo que codifican para múltiples y complejos factores de diferente naturaleza(AU)


Mycobacterium tuberculosis is the causative agent of tuberculosis, one of the most lethal diseases worldwide. BCG is the only available vaccine for tuberculosis control, but at the same time it fails in the protection from pulmonary tuberculosis, which is the most common and responsible form of dissemination. The identification of virulence factors of the causative organism could help in developing a new vaccine candidate capable of neutralizing the action of these pathogenic determinants. The use of different animal models has allowed to reproduce the stages of the disease and to measure or to quantify the virulence of different circulating strains of M tuberculosis. Gene mutations and other molecular biology techniques have made possible to elucidate the specific genes involved in virulence of this organism, that encodes many complex and different factors(AU)


Assuntos
Humanos , Mycobacterium tuberculosis/patogenicidade , Mycobacterium tuberculosis/virologia , Fatores de Virulência
18.
Malays J Med Sci ; 18(4): 5-12, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22589668

RESUMO

Research, development, and production of vaccines are still highly dependent on the use of animal models in the various evaluation steps. Despite this fact, there are strong interests and ongoing efforts to reduce the use of animals in vaccine development. Tuberculosis vaccine development is one important example of the complexities involved in the use of animal models for the production of new vaccines. This review summarises some of the general aspects related with the use of animals in vaccine research and production, as well as achievements and challenges towards the rational use of animals, particularly in the case of tuberculosis vaccine development.

19.
Vaccimonitor ; 19(1)ene.-abr. 2010. ilus, graf, tab
Artigo em Inglês | CUMED | ID: cum-43093

RESUMO

In this study, we scanned multiple published databases of gene expression in vivo of M tuberculosis at different phases of infection in animals and humans, to select 38 proteins that are highly expressed in the active, latent and reactivation phases. The selected proteins were predicted for T and B epitopes. For each proteins, the regions containing T and B epitopes were selected at the same time to look for identical epitopes on M smegmatis based on sequence alignments. Preliminary studies of humoral immunogenicity and cross-reactivity with M tuberculosis in mice using two M smegmatis-derived experimental vaccines were carried out, demonstrating the immunogenicity of M smegmatis proteoliposomes and the recognition of M tuberculosis proteins by the sera of animals immunized with this vaccine candidate. The conjunction of in silico and in vivo studies suggested the potential for future evaluation of M smegmatis as vaccine candidate against tuberculosis using different strategies.(AU)


En este estudio se revisaron múltiples bases de datos publicadas, relacionadas con experimentos de expresión de genes de M tuberculosis in vivo en diferentes estadios de la infeccción en humanos y animales. Se identificaron 38 proteínas con elevada expresión en las fases activa, latente y de reactivación de la infección. Se llevó a cabo la predicción de epítopes T y B en dichas proteínas. Las regiones de cada proteína que contenían simultàneamente epítopes T y B se seleccionaron y utilizaron para identificar regiones idénticas en M smegmatis mediante el alineamiento de secuencias. Se llevaron a cabo estudios de inmunogenicidad humoral y reactividad cruzada con M tuberculosis en ratones inmunizados con dos vacunas experimentales obtenidas a partir de M smegmatis, demostràndose la immunogenicidad de los proteoliposomas y el reconocimiento de proteínas de M tuberculosis por el suero de ratones vacunados con este candidato vacunal. Los resultados obtenidos con los estudios in sílico e in vivo sugieren la potencialidad para evaluación futura de candidatos vacunales obtenidos a partir de M smegmatis para la prevención de la tuberculosis(AU)


Assuntos
Mycobacterium tuberculosis , Mycobacterium smegmatis , Epitopos , Previsões
20.
Vaccimonitor ; 19(3)2010.
Artigo em Espanhol | CUMED | ID: cum-47082

RESUMO

La inmunoglobulina A (IgA) es el isotipo de anticuerpo más abundante en las secreciones de las superficies mucosales del tracto gastrointestinal, respiratorio y genitourinario y en secreciones externas como el calostro, la leche materna, las lágrimas y la saliva. En el presente trabajo se obtuvo inmunoglobulina A secretora a partir de calostro humano mediante una combinación de métodos cromatográficos, y su presencia fue demostrada mediante Dot blot. La fracción de IgA fue analizada mediante electroforesis en gel de poliacrilamida en presencia de dodecilsulfato de sodio (SDS-PAGE) bajo condiciones reductoras, obteniéndose una elevada pureza al identificarse la presencia del componente secretor, la cadena pesada y la cadena ligera de dicha inmunoglobulina, con un patrón de migración correspondiente a sus respectivas masas molares(AU)


Immunoglobulin A is the most abundant antibody isotype in secretions from mucosal surfaces of the gastrointestinal, respiratory, and genitourinary tracts and in external secretions such as colostrums, breast milk, tears and saliva. In this study, secretory immunoglobulin A was obtained from human colostrum by a combination of chromatographic methods which was demonstrated by Dot blot. IgA fraction was analyzed by SDS-PAGE electrophoresis under reducing conditions, obtaining a high purity product by identifying the presence of secretory component, heavy and light chains of the immunoglobulin, with a migration pattern corresponding to their respective molecular mass(AU)


Assuntos
Humanos , Feminino , Gravidez , Imunoglobulina A Secretora , Colostro/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...