Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
PLoS Pathog ; 12(1): e1005356, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808628

RESUMEN

Persistent viral infections are simultaneously associated with chronic inflammation and highly potent immunosuppressive programs mediated by IL-10 and PDL1 that attenuate antiviral T cell responses. Inhibiting these suppressive signals enhances T cell function to control persistent infection; yet, the underlying signals and mechanisms that program immunosuppressive cell fates and functions are not well understood. Herein, we use lymphocytic choriomeningitis virus infection (LCMV) to demonstrate that the induction and functional programming of immunosuppressive dendritic cells (DCs) during viral persistence are separable mechanisms programmed by factors primarily considered pro-inflammatory. IFNγ first induces the de novo development of naive monocytes into DCs with immunosuppressive potential. Type I interferon (IFN-I) then directly targets these newly generated DCs to program their potent T cell immunosuppressive functions while simultaneously inhibiting conventional DCs with T cell stimulating capacity. These mechanisms of monocyte conversion are constant throughout persistent infection, establishing a system to continuously interpret and shape the immunologic environment. MyD88 signaling was required for the differentiation of suppressive DCs, whereas inhibition of stimulatory DCs was dependent on MAVS signaling, demonstrating a bifurcation in the pathogen recognition pathways that promote distinct elements of IFN-I mediated immunosuppression. Further, a similar suppressive DC origin and differentiation was also observed in Mycobacterium tuberculosis infection, HIV infection and cancer. Ultimately, targeting the underlying mechanisms that induce immunosuppression could simultaneously prevent multiple suppressive signals to further restore T cell function and control persistent infections.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Interferones/inmunología , Virosis/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , VIH , Infecciones por VIH/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología , Tuberculosis/inmunología
2.
Infect Immun ; 85(9)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28630063

RESUMEN

A potent vaccine against tuberculosis, one of the world's deadliest diseases, is needed to enhance the immunity of people worldwide, most of whom have been vaccinated with the partially effective Mycobacterium bovis BCG vaccine. Here we investigate novel live attenuated recombinant Listeria monocytogenes (rLm) vaccines expressing the Mycobacterium tuberculosis 30-kDa major secretory protein (r30/antigen 85B [Ag85B]) (rLm30) as heterologous booster vaccines in animals primed with BCG. Using three attenuated L. monocytogenes vectors, L. monocytogenes ΔactA (LmI), L. monocytogenes ΔactA ΔinlB (LmII), and L. monocytogenes ΔactA ΔinlB prfA* (LmIII), we constructed five rLm30 vaccine candidates expressing r30 linked in frame to the L. monocytogenes listeriolysin O signal sequence and driven by the hly promoter (h30) or linked in frame to the ActA N-terminal 100 amino acids and driven by the actA promoter (a30). All five rLm30 vaccines secreted r30 in broth and macrophages; while rLm30 expressing r30 via a constitutively active prfA* regulon (rLmIII/a30) expressed the largest amount of r30 in broth culture, all five rLm30 vaccines expressed equivalent amounts of r30 in infected macrophages. In comparative studies, boosting of BCG-immunized mice with rLmIII/a30 induced the strongest antigen-specific T-cell responses, including splenic and lung polyfunctional CD4+ T cells expressing the three cytokines interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin-2 (IL-2) (P < 0.001) and splenic and lung CD8+ T cells expressing IFN-γ (P < 0.0001). In mice and guinea pigs, the rLmIII/a30 and rLmI/h30 vaccines were generally more potent booster vaccines than r30 with an adjuvant and a recombinant adenovirus vaccine expressing r30. In a setting in which BCG alone was highly immunoprotective, boosting of mice with rLmIII/a30, the most potent of the vaccines, significantly enhanced protection against aerosolized M. tuberculosis (P < 0.01).

3.
Small ; 12(27): 3690-702, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27246117

RESUMEN

Effective and rapid treatment of tularemia is needed to reduce morbidity and mortality of this potentially fatal infectious disease. The etiologic agent, Francisella tularensis, is a facultative intracellular bacterial pathogen which infects and multiplies to high numbers in macrophages. Nanotherapeutics are particularly promising for treatment of infectious diseases caused by intracellular pathogens, whose primary host cells are macrophages, because nanoparticles preferentially target and are avidly internalized by macrophages. A mesoporous silica nanoparticle (MSN) has been developed functionalized with disulfide snap-tops that has high drug loading and selectively releases drug intracellularly in response to the redox potential. These nanoparticles, when loaded with Hoechst fluorescent dye, release their cargo exclusively intracellularly and stain the nuclei of macrophages. The MSNs loaded with moxifloxacin kill F. tularensis in macrophages in a dose-dependent fashion. In a mouse model of lethal pneumonic tularemia, MSNs loaded with moxifloxacin prevent weight loss, illness, and death, markedly reduce the burden of F. tularensis in the lung, liver, and spleen, and are significantly more efficacious than an equivalent amount of free drug. An important proof-of-principle for the potential therapeutic use of a novel nanoparticle drug delivery platform for the treatment of infectious diseases is provided.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fluoroquinolonas/química , Fluoroquinolonas/uso terapéutico , Nanopartículas/química , Dióxido de Silicio/química , Tularemia/tratamiento farmacológico , Animales , Femenino , Fluoroquinolonas/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Moxifloxacino
4.
Small ; 11(38): 5066-78, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26193431

RESUMEN

Tuberculosis is a major global health problem for which improved therapeutics are needed to shorten the course of treatment and combat emergence of drug resistance. Mycobacterium tuberculosis, the etiologic agent of tuberculosis, is an intracellular pathogen of mononuclear phagocytes. As such, it is an ideal pathogen for nanotherapeutics because macrophages avidly ingest nanoparticles even without specific targeting molecules. Hence, a nanoparticle drug delivery system has the potential to target and deliver high concentrations of drug directly into M. tuberculosis-infected cells-greatly enhancing efficacy while avoiding off-target toxicities. Stimulus-responsive mesoporous silica nanoparticles of two different sizes, 100 and 50 nm, are developed as carriers for the major anti-tuberculosis drug isoniazid in a prodrug configuration. The drug is captured by the aldehyde-functionalized nanoparticle via hydrazone bond formation and coated with poly(ethylene imine)-poly(ethylene glycol) (PEI-PEG). The drug is released from the nanoparticles in response to acidic pH at levels that naturally occur within acidified endolysosomes. It is demonstrated that isoniazid-loaded PEI-PEG-coated nanoparticles are avidly ingested by M. tuberculosis-infected human macrophages and kill the intracellular bacteria in a dose-dependent manner. It is further demonstrated in a mouse model of pulmonary tuberculosis that the nanoparticles are well tolerated and much more efficacious than an equivalent amount of free drug.


Asunto(s)
Isoniazida/uso terapéutico , Nanopartículas/química , Tuberculosis/tratamiento farmacológico , Aldehídos/química , Animales , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Concentración de Iones de Hidrógeno , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones Endogámicos BALB C , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Nanopartículas/ultraestructura , Polietilenglicoles/química , Polietileneimina/química , Porosidad , Profármacos/uso terapéutico , Dióxido de Silicio/química , Espectrofotometría Ultravioleta , Tuberculosis/microbiología
5.
Small ; 11(38): 5065, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26450161

RESUMEN

On page 5066, J. I. Zink, M. A. Horwitz, and co-workers use confocal microscopy to demonstrate the avid uptake of RITC-labeled mesoporous silica nanoparticles loaded with the anti-tuberculosis drug isoniazid (shown here in red) by human macrophages (nuclei stained blue with DAPI) infected with GFP-expressing Mycobacterium tuberculosis (shown here in green).


Asunto(s)
Isoniazida/uso terapéutico , Nanopartículas/química , Tuberculosis/tratamiento farmacológico , Animales , Concentración de Iones de Hidrógeno , Ratones
6.
Infect Immun ; 81(5): 1550-61, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23439306

RESUMEN

Francisella tularensis, the causative agent of tularemia, is a category A bioterrorism agent. A vaccine that is safer and more effective than the currently available unlicensed F. tularensis live vaccine strain (LVS) is needed to protect against intentional release of aerosolized F. tularensis, the most dangerous type of exposure. In this study, we employed a heterologous prime-boost vaccination strategy comprising intradermally administered LVS ΔcapB (highly attenuated capB-deficient LVS mutant) as the primer vaccine and rLm/iglC (recombinant attenuated Listeria monocytogenes expressing the F. tularensis immunoprotective antigen IglC) as the booster vaccine. Boosting LVS ΔcapB-primed mice with rLm/iglC significantly enhanced T cell immunity; their splenic T cells secreted significantly more gamma interferon (IFN-γ) and had significantly more cytokine (IFN-γ and/or tumor necrosis factor [TNF] and/or interleukin-2 [IL-2])-producing CD4(+) and CD8(+) T cells upon in vitro IglC stimulation. Importantly, mice primed with LVS ΔcapB or rLVS ΔcapB/IglC, boosted with rLm/iglC, and subsequently challenged with 10 50% lethal doses (LD50) of aerosolized highly virulent F. tularensis Schu S4 had a significantly higher survival rate and mean survival time than mice immunized with only LVS ΔcapB (P < 0.0001); moreover, compared with mice immunized once with LVS, primed-boosted mice had a higher survival rate (75% versus 62.5%) and mean survival time during the first 21 days postchallenge (19 and 20 days for mice boosted after being primed with LVS ΔcapB and rLVS ΔcapB/IglC, respectively, versus 17 days for mice immunized with LVS) and maintained their weight significantly better (P < 0.01). Thus, the LVS ΔcapB-rLm/iglC prime-boost vaccination strategy holds substantial promise for a vaccine that is safer and at least as potent as LVS.


Asunto(s)
Vacunas Bacterianas , Francisella tularensis/inmunología , Inmunización Secundaria/métodos , Listeria monocytogenes/inmunología , Tularemia/prevención & control , Vacunación/métodos , Aerosoles , Análisis de Varianza , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Citocinas/metabolismo , Femenino , Inmunidad Celular , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos BALB C , Bazo/citología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Tularemia/inmunología , Tularemia/metabolismo , Vacunas Atenuadas , Vacunas Sintéticas
7.
bioRxiv ; 2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35233569

RESUMEN

To date, there is no effective oral antiviral against SARS-CoV-2 that is also anti-inflammatory. Herein, we show that the mitochondrial antioxidant mitoquinone/mitoquinol mesylate (Mito-MES), a dietary supplement, has potent antiviral activity against SARS-CoV-2 and its variants of concern in vitro and in vivo . Mito-MES had nanomolar in vitro antiviral potency against the Beta and Delta SARS-CoV-2 variants as well as the murine hepatitis virus (MHV-A59). Mito-MES given in SARS-CoV-2 infected K18-hACE2 mice through oral gavage reduced viral titer by nearly 4 log units relative to the vehicle group. We found in vitro that the antiviral effect of Mito-MES is attributable to its hydrophobic dTPP+ moiety and its combined effects scavenging reactive oxygen species (ROS), activating Nrf2 and increasing the host defense proteins TOM70 and MX1. Mito-MES was efficacious reducing increase in cleaved caspase-3 and inflammation induced by SARS-CoV2 infection both in lung epithelial cells and a transgenic mouse model of COVID-19. Mito-MES reduced production of IL-6 by SARS-CoV-2 infected epithelial cells through its antioxidant properties (Nrf2 agonist, coenzyme Q10 moiety) and the dTPP moiety. Given established safety of Mito-MES in humans, our results suggest that Mito-MES may represent a rapidly applicable therapeutic strategy that can be added in the therapeutic arsenal against COVID-19. Its potential long-term use by humans as diet supplement could help control the SARS-CoV-2 pandemic, especially in the setting of rapidly emerging SARS-CoV-2 variants that may compromise vaccine efficacy. One-Sentence Summary: Mitoquinone/mitoquinol mesylate has potent antiviral and anti-inflammatory activity in preclinical models of SARS-CoV-2 infection.

8.
Infect Immun ; 78(10): 4341-55, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20643859

RESUMEN

Francisella tularensis, the causative agent of tularemia, is in the top category (category A) of potential agents of bioterrorism. The F. tularensis live vaccine strain (LVS) is the only vaccine currently available to protect against tularemia; however, this unlicensed vaccine is relatively toxic and provides incomplete protection against aerosolized F. tularensis, the most dangerous mode of transmission. Hence, a safer and more potent vaccine is needed. As a first step toward addressing this need, we have constructed and characterized an attenuated version of LVS, LVS ΔcapB, both as a safer vaccine and as a vector for the expression of recombinant F. tularensis proteins. LVS ΔcapB, with a targeted deletion in a putative capsule synthesis gene (capB), is antibiotic resistance marker free. LVS ΔcapB retains the immunoprotective O antigen, is serum resistant, and is outgrown by parental LVS in human macrophage-like THP-1 cells in a competition assay. LVS ΔcapB is significantly attenuated in mice; the 50% lethal dose (LD(50)) intranasally (i.n.) is >10,000-fold that of LVS. Providing CapB in trans to LVS ΔcapB partially restores its virulence in mice. Mice immunized with LVS ΔcapB i.n. or intradermally (i.d.) developed humoral and cellular immune responses comparable to those of mice immunized with LVS, and when challenged 4 or 8 weeks later with a lethal dose of LVS i.n., they were 100% protected from illness and death and had significantly lower levels (3 to 5 logs) of LVS in the lung, liver, and spleen than sham-immunized mice. Most importantly, mice immunized with LVS ΔcapB i.n. or i.d. and then challenged 6 weeks later by aerosol with 10× the LD(50) of the highly virulent type A F. tularensis strain SchuS4 were significantly protected (100% survival after i.n. immunization). These results show that LVS ΔcapB is significantly safer than LVS and yet provides potent protective immunity against virulent F. tularensis SchuS4 challenge.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacunas Bacterianas , Francisella tularensis/patogenicidad , Tularemia/prevención & control , Administración Intranasal , Animales , Antibacterianos/farmacología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Línea Celular , Femenino , Francisella tularensis/genética , Eliminación de Gen , Marcadores Genéticos , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulinas/metabolismo , Inyecciones Intradérmicas , Ratones , Ratones Endogámicos BALB C , Tularemia/inmunología , Tularemia/microbiología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Virulencia
9.
PLoS One ; 14(5): e0215607, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31075149

RESUMEN

BACKGROUND: Shorter, more effective treatments for tuberculosis (TB) are urgently needed. While many TB drugs are available, identification of the best regimens is challenging because of the large number of possible drug-dose combinations. We have found consistently that responses of cells or whole animals to drug-dose stimulations fit a parabolic response surface (PRS), allowing us to identify and optimize the best drug combinations by testing only a small fraction of the total search space. Previously, we used PRS methodology to identify three regimens (PRS Regimens I-III) that in murine models are much more effective than the standard regimen used to treat TB. However, PRS Regimens I and II are unsuitable for treating drug-resistant TB and PRS Regimen III includes an experimental drug. Here, we use PRS methodology to identify from an expanded pool of drugs new highly effective near-universal drug regimens comprising only approved drugs. METHODS AND FINDINGS: We evaluated combinations of 15 different drugs in a human macrophage TB model and identified the most promising 4-drug combinations. We then tested 14 of these combinations in Mycobacterium tuberculosis-infected BALB/c mice and chose for PRS dose optimization and further study the two most potent regimens, designated PRS Regimens IV and V, consisting of clofazimine (CFZ), bedaquiline (BDQ), pyrazinamide (PZA), and either amoxicillin/clavulanate (AC) or delamanid (DLM), respectively. We then evaluated the efficacy in mice of optimized PRS Regimens IV and V, as well as a 3-drug regimen, PRS Regimen VI (CFZ, BDQ, and PZA), and compared their efficacy to PRS Regimen III (CFZ, BDQ, PZA, and SQ109), previously shown to reduce the time to achieve relapse-free cure in mice by 80% compared with the Standard Regimen (isoniazid, rifampicin, PZA, and ethambutol). Efficacy measurements included early bactericidal activity, time to lung sterilization, and time to relapse-free cure. PRS Regimens III-VI all rapidly sterilized the lungs and achieved relapse-free cure in 3 weeks (PRS Regimens III, V, and VI) or 5 weeks (PRS Regimen IV). In contrast, mice treated with the Standard Regimen still had high numbers of bacteria in their lungs after 6-weeks treatment and none achieved relapse-free cure in this time-period. CONCLUSIONS: We have identified three new regimens that rapidly sterilize the lungs of mice and dramatically shorten the time required to achieve relapse-free cure. All mouse drug doses in these regimens extrapolate to doses that are readily achievable in humans. Because PRS Regimens IV and V contain only one first line drug (PZA) and exclude fluoroquinolones and aminoglycosides, they should be effective against most TB cases that are multidrug resistant (MDR-TB) and many that are extensively drug-resistant (XDR-TB). Hence, these regimens have potential to shorten dramatically the time required for treatment of both drug-sensitive and drug-resistant TB. If clinical trials confirm that these regimens dramatically shorten the time required to achieve relapse-free cure in humans, then this radically shortened treatment has the potential to improve treatment compliance, decrease the emergence of drug resistance, and decrease the healthcare burden of treating both drug-sensitive and drug-resistant TB.


Asunto(s)
Antituberculosos/uso terapéutico , Tuberculosis/tratamiento farmacológico , Animales , Antituberculosos/farmacología , Inteligencia Artificial , Modelos Animales de Enfermedad , Aprobación de Drogas , Combinación de Medicamentos , Cálculo de Dosificación de Drogas , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/efectos de los fármacos , Células THP-1 , Resultado del Tratamiento
10.
ACS Infect Dis ; 5(2): 281-291, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30480992

RESUMEN

Francisella tularensis causes a serious and often fatal infection, tularemia. We compared the efficacy of moxifloxacin formulated as free drug vs disulfide snap-top mesoporous silica nanoparticles (MSNs) in a mouse model of pneumonic tularemia. We found that MSN-formulated moxifloxacin was more effective than free drug and that the intramuscular and subcutaneous routes were markedly more effective than the intravenous route. Measurement of tissue silica levels and fluorescent flow cytometry assessment of colocalization of MSNs with infected cells revealed that the enhanced efficacy of MSNs and the intramuscular route of delivery was not due to better delivery of MSNs to infected tissues or cells. However, moxifloxacin blood levels demonstrated that the nanoparticle formulation and intramuscular route provided the longest half-life and longest time above the minimal inhibitory concentration. Thus, improved pharmacokinetics are responsible for the greater efficacy of nanoparticle formulation and intramuscular delivery compared with free drug and intravenous delivery.


Asunto(s)
Antibacterianos/farmacocinética , Antibacterianos/uso terapéutico , Moxifloxacino/farmacocinética , Moxifloxacino/uso terapéutico , Nanopartículas/química , Tularemia/tratamiento farmacológico , Administración Intravenosa , Animales , Modelos Animales de Enfermedad , Femenino , Francisella tularensis/efectos de los fármacos , Inyecciones Intramusculares , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Neumonía Bacteriana/tratamiento farmacológico , Tularemia/microbiología
11.
Sci Rep ; 8(1): 7009, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29725025

RESUMEN

Bacillus anthracis, Yersinia pestis, and Francisella tularensis are the causative agents of Tier 1 Select Agents anthrax, plague, and tularemia, respectively. Currently, there are no licensed vaccines against plague and tularemia and the licensed anthrax vaccine is suboptimal. Here we report F. tularensis LVS ΔcapB (Live Vaccine Strain with a deletion in capB)- and attenuated multi-deletional Listeria monocytogenes (Lm)-vectored vaccines against all three aforementioned pathogens. We show that LVS ΔcapB- and Lm-vectored vaccines express recombinant B. anthracis, Y. pestis, and F. tularensis immunoprotective antigens in broth and in macrophage-like cells and are non-toxic in mice. Homologous priming-boosting with the LVS ΔcapB-vectored vaccines induces potent antigen-specific humoral and T-cell-mediated immune responses and potent protective immunity against lethal respiratory challenge with all three pathogens. Protection against anthrax was far superior to that obtained with the licensed AVA vaccine and protection against tularemia was comparable to or greater than that obtained with the toxic and unlicensed LVS vaccine. Heterologous priming-boosting with LVS ΔcapB- and Lm-vectored B. anthracis and Y. pestis vaccines also induced potent protective immunity against lethal respiratory challenge with B. anthracis and Y. pestis. The single vaccine platform, especially the LVS ΔcapB-vectored vaccine platform, can be extended readily to other pathogens.


Asunto(s)
Carbunco/prevención & control , Vacunas Bacterianas/inmunología , Portadores de Fármacos , Vectores Genéticos , Peste/prevención & control , Tularemia/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Bacillus anthracis/genética , Bacillus anthracis/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Francisella tularensis/genética , Francisella tularensis/inmunología , Listeria monocytogenes/genética , Ratones , Linfocitos T/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Yersinia pestis/genética , Yersinia pestis/inmunología
12.
PLoS One ; 13(11): e0207469, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30427938

RESUMEN

As current treatment of tuberculosis is burdensomely long, provoking non-adherence and drug resistance, effective short-course treatments are needed. Using the output-driven parabolic response surface (PRS) platform, we have identified drug regimens that treat tuberculosis more rapidly in mice than the current Standard Regimen used in humans. We show that PRS Regimen III, comprising clofazimine, SQ109, bedaquiline and pyrazinamide, rapidly sterilizes the lung both in conventionally studied BALB/c mice and in C3HeB/FeJ mice, highly susceptible mice that develop massive necrotic granulomatous lung lesions akin to those in humans, achieving relapse-free cure in only 4 weeks (p<0.0001 versus Standard Regimen). In contrast, the Standard Regimen required 16 weeks to attain lung culture negative status and 20 weeks to achieve relapse-free cure. Thus, PRS Regimen III dramatically cuts by ~80% the time to relapse-free cure in mouse tuberculosis models. PRS Regimen III, with three nonstandard drugs, can potentially treat both drug-sensitive and most drug-resistant tuberculosis.


Asunto(s)
Antituberculosos/administración & dosificación , Combinación de Medicamentos , Pulmón/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Adamantano/administración & dosificación , Adamantano/análogos & derivados , Animales , Clofazimina/administración & dosificación , Diarilquinolinas/administración & dosificación , Modelos Animales de Enfermedad , Etilenodiaminas/administración & dosificación , Humanos , Pulmón/fisiopatología , Ratones , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Pirazinamida/administración & dosificación , Tuberculosis/microbiología , Tuberculosis/fisiopatología
13.
Nat Commun ; 8: 14183, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28117835

RESUMEN

The current drug regimens for treating tuberculosis are lengthy and onerous, and hence complicated by poor adherence leading to drug resistance and disease relapse. Previously, using an output-driven optimization platform and an in vitro macrophage model of Mycobacterium tuberculosis infection, we identified several experimental drug regimens among billions of possible drug-dose combinations that outperform the current standard regimen. Here we use this platform to optimize the in vivo drug doses of two of these regimens in a mouse model of pulmonary tuberculosis. The experimental regimens kill M. tuberculosis much more rapidly than the standard regimen and reduce treatment time to relapse-free cure by 75%. Thus, these regimens have the potential to provide a markedly shorter course of treatment for tuberculosis in humans. As these regimens omit isoniazid, rifampicin, fluoroquinolones and injectable aminoglycosides, they would be suitable for treating many cases of multidrug and extensively drug-resistant tuberculosis.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Tuberculosis Pulmonar/tratamiento farmacológico , Animales , Antituberculosos/uso terapéutico , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Quimioterapia Combinada/métodos , Femenino , Humanos , Cumplimiento de la Medicación , Ratones , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/aislamiento & purificación , Organismos Libres de Patógenos Específicos , Factores de Tiempo , Resultado del Tratamiento , Tuberculosis Resistente a Múltiples Medicamentos/microbiología , Tuberculosis Pulmonar/microbiología
14.
Vaccine ; 34(41): 4969-4978, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27577555

RESUMEN

A safer and more effective vaccine than the unlicensed Francisella tularensis Live Vaccine Strain (LVS) is needed to protect against the biowarfare agent F. tularensis. Previously, we developed an LVS ΔcapB mutant that is significantly safer than LVS and provides potent protective immunity against F. tularensis respiratory challenge when administered intranasally but limited protection when administered intradermally unless as part of a prime-boost vaccination strategy. To improve the immunogenicity and efficacy of LVS ΔcapB, we developed recombinant LVS ΔcapB (rLVS ΔcapB) strains overexpressing various F. tularensis Francisella Pathogenicity Island (FPI) proteins - IglA, IglB and IglC, and a fusion protein (IglABC) comprising immunodominant epitopes of IglA, IglB, and IglC downstream of different Francisella promoters, including the bacterioferritin (bfr) promoter. We show that rLVS ΔcapB/bfr-iglA, iglB, iglC, and iglABC express more IglA, IglB, IglC or IglABC than parental LVS ΔcapB in broth and in human macrophages, and stably express FPI proteins in macrophages and mice absent antibiotic selection. In response to IglC and heat-inactivated LVS, spleen cells from mice immunized intradermally with rLVS ΔcapB/bfr-iglC or bfr-iglABC secrete greater amounts of interferon-gamma and/or interleukin-17 than those from mice immunized with LVS ΔcapB, comparable to those from LVS-immunized mice. Mice immunized with rLVS ΔcapB/bfr-iglA, iglB, iglC or iglABC produce serum antibodies at levels similar to LVS-immunized mice. Mice immunized intradermally with rLVS ΔcapB/bfr-iglABC and challenged intranasally with virulent F. tularensis Schu S4 survive longer than sham- and LVS ΔcapB-immunized mice. Mice immunized intranasally with rLVS ΔcapB/bfr-iglABC - but not with LVS - just before or after respiratory challenge with F. tularensis Schu S4 are partially protected; protection is correlated with induction of a strong innate immune response. Thus, rLVS ΔcapB/bfr-iglABC shows improved immunogenicity and protective efficacy compared with parental LVS ΔcapB and, in contrast to LVS, has partial efficacy as immediate pre- and post-exposure prophylaxis.


Asunto(s)
Vacunas Bacterianas/inmunología , Inmunogenicidad Vacunal , Tularemia/prevención & control , Sistemas de Secreción Tipo VI/inmunología , Animales , Cápsulas Bacterianas/genética , Proteínas Bacterianas/genética , Grupo Citocromo b/genética , Femenino , Ferritinas/genética , Francisella tularensis/genética , Islas Genómicas/inmunología , Humanos , Interferón gamma/inmunología , Interleucina-17/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Profilaxis Posexposición , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/inmunología , Eliminación de Secuencia , Células THP-1 , Sistemas de Secreción Tipo VI/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/inmunología
15.
ACS Nano ; 9(11): 10778-89, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26435204

RESUMEN

We have optimized mesoporous silica nanoparticles (MSNs) functionalized with pH-sensitive nanovalves for the delivery of the broad spectrum fluoroquinolone moxifloxacin (MXF) and demonstrated its efficacy in treating Francisella tularensis infections both in vitro and in vivo. We compared two different nanovalve systems, positive and negative charge modifications of the mesopores, and different loading conditions-varying pH, cargo concentration, and duration of loading-and identified conditions that maximize both the uptake and release capacity of MXF by MSNs. We have demonstrated in macrophage cell culture that the MSN-MXF delivery platform is highly effective in killing F. tularensis in infected macrophages, and in a mouse model of lethal pneumonic tularemia, we have shown that the drug-loaded MSNs are much more effective in killing F. tularensis than an equivalent amount of free MXF.


Asunto(s)
Sistemas de Liberación de Medicamentos , Fluoroquinolonas/uso terapéutico , Nanopartículas/química , Neumonía/complicaciones , Dióxido de Silicio/química , Tularemia/complicaciones , Tularemia/tratamiento farmacológico , Animales , Bencimidazoles/química , Fluoroquinolonas/farmacología , Francisella tularensis/efectos de los fármacos , Francisella tularensis/fisiología , Humanos , Concentración de Iones de Hidrógeno , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Viabilidad Microbiana/efectos de los fármacos , Moxifloxacino , Ácidos Fosforosos/química , Neumonía/tratamiento farmacológico , Porosidad , Resultado del Tratamiento
16.
Vaccine ; 27(3): 441-5, 2009 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19007841

RESUMEN

BCG has been administered to over 4 billion persons worldwide, but its efficacy in preventing tuberculosis in adults has been highly variable. One hypothesis for its variability is that different strains of BCG vary in protective efficacy, and moreover, that evolutionarily early strains are more efficacious than the more attenuated evolutionarily late strains, which lack region of deletion 2. To examine this hypothesis, we tested six widely used BCG strains--the evolutionarily early strain BCG Japanese, two evolutionarily late strains in DU2 Group III (BCG Danish and Glaxo), and three evolutionarily late strains in DU2 Group IV (BCG Connaught, Pasteur, and Tice)--in the guinea pig model of pulmonary tuberculosis. With the exception of BCG Glaxo, which had relatively poor efficacy, we found no substantial differences in efficacy between the early strain and the late strains, and only small differences in efficacy among late strains. BCG Tice was the most efficacious BCG vaccine, with significantly fewer Mycobacterium tuberculosis in the lung and spleen than BCG Danish and BCG Japanese, although absolute differences in the organ burden of M. tuberculosis among these three vaccines were small (< or =0.2 log). BCG Tice and Pasteur were not significantly different. rBCG30, a recombinant BCG Tice vaccine overexpressing the M. tuberculosis 30 kDa major secretory protein (Antigen 85B), was more potent than any BCG vaccine (P < 0.0001 for differences in organ burden). Our study shows that late strains are not less potent than an early strain and argues against strain differences as a major factor in the variability of outcomes in BCG vaccine trials.


Asunto(s)
Vacuna BCG/inmunología , Mycobacterium bovis/inmunología , Tuberculosis/prevención & control , Animales , Antígenos Bacterianos/genética , Peso Corporal , Recuento de Colonia Microbiana , Cobayas , Pulmón/microbiología , Pulmón/patología , Masculino , Mycobacterium bovis/genética , Bazo/microbiología , Bazo/patología , Tuberculosis/inmunología , Tuberculosis/patología
17.
Vaccine ; 24(4): 443-51, 2006 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-16125825

RESUMEN

In previous studies, we have described a live recombinant BCG vaccine (rBCG30) overexpressing the 30 kDa major secretory protein of Mycobacterium tuberculosis that induces greater protective immunity against tuberculosis than the current vaccine in the demanding guinea pig model of pulmonary tuberculosis. In this study, we have investigated the impact of vaccine dose on the development of cell-mediated and protective immunity in the guinea pig model. We found that the protective efficacy against M. tuberculosis aerosol challenge of both BCG and rBCG30 was essentially dose-independent over a dose range of 10(1)-10(6) live organisms. As previously observed, rBCG30 was more potent, reducing colony-forming units (CFU) below the level observed in animals immunized with the parental BCG vaccine by 0.7 logs in the lungs and 1.0 logs in the spleen (P<0.0001). To gain a better understanding of the influence of dose on bacterial clearance and immunity, we assessed animals immunized with 10(1), 10(3), or 10(6)CFU of rBCG30. The higher the dose, the higher the peak CFU level achieved in animal organs. However, whereas humoral immune responses to the 30 kDa protein reflected the disparate CFU levels, cell-mediated immune responses did not; high and low doses of rBCG30 ultimately induced comparable peak lymphocyte proliferative responses and cutaneous delayed-type hypersensitivity responses to the 30 kDa protein. We estimate that the amount of the 30 kDa protein required to induce a strong cell-mediated immune response when delivered via 10 rBCG30 organisms is about 9 orders of magnitude less than that required when the protein is delivered in a conventional protein/adjuvant vaccine. This study demonstrates that a very low inoculum of rBCG30 organisms has the capacity to induce strong protective immunity against tuberculosis and that rBCG30 is an extremely potent delivery system for mycobacterial antigens.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacuna BCG/inmunología , Inmunidad Celular/efectos de los fármacos , Mycobacterium tuberculosis/inmunología , Vacunas Sintéticas/inmunología , Animales , Anticuerpos Antibacterianos/análisis , Antígenos Bacterianos/genética , Vacuna BCG/administración & dosificación , Vacuna BCG/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Cobayas , Tuberculosis/prevención & control , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología
18.
Vaccine ; 24(10): 1593-600, 2006 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-16257099

RESUMEN

Mycobacterium bovis infection of cattle and other domesticated animals exacts a significant economic toll in both economically developing and industrialized countries. Vaccination of herds and/or wild animals that share their grazing land and serve as reservoirs of infection has been proposed as a strategy to combat bovine tuberculosis. However, the only currently available vaccine, M. bovis Bacille Calmette-Guerin (BCG), is not highly efficacious. Here we show that a live recombinant vaccine, rBCG30, which expresses large amounts of the Mycobacterium tuberculosis 30 kDa major secretory protein, is more efficacious against bovine tuberculosis than BCG in the highly demanding guinea pig model of pulmonary tuberculosis. Compared with the parental wild-type BCG strain, rBCG30 administered intradermally induced significantly greater cell-mediated and humoral immune responses against the 30 kDa protein, as determined by measuring cutaneous delayed-type hypersensitivity and antibody titers. As for potency, in three independent experiments, rBCG30 induced greater protective immunity than BCG against aerosol challenge with a highly virulent strain of M. bovis, reducing the burden of M. bovis by 0.4 +/- 0.2 log colony-forming units (CFU) in the lung (P < 0.05) and by 1.1 +/- 0.4 log CFU in the spleen (P = 0.0005) below the level in BCG-immunized animals. A recombinant BCG vaccine overexpressing the identical M. bovis 30 kDa protein, rBCG30Mb, also induced greater cell-mediated and humoral immunity against the 30 kDa protein than BCG and greater protective immunity against M. bovis challenge; however, its potency was not significantly different from rBCG30. As rBCG30 is significantly more potent than BCG against M. bovis challenge, it has potential as a vaccine against bovine tuberculosis in domesticated animals and in wild animal reservoirs.


Asunto(s)
Vacuna BCG/inmunología , Tuberculosis Bovina/prevención & control , Vacunas Sintéticas/inmunología , Aerosoles , Animales , Bovinos , Cobayas , Inmunización , Hígado/patología , Masculino , Tuberculosis Bovina/patología
19.
Infect Immun ; 73(8): 4676-83, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16040980

RESUMEN

Tuberculosis continues to ravage humanity, killing 2 million people yearly. Most cases occur in areas of the world to which the disease is endemic, where almost everyone is vaccinated early in life with Mycobacterium bovis BCG, the currently available vaccine against tuberculosis. Thus, while more-potent vaccines are needed to replace BCG, new vaccines are also needed to boost the immune protection of the 4 billion people already vaccinated with BCG. Until now, no booster vaccine has been shown capable of significantly enhancing the level of protective immunity induced by BCG in the stringent guinea pig model of pulmonary tuberculosis, the "gold standard" for testing tuberculosis vaccines. In this paper, we describe a booster vaccine for BCG comprising the purified recombinant Mycobacterium tuberculosis 30-kDa protein, the major secreted protein of this pathogen. In the guinea pig model of pulmonary tuberculosis, boosting BCG-immunized animals once with the 30-kDa protein greatly increased cell-mediated and humoral immune responses to the protein in three consecutive experiments. Most importantly, boosting BCG-immunized animals once with the 30-kDa protein significantly enhanced protective immunity against aerosol challenge with highly virulent M. tuberculosis, as evidenced by a significantly reduced lung and spleen burden of M. tuberculosis compared with those for nonboosted BCG-immunized animals (mean additional reduction in CFU of 0.4 +/- 0.1 log in the lung [P = 0.03] and 0.6 +/- 0.1 log in the spleen [P = 0.002]). This study suggests that administering BCG-immunized people a booster vaccine comprising the 30-kDa protein may enhance their level of immunoprotection against tuberculosis.


Asunto(s)
Formación de Anticuerpos/inmunología , Vacuna BCG/inmunología , Proteínas Bacterianas/inmunología , Inmunización Secundaria , Tuberculosis/prevención & control , Animales , Peso Corporal/inmunología , Cobayas , Factores de Tiempo , Tuberculosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA