Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Malar J ; 15(1): 260, 2016 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-27150250

RESUMO

BACKGROUND: The potential emergence and spread of resistance to artemisinins in the Plasmodium falciparum malaria parasite constitutes a major global health threat. Hence, improving the efficacy of artemisinins and of artemisinin-based combination therapy (ACT) represents a major short-term goal in the global fight against malaria. Mice defective in the enzyme pantetheinase (Vnn3) show increased susceptibility to blood-stage malaria (increased parasitaemia, reduced survival), and supplementation of Vnn3 mutants with the reaction product of pantetheinase, cysteamine, corrects in part the malaria-susceptibility phenotype of the mutants. Cysteamine (Cys) is a small, naturally occurring amino-thiol that has very low toxicity in vivo and is approved for clinical use in the life-long treatment of the kidney disorder nephropathic cystinosis. METHODS: The ability of Cys to improve the anti-plasmodial activity of different clinically used artemisinins was tested. The effect of different CYS/ART combinations on malarial phenotypes (parasite blood-stage replication, overall and survival from lethal infection) was assessed in a series of in vivo experiments using Plasmodium strains that induce either blood-stage (Plasmodium chabaudi AS) or cerebral disease (Plasmodium berghei ANKA). This was also evaluated in an ex vivo experimental protocol that directly assesses the effect of such drug combinations on the viability of Plasmodium parasites, as measured by the ability of tested parasites to induce a productive infection in vivo in otherwise naïve animals. RESULTS: Cys is found to potentiate the anti-plasmodial activity of artesunate, artemether, and arteether, towards the blood-stage malaria parasite P. chabaudi AS. Ex vivo experiments, indicate that potentiation of the anti-plasmodial activity of artemisinins by Cys is direct and does not require the presence of host factors. In addition, potentiation occurs at sub-optimal concentrations of artemisinins and Cys that on their own have little or no effect on parasite growth. Cys also dramatically enhances the efficacy and protective effect of artemisinins against cerebral malaria induced by infection with the P. berghei ANKA parasite. CONCLUSION: These findings indicate that inclusion of Cys in current formulations of ACT, or its use as adjunct therapy could improve the anti-plasmodial activity of artemisinin, decrease mortality in cerebral malaria patients, and prevent or delay the development and spread of artemisinin resistance.


Assuntos
Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Cisteamina/administração & dosagem , Sinergismo Farmacológico , Malária/tratamento farmacológico , Animais , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Malária Cerebral/tratamento farmacológico , Camundongos , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/fisiologia , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium chabaudi/fisiologia , Análise de Sobrevida , Resultado do Tratamento
2.
J Antimicrob Chemother ; 71(7): 1892-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27084920

RESUMO

OBJECTIVES: This study evaluated the pharmacokinetic properties of oleylphosphocholine (OlPC) in hamsters following a single oral dose. Its prophylactic activity was tested to establish exposure-activity relationships, while a 5 + 5 day oral regimen at 20 mg/kg with long post-treatment follow-up was performed to assess its curative potential. METHODS: Single oral doses of 20, 50 and 100 mg/kg were administered for pharmacokinetic analysis while a 100 mg/kg single oral dose was given on day 7, 4 or 1, or 4 h prior to infection in the prophylactic efficacy study. The animals were infected on day 0 with Leishmania infantum and the resulting parasite burdens were measured in target organs on day 21. In the curative model, treatment started on day 21 post-infection at 20 mg/kg for 5 + 5 days and amastigote burdens were determined in target organs either on day 42 [10 days after the end of treatment (dpt)] or day 72 (40 dpt). RESULTS: OlPC showed elimination t1/2 of ∼50 h and dose-proportional exposure. The prophylactic action of OlPC was in agreement with model-simulated drug exposures, showing dose-dependent residual activity. Interestingly, the 100 mg/kg single dose administered 4 days before infection (day -4) still reduced the overall parasite burden by ∼50%. In the curative model, >99% clearance of infection was observed at 10 dpt in all OlPC-treated animals and remained so at 40 dpt. CONCLUSIONS: This study reveals that total plasma exposure (AUCt-∞) correlates well with the prophylactic and curative efficacy of OlPC in the L. infantum hamster model.


Assuntos
Antiprotozoários/farmacologia , Antiprotozoários/farmacocinética , Leishmania infantum/efeitos dos fármacos , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/prevenção & controle , Fosforilcolina/análogos & derivados , Administração Oral , Animais , Antiprotozoários/administração & dosagem , Área Sob a Curva , Quimioprevenção/métodos , Modelos Animais de Doenças , Feminino , Mesocricetus , Fosforilcolina/administração & dosagem , Fosforilcolina/farmacocinética , Fosforilcolina/farmacologia , Plasma/química
3.
Front Microbiol ; 6: 973, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26441906

RESUMO

Cryptosporidium parvum is a species of protozoa that causes cryptosporidiosis, an intestinal disease affecting many mammals including humans. Typically, in healthy individuals, cryptosporidiosis is a self-limiting disease. However, C. parvum can cause a severe and persistent infection that can be life-threatening for immunocompromised individuals, such as AIDS patients. As there are no available treatments for these patients that can cure the disease, there is an urgent need to identify treatment options. We tested the anti-parasitic activity of the alkylphosphocholine oleylphosphocholine (OlPC), an analog of miltefosine, against C. parvum in in vitro and in vivo studies. In vitro experiments using C. parvum infected human ileocecal adenocarcinoma cells (HCT-8 cells) showed that OlPC has an EC50 of 18.84 nM. Moreover, no cell toxicity has been seen at concentrations ≤50 µM. C57BL/6 interferon gamma receptor knock-out mice, were infected by gavage with 4000 C. parvum oocysts on Day 0. Oral treatments, with OlPC, miltefosine, paromomycin or PBS, began on Day 3 post-infection for 10 days. Treatment with OlPC, at 40 mg/kg/day resulted in 100% survival, complete clearance of parasite in stools and a 99.9% parasite burden reduction in the intestines at Day 30. Doses of 30 and 20 mg/kg/day also demonstrated an increased survival rate and a dose-dependent parasite burden reduction. Mice treated with 10 mg/kg/day of miltefosine resulted in 50% survival at Day 30. In contrast, control mice, treated with PBS or 100 mg/kg/day of paromomycin, died or had to be euthanized between Days 6 and 13 due to severe illness. Results of parasite burden were obtained by qPCR and cross-validated by both flow cytometry of stool oocysts and histological sections of the ileum. Together, our results strongly support that OlPC represents a potential candidate for the treatment of C. parvum infections in immunocompromised patients.

4.
Mycoses ; 58(3): 127-32, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25590577

RESUMO

Invasive aspergillosis (IA) has become increasingly common and is characterised by high morbidity and mortality. Upcoming resistance threatens treatment with azoles and highlights the continuous need for novel therapeutics. This laboratory study investigated the in vitro and in vivo potential of the alkylphospholipid oleylphosphocholine (OlPC) against Aspergillus. In vitro activities of OlPC, miltefosine, posaconazole and voriconazole were determined for Aspergillus fumigatus, A. niger, A. terreus and A. flavus. In vivo efficacy of OlPC was evaluated in a systemic A. fumigatus mouse model, adopting a short-term and long-term oral or intraperitoneal dosing regimen. OlPC showed good in vitro activity against A. fumigatus (IC50 = 1.04 µmol l(-1)). Intraperitoneal administration of 50 mg kg(-1) day(-1) OlPC significantly reduced the fungal organ burdens at 4 days post-infection (dpi). Although 5- and 10-day OlPC treatment improved survival, organ burdens were not affected at 10 and 15 dpi. While this study showed excellent in vitro activity of OlPC against Aspergillus spp., its therapeutic efficacy in an acute mouse model for IA was less convincing. Given the limited therapeutic options in the current antifungal market for invasive infections, OlPC activity should be assessed in a less stringent in vivo model, potentially in combination treatment with other already marketed antifungal drugs.


Assuntos
Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Aspergilose/tratamento farmacológico , Aspergillus/efeitos dos fármacos , Fosforilcolina , Pirimidinas/uso terapêutico , Animais , Aspergilose/microbiologia , Aspergillus fumigatus/efeitos dos fármacos , Azóis/uso terapêutico , Modelos Animais de Doenças , Camundongos , Testes de Sensibilidade Microbiana , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacologia , Fosforilcolina/uso terapêutico , Triazóis/farmacologia , Voriconazol/farmacologia
5.
PLoS Negl Trop Dis ; 8(9): e3144, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25210745

RESUMO

BACKGROUND: Cutaneous leishmaniasis (CL) represents a range of skin diseases caused by infection with Leishmania parasites and associated with tissue inflammation and skin ulceration. CL is clinically widespread in both the Old and New World but lacks treatments that are well tolerated, effective and inexpensive. Oleylphosphocholine (OlPC) is a new orally bioavailable drug of the alkylphosphocholine family with potent antileishmanial activity against a broad range of Leishmania species/strains. METHODOLOGY/PRINCIPAL FINDINGS: The potential of OlPC against Old World CL was evaluated in a mouse model of Leishmania (L.) major infection in BALB/c mice. Initial dose-response experiments showed that an oral daily dose of 40 mg/kg of OlPC was needed to impact time to cure and lesion sizes. This dose was then used to directly compare the efficacy of OlPC to the efficacy of the antileishmanial drugs miltefosine (40 mg/kg/day), fluconazole (160 mg/kg/day) and amphotericin B (25 mg/kg/day). OlPC, miltefosine and fluconazole were given orally for 21 days while amphotericin B was administered intraperitoneally for 10 days. Ulcer sizes and animal weights were followed up on a weekly basis and parasitemia was determined by means of a real-time in vivo imaging system which detects luminescence emitted from luciferase-expressing infecting L. major parasites. Amphotericin B and OlPC showed excellent efficacy against L. major lesions in terms of reduction of parasitic loads and by inducing complete healing of established lesions. In contrast, treatment with miltefosine did not significantly affect parasitemia and lesion sizes, while fluconazole was completely ineffective at the dose regimen tested. CONCLUSIONS/SIGNIFICANCE: Given the data showing the outstanding efficacy and tolerability of OlPC, our results suggest that OlPC is a promising new drug candidate to improve and simplify current clinical management of L. major CL.


Assuntos
Antiprotozoários/efeitos adversos , Antiprotozoários/farmacologia , Leishmania major/efeitos dos fármacos , Leishmaniose Cutânea/tratamento farmacológico , Fosfatidilcolinas/efeitos adversos , Fosfatidilcolinas/farmacologia , Fosforilcolina/análogos & derivados , Administração Oral , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Fosforilcolina/efeitos adversos , Fosforilcolina/farmacologia
6.
Global Health ; 10: 27, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24735677

RESUMO

BACKGROUND: The grand challenges approach aims to spark innovative and transformative strategies to overcome barriers to significant global health issues. Grand Challenges Canada endorses an 'Integrated Innovation™' approach that focuses on the intersection of scientific/technological, social and business innovation. In this article we explore themes emerging from a dialogue between the authors, who are multidisciplinary recipients of the 'Rising Stars in Global Health' award from Grand Challenges Canada, regarding benefits of engaging in integrated innovation research, and recommendations for how this approach may develop in the future. DISCUSSION: Our dialogue followed a semi-structured interview format that addressed three topics: 1) reflections on applying an Integrated Innovation™ approach for global health; 2) thoughts on participation in the Grand Challenges 2012 meeting; and 3) authors' visions of Grand Challenges Canada and the Grand Challenge movement towards 2020. The dialogue was transcribed verbatim and we used thematic analysis techniques to identify, analyze and report themes in the data. Benefits of working using the Grand Challenges approach centered on two themes: a) the potential for scientific breakthrough and b) building interdisciplinary collaborations and a community of scholars. Challenges and opportunities for Grand Challenges in moving forward included: a) capacity building, particularly regarding Integrated Innovation™ and scale-up planning; b) interdisciplinary and international mentorship for new investigators; and c) potential for future commercialization. CONCLUSIONS: Our discussion highlighted that Integrated Innovation™ offers the opportunity to develop new theories, methods and approaches to global health while simultaneously fostering a collaborative spirit grounded in international, interdisciplinary collaborations. However, the arguable over-emphasis on corporatization poses a major challenge for new investigators. We propose a more balanced way forward that can harness technology to foster mentorship across time and space to support the development of such skills and ideas among new investigators.


Assuntos
Comportamento Cooperativo , Saúde Global , Pesquisa/organização & administração , Pesquisa Biomédica/organização & administração , Canadá , Humanos , Inovação Organizacional , Pesquisadores , Seguridade Social
7.
Sci Rep ; 4: 4156, 2014 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-24561704

RESUMO

This study establishes a proof-of-concept that a tattoo device can target intra-dermal drug delivery against cutaneous leishmaniasis (CL). The selected drug is oleylphosphocholine (OlPC) formulated as liposomes, particles known to be prone to macrophage ingestion. We first show that treatment of cultured Leishmania-infected macrophages with OlPC-liposomes results in a direct dose-dependent killing of intracellular parasites. Based on this, in vivo efficacy is demonstrated using a 10 day tattooing-mediated treatment in mice infected with L. major and L. mexicana. In both models this regimen results in rapid clinical recovery with complete regression of skin lesions by Day 28. Parasite counts and histopathology examination confirm high treatment efficacy at the parasitic level. Low amount of drug required for tattooing combined with fast clinical recovery may have a positive impact on CL patient management. This first example of tattoo-mediated drug delivery could open to new therapeutic interventions in the treatment of skin diseases.


Assuntos
Leishmaniose Cutânea/tratamento farmacológico , Lipossomos/química , Fosfatidilcolinas/administração & dosagem , Tatuagem , Animais , Linhagem Celular , Feminino , Leishmania/isolamento & purificação , Leishmaniose Cutânea/parasitologia , Leishmaniose Cutânea/patologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilcolinas/química
8.
Parasitol Res ; 113(1): 157-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24192865

RESUMO

The alkylphosphocholine oleylphosphocholine (OlPC) represents a potential new therapy for the treatment of canine leishmaniosis caused by Leishmania infantum. The aim of the present study was to evaluate the efficacy and safety of OlPC in a small cohort of dogs naturally infected with L. infantum and defined as clinically sick (LeishVet stages II and III). A total of eight dogs were included in the study and were treated orally with 4 mg/kg OlPC for 14 days. Dogs were assessed at the clinical and parasitological level at four time points during a total follow-up period of 90 days (before treatment and at 15, 30, and 90 days post-treatment onset). Ln-PCR, real-time quantitative PCR, antibody testing (IFAT), and culture of bone marrow aspirates were evaluated at the four time points. OlPC treatment induced a rapid and satisfactory clinical recovery in terms of clinical score reduction and weight gain, and treatment efficacy was found to be associated with a decrease in bone marrow parasitic load. Serological titers measured by IFAT were stable in any of the treated dogs at any time point after treatment. OlPC was well tolerated and no severe adverse events were noted in any of the treated dogs; even some dogs showed slight intestinal disorders. This proof-of-principle study is the first to show that short oral treatment with OlPC improves clinical signs of canine L. infantum leishmaniosis, highlighting the need to perform additional studies to optimize the dosing regimen and to assess long-term treatment efficacy of this drug.


Assuntos
Antiprotozoários/uso terapêutico , Doenças do Cão/tratamento farmacológico , Cães/parasitologia , Leishmaniose Visceral/veterinária , Fosforilcolina/análogos & derivados , Animais , Medula Óssea/parasitologia , Doenças do Cão/diagnóstico , Doenças do Cão/parasitologia , Feminino , Leishmania infantum/genética , Leishmania infantum/imunologia , Leishmaniose Visceral/diagnóstico , Leishmaniose Visceral/tratamento farmacológico , Masculino , Carga Parasitária , Fosforilcolina/uso terapêutico , Projetos Piloto , Reação em Cadeia da Polimerase/veterinária
9.
Physiol Genomics ; 44(17): 843-52, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22805347

RESUMO

Epidemiological studies show that high HDL-cholesterol (HDLc) decreases the risk of cardiovascular disease. To map genes controlling lipid metabolism, particularly HDLc levels, we screened the plasma lipids of 36 AcB/BcA RC mouse strains subjected to either a normal or a high-fat/cholesterol diet. Strains BcA68 and AcB65 showed deviant HDLc plasma levels compared with the parental A/J and C57BL/6J strains; they were thus selected to generate informative F2 crosses. Linkage analyses in the AcB65 strain identified a locus on chromosome 4 (Hdlq78) responsible for high post-high fat diet HDLc levels. This locus has been previously associated at genome-wide significance to two regions in the human genome. A second linkage analysis in strain BcA68 identified linkage in the vicinity of a gene cluster known to control HDLc levels. Sequence analysis of these candidates identified a de novo, loss-of-function mutation in the ApoA1 gene of BcA68 that prematurely truncates the ApoA1 protein. The possibility of dissecting the specific effects of this new ApoA1 deficiency in the context of isogenic controls makes the BcA68 mouse a valuable new tool.


Assuntos
Apolipoproteína A-I/genética , HDL-Colesterol/sangue , HDL-Colesterol/genética , Dieta Hiperlipídica , Camundongos Congênicos/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Cruzamentos Genéticos , Loci Gênicos/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação/genética , Análise de Sequência de DNA , Especificidade da Espécie
10.
J Antimicrob Chemother ; 67(11): 2707-12, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22782488

RESUMO

OBJECTIVES: The alkylphospholipid oleylphosphocholine (OlPC) is a structural analogue of miltefosine and may represent a potential therapeutic backup for the treatment of visceral leishmaniasis (VL). This laboratory study compared the in vitro and in vivo activity profile of both OlPC and miltefosine. METHODS: The in vitro potency of OlPC was compared with that of miltefosine, amphotericin B, paromomycin and pentavalent antimony (Sb(V)) using the intracellular amastigote assay on different Old World and New World Leishmania species. The in vivo efficacy was dose titrated in the Leishmania infantum hamster model after infection with 2 × 10(7) amastigotes (day 0) and oral treatment at day 21 using an aqueous (OlPC/H(2)O) and liposomal formulation of OlPC in single and repeated (5 day) oral dosing regimens. The amastigote reductions in the liver, spleen and bone marrow were assessed (day 35). RESULTS: The in vitro activity of OlPC against Leishmania donovani, L. infantum, Leishmania tropica, Leishmania mexicana and Leishmania panamensis showed mean IC(50) values <5 µM, while the IC(50) values for Leishmania major and Leishmania braziliensis were 7.7 and 13.5 µM, respectively. These results are fairly similar to those obtained for miltefosine. In the hamster model, treatment with 20 and 40 mg/kg for 5 days proved that both OlPC formulations were equipotent and showed a markedly higher efficacy compared with miltefosine. A single dosing of 100 mg/kg of OlPC/H(2)O or OlPC liposomes reduced the parasite burdens by 96.2% and 99.3% in liver, 99.8% and 99.9% in spleen, and 87.6% and 96.9% in bone marrow, respectively. No signs of toxicity or adverse drug-related effects were noted. CONCLUSIONS: These data suggest that OlPC may become a promising candidate to improve and simplify current case management of VL. Additional pharmacological and pharmacokinetic studies are ongoing to assess the full potential of OlPC as a 'drug candidate'.


Assuntos
Antiprotozoários/administração & dosagem , Antiprotozoários/efeitos adversos , Leishmaniose Visceral/tratamento farmacológico , Fosfatidilcolinas/administração & dosagem , Fosfatidilcolinas/efeitos adversos , Administração Oral , Animais , Antiprotozoários/farmacologia , Medula Óssea/parasitologia , Cricetinae , Modelos Animais de Doenças , Feminino , Concentração Inibidora 50 , Fígado/parasitologia , Mesocricetus , Carga Parasitária , Testes de Sensibilidade Parasitária , Fosfatidilcolinas/farmacologia , Baço/parasitologia
11.
PLoS Negl Trop Dis ; 5(12): e1435, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22206032

RESUMO

Human neurocysticercosis (NC) caused by Taenia solium is a parasitic disease of the central nervous system that is endemic in many developing countries. In this study, a genetic approach using the murine intraperitoneal cysticercosis caused by the related cestode Taenia crassiceps was employed to identify host factors that regulate the establishment and proliferation of the parasite. A/J mice are permissive to T. crassiceps infection while C57BL/6J mice (B6) are comparatively restrictive, with a 10-fold difference in numbers of peritoneal cysticerci recovered 30 days after infection. The genetic basis of this inter-strain difference was explored using 34 AcB/BcA recombinant congenic strains derived from A/J and B6 progenitors, that were phenotyped for T. crassiceps replication. In agreement with their genetic background, most AcB strains (A/J-derived) were found to be permissive to infection while most BcA strains (B6-derived) were restrictive with the exception of a few discordant strains, together suggesting a possible simple genetic control. Initial haplotype association mapping using >1200 informative SNPs pointed to linkages on chromosomes 2 (proximal) and 6 as controlling parasite replication in the AcB/BcA panel. Additional linkage analysis by genome scan in informative [AcB55xDBA/2]F1 and F2 mice (derived from the discordant AcB55 strain), confirmed the effect of chromosome 2 on parasite replication, and further delineated a major locus (LOD = 4.76, p<0.01; peak marker D2Mit295, 29.7 Mb) that we designate Tccr1 (T. crassiceps cysticercosis restrictive locus 1). Resistance alleles at Tccr1 are derived from AcB55 and are inherited in a dominant fashion. Scrutiny of the minimal genetic interval reveals overlap of Tccr1 with other host resistance loci mapped to this region, most notably the defective Hc/C5 allele which segregates both in the AcB/BcA set and in the AcB55xDBA/2 cross. These results strongly suggest that the complement component 5 (C5) plays a critical role in early protective inflammatory response to infection with T. crassiceps.


Assuntos
Complemento C5/genética , Complemento C5/imunologia , Cisticercose/genética , Cisticercose/imunologia , Taenia/imunologia , Animais , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Estudos de Associação Genética , Haplótipos , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único
12.
Nat Neurosci ; 14(12): 1569-73, 2011 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-22019732

RESUMO

Quantitative trait locus mapping of chemical/inflammatory pain in the mouse identified the Avpr1a gene, which encodes the vasopressin-1A receptor (V1AR), as being responsible for strain-dependent pain sensitivity to formalin and capsaicin. A genetic association study in humans revealed the influence of a single nucleotide polymorphism (rs10877969) in AVPR1A on capsaicin pain levels, but only in male subjects reporting stress at the time of testing. The analgesic efficacy of the vasopressin analog desmopressin revealed a similar interaction between the drug and acute stress, as desmopressin inhibition of capsaicin pain was only observed in nonstressed subjects. Additional experiments in mice confirmed the male-specific interaction of V1AR and stress, leading to the conclusion that vasopressin activates endogenous analgesia mechanisms unless they have already been activated by stress. These findings represent, to the best of our knowledge, the first explicit demonstration of analgesic efficacy depending on the emotional state of the recipient, and illustrate the heuristic power of a bench-to-bedside-to-bench translational strategy.


Assuntos
Analgésicos/uso terapêutico , Limiar da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Dor/genética , Dor/fisiopatologia , Vasopressinas/uso terapêutico , Animais , Animais Recém-Nascidos , Capsaicina/efeitos adversos , Desamino Arginina Vasopressina/uso terapêutico , Modelos Animais de Doenças , Feminino , Estudos de Associação Genética , Habituação Psicofisiológica/efeitos dos fármacos , Habituação Psicofisiológica/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peso Molecular , Dor/induzido quimicamente , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Limiar da Dor/fisiologia , Polimorfismo de Nucleotídeo Único/genética , Locos de Características Quantitativas , Receptores de Vasopressinas/deficiência , Receptores de Vasopressinas/genética , Fatores Sexuais , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia
14.
N Engl J Med ; 365(2): 127-38, 2011 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-21524210

RESUMO

BACKGROUND: The genetic analysis of human primary immunodeficiencies has defined the contribution of specific cell populations and molecular pathways in the host defense against infection. Disseminated infection caused by bacille Calmette-Guérin (BCG) vaccines is an early manifestation of primary immunodeficiencies, such as severe combined immunodeficiency. In many affected persons, the cause of disseminated BCG disease is unexplained. METHODS: We evaluated an infant presenting with features of severe immunodeficiency, including early-onset disseminated BCG disease, who required hematopoietic stem-cell transplantation. We also studied two otherwise healthy subjects with a history of disseminated but curable BCG disease in childhood. We characterized the monocyte and dendritic-cell compartments in these three subjects and sequenced candidate genes in which mutations could plausibly confer susceptibility to BCG disease. RESULTS: We detected two distinct disease-causing mutations affecting interferon regulatory factor 8 (IRF8). Both K108E and T80A mutations impair IRF8 transcriptional activity by disrupting the interaction between IRF8 and DNA. The K108E variant was associated with an autosomal recessive severe immunodeficiency with a complete lack of circulating monocytes and dendritic cells. The T80A variant was associated with an autosomal dominant, milder immunodeficiency and a selective depletion of CD11c+CD1c+ circulating dendritic cells. CONCLUSIONS: These findings define a class of human primary immunodeficiencies that affect the differentiation of mononuclear phagocytes. They also show that human IRF8 is critical for the development of monocytes and dendritic cells and for antimycobacterial immunity. (Funded by the Medical Research Council and others.).


Assuntos
Células Dendríticas/imunologia , Síndromes de Imunodeficiência/genética , Fatores Reguladores de Interferon/genética , Mutação , Adolescente , Adulto , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos , Vacina BCG/genética , Vacina BCG/imunologia , Feminino , Genes Dominantes , Humanos , Lactente , Fatores Reguladores de Interferon/deficiência , Interleucina-12/biossíntese , Leucócitos Mononucleares/imunologia , Masculino , Modelos Moleculares , Infecções por Mycobacterium/genética , Infecções por Mycobacterium/imunologia , Linhagem , Conformação Proteica , Alinhamento de Sequência
15.
Eur J Clin Pharmacol ; 67(3): 267-75, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20949263

RESUMO

OBJECTIVE: The aim of the present study was to compare the pharmacokinetic properties, bioavailability and tolerability of artesunate (AS) and amodiaquine (AQ) administered as a fixed-dose combination (Amonate FDC tablets; Dafra Pharma, Turnhout, Belgium) or as a non-fixed dose combination of separate AS tablets (Arsuamoon; Guilin Pharmaceutical Co, Shanghai, China) and AQ tablets (Flavoquine; Sanofi-Aventis, Paris, France). METHODS: This was a randomized, open label, two-period, two-treatment, two-sequence, cross-over study in which 60 healthy male Indian volunteers were given a single total oral dose of 100 mg AS and 400 mg AQ hydrochloride either as two tablets of Amonate FDC (AS 50 mg and AQ hydrochloride 200 mg) or as two AS tablets of the co-blister Arsuamoon (50 mg AS) together with two Flavoquine tablets (200 mg AQ hydrochloride). Plasma AS and blood AQ concentrations, as well as those of their respective active metabolites dihydroartemisinin (DHA) and desethylamodiaquine (DEAQ), were measured by high-performance liquid chromatography-tandem spectrometry. The pharmacokinetic parameters of AS, DHA, AQ and DEAQ were determined by non-compartmental analysis. Bioequivalence assessment was performed by analysis of variance (ANOVA), and calculation of the 90% confidence intervals of the geometric mean ratio test (fixed)/reference (non-fixed) for AUC(0-t) and C(max) for AS, AQ, DHA and DEAQ. RESULTS: Interim analysis showed that both treatments were not bioequivalent; therefore, statistical analysis was carried out on the results of all subjects for whom blood/plasma concentrations were available for all four analytes (n=26). The C(max) (maximum plasma/blood concentration) of AS was 67.0 ± 37.1 and 154.8 ± 116.2 ng/mL for the fixed-dose and non-fixed dose administration, respectively. The AUC(0-t) (area under the plasma concentration-time curve from time zero to the last measurable concentration) of AS was 60.1 ± 27.5 and 81.8 ± 44.3 ng h/mL for the fixed-dose and non-fixed dose administration, respectively. The 90% confidence intervals for C(max) and AUC(0-t) of AS were outside the 80-125% acceptance range: 37.02-61.62% and 70.10-83.47%, respectively. The C(max) of AQ was 33.8 ± 13.6 and 31.4 ± 14.1 ng/mL for the fixed-dose and non-fixed dose administration, respectively. The AUC(0-t) of AQ was 332.3 ± 116.6 and 329.8 ± 99.5 ng h/mL for the fixed and non-fixed dose administration, respectively. For AQ, the 90% CIs for C(max) and AUC(0-t) were within the 80-125% acceptance range: 99.17-121.71 and 89.53-107.35%, respectively. Bioequivalence assessment based on the active metabolite data supported the bioequivalence conclusions based on the parent compound data. Both the fixed-dose and non-fixed dose administration of 100 mg AS and 400 mg AQ were well tolerated. CONCLUSION: Bioequivalence of the fixed-dose AS/AQ formulation with the non-fixed dose combination of the same drugs was not demonstrated for AS, but it was shown for AQ for both C(max) and AUC(0-t). The results obtained on the active metabolites support this conclusion. Overall, the fixed-dose 50 mg AS/200 mg AQ tablets were not technically bioequivalent with 50 mg AS tablets and 200 mg AQ tablets administered separately. The difference cannot be explained by the pharmaceutical properties of the tablets and seems to be biologically related.


Assuntos
Amodiaquina/farmacocinética , Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Administração Oral , Adolescente , Adulto , Amodiaquina/administração & dosagem , Amodiaquina/análogos & derivados , Análise de Variância , Antimaláricos/administração & dosagem , Área Sob a Curva , Artemisininas/administração & dosagem , Artesunato , Disponibilidade Biológica , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Quimioterapia Combinada , Humanos , Índia , Masculino , Pessoa de Meia-Idade , Comprimidos , Espectrometria de Massas em Tandem , Equivalência Terapêutica , Adulto Jovem
16.
Mamm Genome ; 22(1-2): 32-42, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21116636

RESUMO

Malaria is a disease that infects over 500 million people, causing at least 1 million deaths every year, with the majority occurring in developing countries. The current antimalarial arsenal is becoming dulled due to the rapid rate of resistance of the parasite. However, in populations living in malaria-endemic regions there are many examples of genetic-based resistance to the severe effects of the parasite Plasmodium. Defining the genetic factors behind host resistance has been an area of great scientific interest over the last few decades; this review summarizes the current knowledge of the genetic loci involved. Perhaps the lessons learned from the natural variation in both the human populations and experimental mouse models of infection may pave the way for novel resistance-proof antimalarials.


Assuntos
Modelos Animais de Doenças , Imunidade Inata , Malária/genética , Malária/imunologia , Camundongos , Animais , Suscetibilidade a Doenças , Humanos , Malária/parasitologia , Plasmodium/imunologia , Plasmodium/fisiologia
17.
Antimicrob Agents Chemother ; 54(8): 3262-70, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20479197

RESUMO

Malaria continues to be a major threat to global health. Artemisinin combination therapy (ACT) is the recommended treatment for clinical malaria; however, recent reports of parasite resistance to artemisinin in certain areas where malaria is endemic have stressed the need for developing more efficacious ACT. We report that cysteamine (Cys), the aminothiol used to treat nephropathic cystinosis in humans, strongly potentiates the efficacy of artemisinin against the Plasmodium parasite in vivo. Using a mouse model of infection with Plasmodium chabaudi AS, we observe that Cys dosing used to treat cystinosis in humans can strongly potentiate (by 3- to 4-fold) the antimalarial properties of the artemisinin derivatives artesunate and dihydroartemisinin. Addition of Cys to suboptimal doses of artemisinin delays the appearance of blood parasitemia, strongly reduces the extent of parasite replication, and significantly improves survival in a model of lethal P. chabaudi infection. Cys, the natural product of the enzyme pantetheinase, has a history of safe use for the clinical management of cystinosis. Our findings suggest that Cys could be included in novel ACTs to improve efficacy against Plasmodium parasite replication, including artemisinin-resistant isolates. Future work will include clinical evaluation of novel Cys-containing ACTs and elucidation of the mechanism underlying the potentiation effect of Cys.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Cisteamina/uso terapêutico , Cistinose/tratamento farmacológico , Malária/tratamento farmacológico , Plasmodium chabaudi/efeitos dos fármacos , Animais , Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Cisteamina/administração & dosagem , Cisteamina/farmacocinética , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Malária/mortalidade , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium chabaudi/crescimento & desenvolvimento
19.
Exp Parasitol ; 125(4): 315-24, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20219464

RESUMO

In mice, loss of pantetheinase activity causes susceptibility to infection with Plasmodium chabaudi AS. Treatment of mice with the pantetheinase metabolite cysteamine reduces blood-stage replication of P. chabaudi and significantly increases survival. Similarly, a short exposure of Plasmodium to cysteamine ex vivo is sufficient to suppress parasite infectivity in vivo. This effect of cysteamine is specific and not observed with a related thiol (dimercaptosuccinic acid) or with the pantethine precursor of cysteamine. Also, cysteamine does not protect against infection with the parasite Trypanosoma cruzi or the fungal pathogen Candida albicans, suggesting cysteamine acts directly against the parasite and does not modulate host inflammatory response. Cysteamine exposure also blocks replication of P. falciparum in vitro; moreover, these treated parasites show higher levels of intact hemoglobin. This study highlights the in vivo action of cysteamine against Plasmodium and provides further evidence for the involvement of pantetheinase in host response to this infection.


Assuntos
Antimaláricos/farmacologia , Cisteamina/farmacologia , Malária/tratamento farmacológico , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Amidoidrolases/metabolismo , Animais , Antimaláricos/uso terapêutico , Candidíase/tratamento farmacológico , Doença de Chagas/tratamento farmacológico , Cloroquina/farmacologia , Cisteamina/uso terapêutico , Citocinas/sangue , Citocinas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Proteínas Ligadas por GPI , Hemoglobinas/metabolismo , Humanos , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/tratamento farmacológico , Parasitemia/parasitologia , Plasmodium falciparum/metabolismo , Trypanosoma cruzi/efeitos dos fármacos
20.
Hum Mol Genet ; 19(3): 434-44, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19880857

RESUMO

IFN-gammaR1 deficiency is a genetic etiology of Mendelian susceptibility to mycobacterial diseases, and includes two forms of complete recessive deficiency, with or without cell surface expression, and two forms of partial deficiency, dominant or recessive. We report here a novel form of partial and recessive Interferon gamma receptor 1 (IFN-gammaR1) deficiency, which is almost as severe as complete deficiency. The patient is homozygous for a mutation of the initiation codon (M1K). No detectable expression and function of IFN-gammaR1 were found in the patient's fibroblasts. However, IFN-gammaR1 expression was found to be impaired, but not abolished, on the EBV-transformed B cells, which could respond weakly to IFN-gamma. The mechanism underlying this weak expression involves leaky translation initiation at both non-AUG codons and the third AUG codon at position 19. It results in the residual expression of IFN-gammaR1 protein of normal molecular weight and function. The residual IFN-gamma signaling documented in this novel form of partial IFN-gammaR1 deficiency was not ubiquitous and was milder than that seen in other forms of partial IFN-gammaR1 deficiency, accounting for the more severe clinical phenotype of the patient, which was almost as severe as that of patients with complete deficiency.


Assuntos
Códon de Iniciação , Mutação em Linhagem Germinativa , Infecções por Mycobacterium/genética , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Linfócitos B/metabolismo , Linhagem Celular , Células Cultivadas , Criança , Feminino , Fibroblastos/metabolismo , Predisposição Genética para Doença , Humanos , Interferon gama/metabolismo , Mutação de Sentido Incorreto , Infecções por Mycobacterium/metabolismo , Infecções por Mycobacterium/microbiologia , Especificidade da Espécie , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA