Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Med ; 10(3): 1128-1140, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33347715

RESUMO

PURPOSE: Despite the availability of new drugs, many patients with acute myeloid leukemia (AML) do not achieve remission and outcomes remain poor. Venetoclax is a promising new therapy approved for use in combination with a hypomethylating agent or with low-dose cytarabine for the treatment of newly diagnosed older AML patients or those ineligible for intensive chemotherapy. 225 Actinium-lintuzumab (225 Ac-lintuzumab) is a clinical stage radioimmunotherapy targeting CD33 that has shown evidence of single-agent activity in relapsed/refractory AML. Increased expression of MCL-1 is a mediator of resistance to venetoclax in cancer. EXPERIMENTAL DESIGN: Here we investigated the potential for 225 Ac-lintuzumab-directed DNA damage to suppress MCL-1 levels as a possible mechanism of reversing resistance to venetoclax in two preclinical in vivo models of AML. RESULTS: We demonstrated that 225 Ac-lintuzumab in combination with venetoclax induced a synergistic increase in tumor cell killing compared to treatment with either drug alone in venetoclax-resistant AML cell lines through both an induction of double-stranded DNA breaks (DSBs) and depletion of MCL-1 protein levels. Further, this combination led to significant tumor growth control and prolonged survival benefit in venetoclax-resistant in vivo AML models. CONCLUSIONS: There results suggest that the combination of 225 Ac-lintuzumab with venetoclax is a promising therapeutic strategy for the treatment of patients with venetoclax-resistant AML. Clinical trial of this combination therapy (NCT03867682) is currently ongoing.


Assuntos
Actínio/química , Anticorpos Monoclonais Humanizados/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Sulfonamidas/farmacologia , Animais , Anticorpos Monoclonais Humanizados/química , Antineoplásicos/farmacologia , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacologia , Apoptose , Proliferação de Células , Feminino , Humanos , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Oncotarget ; 11(39): 3571-3581, 2020 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-33062193

RESUMO

Chimeric antigen receptor (CAR) T cell therapies, and adoptive cell therapy (ACT) in general, represent one of the most promising anti-cancer strategies. Conditioning has been shown to improve the immune homeostatic environment to enable successful ACT or CAR-T engraftment and expansion in vivo following infusion, and represents potential point of intervention to decrease serious toxicities following CAR-T treatment. In contrast to relatively non-specific chemotherapy-derived lymphodepletion, targeted lymphodepletion with radioimmunotherapy (RIT) directed to CD45 may be a safer and more effective alternative to target and deplete immune cells. Here we describe the results of preclinical studies with an anti-mouse CD45 antibody 30F11, labeled with two different beta-emitters 131Iodine (131I) and 177Lutetium (177Lu), to investigate the effect of anti-CD45 RIT lymphodepletion on immune cell types and on tumor control in a model of adoptive cell therapy. Treatment of mice with 3.7 MBq 131I-30F11 or 1.48 MBq 177Lu-30F11 safely depleted immune cells such as spleen CD4+ and CD8+ T Cells, B and NK cells as well as Tregs in OT I tumor model while sparing RBC and platelets and enabled E. G7 tumor control. Our results support the application of CD45-targeted RIT lymphodepletion with a non-myeloablative dose of 131I-30F11 or 177Lu-30F11 antibody prior to adoptive cell therapy.

3.
Nucl Med Biol ; 82-83: 80-88, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32113033

RESUMO

BACKGROUND: cART has significantly improved the life expectancy of people living with HIV (PLWH). However, it fails to eliminate the long-lived reservoir of latent HIV-infected cells. Radioimmunotherapy (RIT) relies on antigen-specific monoclonal antibodies (mAbs) for targeted delivery of lethal doses of ionizing radiation to cells. Previously, we have demonstrated that human mAb 2556 against HIV gp41 conjugated with 213Bismuth radioisotope (t1/2 = 46 min, alpha-emitter) selectively killed HIV-infected cells. 225Actinium (t1/2 = 9.92 d, alpha-emitter) and 177Lutetium (t1/2 = 6.7 d, beta-emitter) are two long-lived clinically proven radioisotopes for cancer treatment which might be more effective in killing infected cells systemically and in CNS. METHODS: In this study we have conjugated 2556 mAb with 213Bi, 225Ac and 177Lu, and compared their ability to kill HIV-infected human peripheral blood mononuclear cells (PBMCs) and monocytes. PBMCs and monocytes from healthy donors were infected with HIVp49.5 and treated in vitro with increasing concentrations of 213Bi (4-20 µCi)-, 225Ac (20-100 nCi)- and 177Lu (4-50 µCi)-2556 mAb. RESULTS: After three days post-treatment of infected PBMCs and monocytes, 213Bi- and 177Lu-conjugated 2556 mAb reduced virus production measured by p24 level in a dose-dependent manner, whereas, 225Ac-2556 showed minimal effect. However, seven days post-treatment all three radioisotopes showed significantly more pronounced reduction of virus replication as compared to control labeled mAb with 225Ac-2556 showing the least non-specific killing. CONCLUSION: These results indicate that RIT holds promise as a novel treatment option for the eradication of HIV-infected cells that merits further study in combination with cART and reactivation drugs.


Assuntos
Anticorpos Monoclonais/imunologia , HIV-1/fisiologia , Glicoproteínas de Membrana/imunologia , Anticorpos Monoclonais/química , Linhagem Celular , Quebras de DNA de Cadeia Dupla/efeitos da radiação , HIV-1/efeitos da radiação , Humanos , Marcação por Isótopo , Leucócitos Mononucleares/virologia , Monócitos/virologia
4.
Antiviral Res ; 171: 104594, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31470041

RESUMO

Respiratory syncytial virus (RSV) is responsible for a large proportion of acute lower respiratory tract infections, specifically in children. Pneumonia virus of mice (PVM) causes similar lung pathology and clinical disease in rodents, and is therefore an appropriate model of RSV infection. Previously, we demonstrated that a single intranasal dose of P-I-P, a novel immunomodulator composed of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene, confers protection in Balb/c mice for up to 3 days from lethal PVM-15 infection. In the present study a dual intranasal treatment with P-I-P was shown to extend the duration of the protection conferred by P-I-P from PVM-15 challenge. Balb/c mice treated twice with P-I-P showed higher survival rates and milder clinical signs when compared to animals that received a single P-I-P dose. While the mice treated with two consecutive doses of P-I-P experienced some weight loss, they all recovered. The dual P-I-P treatment mediated infiltration of several innate immune cells into the BALF and lung, including alveolar macrophages, neutrophils, and γδ T cells. Partial depletion of alveolar macrophages decreased survival rates and exacerbated clinical signs of mice subjected to the P-I-P dual treatment regime followed by PVM-15 challenge. This suggests that the alveolar macrophage is at least partially responsible for the protection elicited by this novel prophylactic treatment strategy.


Assuntos
Imunidade Inata , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Vírus da Pneumonia Murina/efeitos dos fármacos , Vírus da Pneumonia Murina/imunologia , Infecções por Pneumovirus/imunologia , Infecções por Pneumovirus/virologia , Animais , Linhagem Celular , Citocinas/biossíntese , Citocinas/sangue , Feminino , Interações Hospedeiro-Patógeno , Fatores Imunológicos/administração & dosagem , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Infecções por Pneumovirus/tratamento farmacológico , Infecções por Pneumovirus/mortalidade
5.
Vaccine ; 36(17): 2326-2336, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29559168

RESUMO

Respiratory syncytial virus (RSV) causes acute respiratory tract infections in infants, the elderly and immunocompromised individuals. No licensed vaccine is available against RSV. We previously reported that intranasal immunization of rodents and lambs with a RSV vaccine candidate (ΔF/TriAdj) induces protective immunity with a good safety profile. ΔF/TriAdj promoted innate immune responses in respiratory mucosal tissues in vivo, by local chemokine and cytokine production, as well as infiltration and activation of immune cells including macrophages. The macrophage is an important cell type in context of both innate and adaptive immune responses against RSV. Therefore, we characterized the effects of ΔF/TriAdj on a murine macrophage cell line, RAW264.7, and bone marrow-derived macrophages (BMMs). A gene expression study of pattern recognition receptors (PRRs) revealed induction of endosomal and cytosolic receptors in RAW264.7 cells and BMMs by ΔF/TriAdj, but no up-regulation by ΔF in PBS. As a secondary response to the PRR gene expression, induction of several chemokines and pro-inflammatory cytokines, as well as up-regulation of MHC-II and co-stimulatory immune markers, was observed. To further investigate the mechanisms involved in ΔF/TriAdj-mediated secondary responses, we used relevant signal transduction pathway inhibitors. Based on inhibition studies at both transcript and protein levels, JNK, ERK1/2, CaMKII, PI3K and JAK pathways were clearly responsible for ΔF/TriAdj-mediated chemokine and pro-inflammatory cytokine responses, while the p38 and NF-κB pathways appeared to be not or minimally involved. ΔF/TriAdj induced IFN-ß, which may participate in the JAK-STAT pathway to further amplify CXCL-10 production, which was strongly up-regulated. Blocking this pathway by a JAK inhibitor almost completely abrogated CXCL-10 production and caused a significant reduction in the cell surface expression of MHC-II and co-stimulatory immune markers. These data demonstrate that ΔF/TriAdj induces multiple signaling pathways in macrophages.


Assuntos
Macrófagos/imunologia , Polímeros/química , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Transdução de Sinais/imunologia , Proteínas Virais de Fusão/imunologia , Adjuvantes Imunológicos/química , Animais , Biomarcadores/metabolismo , Linhagem Celular , Quimiocinas/imunologia , Imunidade Inata/imunologia , Imunização/métodos , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/virologia , Camundongos , Células RAW 264.7 , Infecções por Vírus Respiratório Sincicial/metabolismo , Vacinas contra Vírus Sincicial Respiratório/química , Vacinação/métodos , Proteínas Virais de Fusão/química
6.
Vaccine ; 35(35 Pt A): 4486-4489, 2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28599794

RESUMO

Adjuvants are crucial components of many vaccines. They are used to improve the immunogenicity of vaccines with the aim of conferring long-term protection, to enhance the efficacy of vaccines in newborns, elderly or immunocompromised persons, and to reduce the amount of antigen or the number of doses required to elicit effective immunity. Novel combination adjuvants have been tested in both candidate animals and humans vaccines and have generated encouraging results. Recently, we developed a combination adjuvant platform (TriAdj) comprising of three components, namely a TLR agonist, either polyI:C or CpG oligodeoxynucleotides (ODN), host defense peptide and polyphosphazene. This adjuvant platform is stable and highly effective in a wide range of animal and human vaccines tested in mice, cotton rats, pigs, sheep, and koalas. TriAdj with various vaccines antigens induced effective long-term humoral and cellular immunity. Moreover, the adjuvant platform is suitable for maternal immunization and highly effective in neonates even in the presence of maternal antibodies. This novel vaccine platform, offers excellent opportunity for use in present and future generations of vaccines against multiple infectious agents and targets challenging populations.


Assuntos
Adjuvantes Imunológicos/química , Oligodesoxirribonucleotídeos/química , Compostos Organofosforados/química , Polímeros/química , Vacinas Combinadas/química , Vacinas Combinadas/imunologia , Animais , Desenho de Fármacos , Feminino , Humanos , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Imunidade Materno-Adquirida/imunologia , Lactente , Gravidez , Vacinas Combinadas/síntese química
7.
Vaccine ; 35(27): 3466-3473, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28527686

RESUMO

Bovine viral diarrhea virus (BVDV) is one of the most serious pathogens in cattle. Recently, we developed a novel adjuvant platform (TriAdj) that includes a toll-like receptor 3 agonist, poly (I:C); an innate defense regulatory peptide; and water-soluble polymer, poly[di(sodiumcarboxylatoethylphenoxy)]-phosphazene (PCEP). To develop a needle-free intradermal (ID) subunit vaccine, the BVDV type-2 E2 protein was formulated with TriAdj, and immune protection was evaluated in calves against a BVDV-2 strain. Intradermal delivery of E2/TriAdj elicited robust virus neutralizing antibodies and cell-mediated immune responses including CD4+ and CD8+ T-cell responses. The development of CD8+ T-cell responses in vaccinated calves indicates that TriAdj promotes cross-presentation. Upon challenge with virulent BVDV-2, the vaccinated calves showed no weight loss, leukopenia or virus shedding, and almost no temperature increase, in contrast to the control animals, which had severe clinical disease and shed virus for three to six days in nasal fluids and white blood cells. Intradermal vaccination was shown to attract various immune cell populations including dendritic cells, the most important antigen presenting cells. These data demonstrate that ID delivery is suitable as an administration route in cattle and that ID delivered, TriAdj-formulated E2 can protect cattle from BVDV-2.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Vírus da Diarreia Viral Bovina Tipo 1/imunologia , Imunidade Inata , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Peso Corporal , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Bovinos , Febre/prevenção & controle , Injeções Intradérmicas , Leucopenia/prevenção & controle , Vacinas Virais/administração & dosagem , Eliminação de Partículas Virais
8.
Antiviral Res ; 135: 108-119, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27771388

RESUMO

Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in infants and young children. There are no licensed RSV vaccines available, and the few treatment options for high-risk individuals are either extremely costly or cause severe side effects and toxicity. Immunomodulation mediated by a novel formulation consisting of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene (P-I-P) was evaluated in the context of lethal infection with pneumonia virus of mice (PVM). Intranasal delivery of a single dose of P-I-P protected adult mice against PVM when given 24 h prior to challenge. These animals experienced minimal weight loss, no clinical disease, 100% survival, and reduced lung pathology. Similar clinical outcomes were observed in mice treated up to 3 days prior to infection. P-I-P pre-treatment induced early mRNA and protein expression of key chemokine and cytokine genes, reduced the recruitment of neutrophils and eosinophils, decreased virus titers in the lungs, and modulated the delayed exacerbated nature of PVM disease without any short-term side effects. On day 14 post-infection, P-I-P-treated mice were confirmed to be PVM-free. These results demonstrate the capacity of this formulation to prevent PVM and possibly other viral respiratory infections.


Assuntos
Imunidade Inata , Fatores Imunológicos/administração & dosagem , Vírus da Pneumonia Murina/imunologia , Compostos Organofosforados/administração & dosagem , Infecções por Pneumovirus/prevenção & controle , Poli I-C/administração & dosagem , Polímeros/administração & dosagem , Adjuvantes Imunológicos , Administração Intranasal , Animais , Citocinas/imunologia , Fatores Imunológicos/química , Fatores Imunológicos/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Compostos Organofosforados/imunologia , Infecções por Pneumovirus/imunologia , Poli I-C/imunologia , Receptor 3 Toll-Like/agonistas
9.
Vaccine ; 34(42): 5114-5124, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27591951

RESUMO

Respiratory syncytial virus (RSV) causes serious upper and lower respiratory tract infections in newborns and infants. Presently, there is no licensed vaccine against RSV. We previously reported the safety and efficacy of a novel vaccine candidate (ΔF/TriAdj) in rodent and lamb models following intranasal immunization. However, the effects of the vaccine on the innate immune system in the upper and lower respiratory tracts, when delivered intranasally, have not been characterized. In the present study, we found that ΔF/TriAdj triggered transient production of chemokines, cytokines and interferons in the nasal tissues and lungs of BALB/c mice. The types of chemokines produced were consistent with the populations of immune cells recruited, i.e. dendritic cells, macrophages and neutrophils, in the nose-associated lymphoid tissue (NALT), lung and their draining lymph nodes of the ΔF/TriAdj-immunized group. In addition, ΔF/TriAdj stimulated cellular activation with generation of mucosal and systemic antibody responses, and conferred complete protection from viral infection in the lungs upon RSV challenge. The effect of ΔF/TriAdj was short-lived in the nasal tissues and more prolonged in the lungs. In addition, both innate and adaptive immune responses were lower when mice were immunized with ΔF alone. These results suggest that ΔF/TriAdj modulates the innate mucosal environment in both upper and lower respiratory tracts, which contributes to robust adaptive immune responses and long-term protective efficacy of this novel vaccine formulation.


Assuntos
Imunidade Adaptativa , Adjuvantes Imunológicos , Imunidade Inata , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/química , Proteínas Virais de Fusão/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Quimiocinas/biossíntese , Citocinas/biossíntese , Interferons/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Polímeros/química , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sinciciais Respiratórios/química , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/química
10.
Parasite Immunol ; 37(7): 368-75, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25824598

RESUMO

Isolation of the soluble exogenous antigens (SEAgs), its immune response study and proteome profiling is an essential prerequisite for understanding the molecular pathogenesis of Leishmania donovani. The immunostimulatory potential of L. donovani SEAgs, purified from culture of L. donovani clinical isolate, was evaluated for their ability to induce cellular responses in treated/cured hamsters. SEAgs induced significant proliferative responses in lymphocytes (SI 5.6 ± 2.3; P < 0.01) isolated from cured hamster. In addition, significant NO production in response to SEAgs was also noticed in macrophages of hamsters, mouse and human cell lines (J774A-1 and THP1). Western blot analyses with antibodies against proteophosphoglycan (PPG; surface-expressed and secreted molecule) of L. donovani revealed that PPG molecules are also present in L. donovani SEAgs. Mass spectrometry (MS)-based proteome analysis of 12 protein bands of SEAgs through MALDI-TOF/TOF endorsed the identification of some Th1-stimulatory immunogenic proteins. These immunogenic proteins may offer increased hope for the discovery of new promising vaccine candidates against visceral leishmaniasis (VL). The overall results suggest that immunostimulatory molecules are present in the SEAgs, which may be further exploited, for developing a subunit vaccine against VL a fatal human disease.


Assuntos
Antígenos de Protozoários/imunologia , Leishmania donovani/imunologia , Leishmaniose Visceral/imunologia , Macrófagos/imunologia , Animais , Anticorpos/imunologia , Antígenos de Protozoários/isolamento & purificação , Linhagem Celular , Proliferação de Células , Cricetinae , Humanos , Imunização , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Ativação de Macrófagos/imunologia , Masculino , Camundongos , Proteoma/imunologia
11.
Vaccine ; 32(50): 6758-64, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25454860

RESUMO

Bovine viral diarrhea virus (BVDV) is still one of the most serious pathogens in cattle, meriting the development of improved vaccines. Recently, we developed a new adjuvant consisting of poly[di(sodium carboxylatoethylphenoxy)]-phosphazene (PCEP), either CpG ODN or poly(I:C), and an immune defense regulator (IDR) peptide. As this adjuvant has been shown to mediate the induction of robust, balanced immune responses, it was evaluated in an E2 subunit vaccine against BVDV in lambs and calves. The BVDV type 2 E2 protein was produced at high levels in a mammalian expression system and purified. When formulated with either CpG ODN or poly(I:C), together with IDR and PCEP, the E2 protein elicited high antibody titers and production of IFN-γ secreting cells in lambs. As the immune responses were stronger when poly(I:C) was used, the E2 protein with poly(I:C), IDR and PCEP was subsequently tested in cattle. Robust virus neutralizing antibodies as well as cell-mediated immune responses, including CD8(+) cytotoxic T cell (CTL) responses, were induced. The fact that CTL responses were demonstrated in calves vaccinated with an E2 protein subunit vaccine indicates that this adjuvant formulation promotes cross-presentation. Furthermore, upon challenge with a high dose of virulent BVDV-2, the vaccinated calves showed almost no temperature response, weight loss, leukopenia or virus replication, in contrast to the control animals, which had severe clinical disease. These data suggest that this E2 subunit formulation induces significant protection from BVDV-2 challenge, and thus is a promising BVDV vaccine candidate; in addition, the adjuvant platform has applications in bovine vaccines in general.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Doenças dos Bovinos/prevenção & controle , Vírus da Diarreia Viral Bovina Tipo 2/imunologia , Infecções por Pestivirus/veterinária , Doenças dos Ovinos/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Bovinos , Doenças dos Bovinos/patologia , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Infecções por Pestivirus/patologia , Infecções por Pestivirus/prevenção & controle , Ovinos , Doenças dos Ovinos/patologia , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Virais/administração & dosagem
12.
Virology ; 432(2): 271-6, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22704066

RESUMO

Miltefosine (Milt) was originally synthesized as an antineoplastic agent but this phospholipid drug is now clinically used as an antiprotozoal compound. We demonstrate here that Milt reduces replication of HIV-1 in cocultures of human dendritic cells (DCs) and CD4(+) T cells. This phenomenon is due to a rapid secretion of soluble factors by DCs. We present evidence that the Milt-mediated repression in virus production is associated with induction of type-I interferon (IFN) in DCs. The Milt-dependent diminution in HIV-1 production was not totally abrogated by B18R, a vaccinia virus-encoded neutralizing type-I IFN receptor, which suggests the involvement of another yet to be identified soluble factor. Altogether, these results suggest that a therapy with Milt when used to control protozoan infections in individuals also carrying HIV-1 might also help to limit viral load. Additional studies are warranted to estimate the exact therapeutic potential of Milt as an anti-HIV-1 agent.


Assuntos
Linfócitos T CD4-Positivos/virologia , Células Dendríticas/imunologia , HIV-1/efeitos dos fármacos , Interferon Tipo I/metabolismo , Fosforilcolina/análogos & derivados , Replicação Viral/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Técnicas de Cocultura , HIV-1/fisiologia , Humanos , Fosforilcolina/farmacologia
13.
PLoS Negl Trop Dis ; 3(5): e441, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19468304

RESUMO

BACKGROUND: Visceral leishmaniasis has emerged as an important opportunistic disease among patients infected with HIV-1. Both HIV-1 and the protozoan parasite Leishmania can productively infect cells of the macrophage-dendritic cell lineage. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that Leishmania infantum amastigotes increase HIV-1 production when human primary dendritic cells (DCs) are cocultured together with autologous CD4(+) T cells. Interestingly, the promastigote form of the parasite does not modulate virus replication. Moreover, we report that amastigotes promote virus replication in both cell types. Our results indicate that this process is due to secretion of parasite-induced soluble factors by DCs. Luminex micro-beads array system analyses indicate that Leishmania infantum amastigotes induce a higher secretion of several cytokines (i.e. IL-1alpha, IL-2, IL-6, IL-10 and TNF-alpha) and chemokines (i.e. MIP-1alpha, MIP-1beta and RANTES) in these cells. Studies conducted with pentoxifylline and neutralizing antibodies revealed that the Leishmania-dependent augmentation in HIV-1 replication is due to a higher secretion of IL-6 and TNF-alpha. CONCLUSIONS/SIGNIFICANCE: Altogether these findings suggest that the presence of Leishmania within DC/T-cell conjugates leads to an enhancement of virus production and demonstrate that HIV-1 and Leishmania can establish complex interactions in such a cellular microenvironment.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/parasitologia , HIV-1/crescimento & desenvolvimento , Interleucina-6/metabolismo , Leishmania infantum/crescimento & desenvolvimento , Leishmania infantum/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas , Células Dendríticas/virologia , Humanos
14.
J Infect Dis ; 198(9): 1292-9, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18816190

RESUMO

Visceral leishmaniasis is now recognized as an opportunistic disease in individuals infected with human immunodeficiency virus type 1 (HIV-1). Although the usefulness of HIV-1 protease inhibitors (PIs) in antiretroviral regimens is well documented, little is known about their potential impact in the setting of Leishmania/HIV-1 coinfections. We now report that, although selected PIs do not inhibit the growth of Leishmania infantum promastigotes alone in culture, these drugs significantly inhibit the intracellular survival of parasites in phorbol myristate acetate-differentiated THP-1 macrophages and human primary monocyte-derived macrophages (MDMs). Furthermore, a field isolate of Leishmania donovani resistant to sodium stibogluconate (SbV), one of the drugs most commonly used to treat leishmaniasis, is equally susceptible to the tested PIs compared with a sensitive strain, thus suggesting that resistance to SbV does not result in cross-resistance to PIs. Importantly, the efficacy of PIs to reduce the intracellular growth of Leishmania parasites is also observed in MDMs coinfected with HIV-1.


Assuntos
Inibidores da Protease de HIV/farmacologia , Leishmania/efeitos dos fármacos , Leishmania/fisiologia , Leishmaniose Visceral/parasitologia , Animais , Linhagem Celular Tumoral , Humanos , Dose Letal Mediana , Macrófagos/parasitologia , Nelfinavir/farmacologia , Ritonavir/farmacologia , Saquinavir/farmacologia
15.
J Infect Dis ; 197(12): 1701-8, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18422456

RESUMO

Visceral leishmaniasis is now recognized as an opportunistic disease in patients infected with human immunodeficiency virus type 1 (HIV-1). We report here that Leishmania infantum promastigotes enhance HIV-1 replication in monocyte-derived macrophages (MDMs) at late time points in the virus growth curve but also that, surprisingly, a reduction in HIV-1 production is seen during the initial days after infection. This early effect is caused by a Leishmania-mediated inhibition of virus entry into MDMs through the action of lipophosphoglycan (LPG), the major promastigote surface glycolipid. The impact of LPG in the observed phenomenon was confirmed using LPG-defective lpg1-/- knockout mutant promastigotes. Our results suggest that the LPG-mediated effect results from the disruption of lipid rafts. Altogether, these findings suggest that the presence of Leishmania within the same cellular microenvironment leads to 2 opposite, time-dependent effects on HIV-1 replication. Leishmania and HIV-1 can thus establish complex interactions in their common natural host cells.


Assuntos
Glicoesfingolipídeos/metabolismo , HIV-1/fisiologia , Leishmania infantum/fisiologia , Macrófagos/parasitologia , Macrófagos/virologia , Microdomínios da Membrana/fisiologia , Animais , Células Cultivadas , Humanos , Fatores de Tempo , Replicação Viral
16.
Vaccine ; 24(15): 2900-9, 2006 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-16448729

RESUMO

Most of the studies for the identification of prophylactic antigens that elicit T cell responses were concentrated on membrane proteins of Leishmania donovani. This study was taken up to assess L. donovani soluble promastigote antigens for their ability to stimulate proliferation of peripheral blood mononuclear cells (PBMCs) from cured visceral leishmaniasis (VL) patients, endemic and non-endemic controls and lymphocytes/peritoneal macrophages of cured hamsters. The soluble protein was subjected to sequential precipitation with saturated ammonium sulphate (20%, 40%, 60% and 80%), of which largely 80% fractioned protein showed significant cellular responses in cured patients and hamsters. This fraction was further fractionated into five sub fractions by preparative SDS-PAGE and subjected to re-evaluation for their ability to induce cellular responses. Out of these, only F2 sub fraction belonging to the MW of 97.4-68 kDa stimulated remarkable lymphoproliferative and IFN-gamma responses in cured VL patients and in endemic controls. Similarly, significant lymphoproliferative responses and nitric oxide production were also noticed in cured Leishmania infected animals indicating an element of uniformity in responses between hamster and human. F2 sub fraction, when evaluated for its prophylactic efficacy with BCG against L. donovani challenge in hamster exhibited significant parasite inhibition in spleen (71.1%; p<0.001) and liver (68.2%; p<0.001) as compared to their unvaccinated counterpart. The vaccinated animals showed significant lymphoproliferative response and nitric oxide production but leishmania specific IgG level were suppressed. The results indicate the presence of immunostimulatory and protective molecules in F2 sub fraction which may further be exploited for the development of a vaccine against VL, hitherto an unrealized goal.


Assuntos
Antígenos de Protozoários/imunologia , Leishmania donovani/imunologia , Leishmaniose Visceral/prevenção & controle , Linfócitos/imunologia , Adolescente , Adulto , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/isolamento & purificação , Células Cultivadas , Fracionamento Químico , Criança , Pré-Escolar , Cricetinae , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Interferon gama/biossíntese , Macrófagos Peritoneais/imunologia , Masculino , Óxido Nítrico/biossíntese
17.
Vaccine ; 23(9): 1189-96, 2005 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-15629362

RESUMO

The integral membrane proteins (IMP's) of promastigotes of two virulent strain of Leishmania (L.) donovani Dd8 and Leishmania (L.) infantum LV9 and one avirulent viscerotropic strain Leishmania tropica UR6 were extracted by phase separation technique using a non-ionic detergent "Triton X-114". This detergent is homogeneous at 0 degrees C but divides in an aqueous phase and a detergent phase at above 20 degrees C. The phase partition resulted in solubilisation of hydrophilic proteins in aqueous phase, and IMP's with an amphiphilic nature were recovered in the detergent phase. The strain wise quantitative recovery rates of IMP's were estimated. These proteins were purified by chill methanol centrifugation and used as vaccinogen, alone or in combination with adjuvant against L. donovani challenge in hamster model. Among all the combinations, hamsters immunised with IMP of L. donovani (Dd8) in combination with CFA resulted in 75% parasite inhibition in spleen (P <0.001), however, 61.14% (P <0.001) and 77.60% (P <0.001) parasitic inhibition was achieved in liver and bone marrow respectively as compared to their unvaccinated counter part. Similar combinations with UR6 and LV9 strain inhibited the parasite establishment up to 65.12% (P <0.001) and 66.87% (P <0.001), respectively on splenic site. The specific IgG level against (Dd8 strain) soluble leishmania promastigote antigen was monitored at different stages by enzyme linked immunosorbent assay (ELISA) corresponds to the level of parasitic establishment. Similar observations were made in cases of LV9 and UR6 strains. However, significant lymphoproliferative response to IMPs of Dd8 strain (SI 3.5-4.9, P <0.001) was noticed in all IMP + CFA vaccinated animals. Thus, this study will provide a lead for more manipulative trials to develop a subunit vaccine against the fatal disease.


Assuntos
Leishmania/imunologia , Leishmaniose/prevenção & controle , Proteínas de Membrana/imunologia , Proteínas de Membrana/isolamento & purificação , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/uso terapêutico , Vacinas Protozoárias/uso terapêutico , Animais , Cricetinae , Leishmaniose/imunologia , Masculino , Proteínas de Membrana/uso terapêutico , Proteínas de Protozoários/isolamento & purificação , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA