Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Vaccines (Basel) ; 12(5)2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38793801

RESUMO

This work evaluated in vivo an experimental-multivalent-vaccine (EMV) based on three Porcine Respiratory Complex (PRC)-associated antigens: Porcine Circovirus Type 2 (PCV2), M. hyopneumoniae (Mhyop) and M. hyorhinis (Mhyor), microencapsulated with sulfated chitosan (M- ChS + PRC-antigens), postulating chitosan sulphate (ChS) as a mimetic of the heparan sulfate receptor used by these pathogens for cell invasion. The EMV was evaluated physicochemically by SEM (Scanning-Electron-Microscopy), EDS (Energy-Dispersive-Spectroscopy), Pdi (Polydispersity-Index) and zeta potential. Twenty weaned pigs, distributed in four groups, were evaluated for 12 weeks. The groups 1 through 4 were as follows: 1-EMV intramuscular-route (IM), 2-EMV oral-nasal-route (O/N), 3-Placebo O/N (M-ChS without antigens), 4-Commercial-vaccine PCV2-Mhyop. qPCR was used to evaluate viral/bacterial load from serum, nasal and bronchial swab and from inguinal lymphoid samples. Specific humoral immunity was evaluated by ELISA. M-ChS + PRC-antigens measured between 1.3-10 µm and presented low Pdi and negative zeta potential, probably due to S (4.26%). Importantly, the 1-EMV protected 90% of challenged animals against PCV2 and Mhyop and 100% against Mhyor. A significant increase in antibody was observed for Mhyor (1-EMV and 2-EMV) and Mhyop (2-EMV), compared with 4-Commercial-vaccine. No difference in antibody levels between 1-EMV and 4-Commercial-vaccine for PCV2-Mhyop was observed. Conclusion: The results demonstrated the effectiveness of the first EMV with M-ChS + PRC-antigens in pigs, which were challenged with Mhyor, PCV2 and Mhyop, evidencing high protection for Mhyor, which has no commercial vaccine available.

2.
Am J Reprod Immunol ; 79(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29048721

RESUMO

PROBLEM: Immunocastration or vaccination against the GnRH-I hormone is a promising alternative to reproductive control in different animal species. Given the low immunogenicity of this hormone, the use of adjuvants becomes necessary. METHOD OF STUDY: This study evaluated the effects of three adjuvants that induce different immune response profiles over gonadal function, fertility, and expression of GnRH-I. Female mice (n = 6) were vaccinated at days 1 and 30 with a recombinant antigen for immunocastration and different adjuvants that induced preferentially Th1/Th2, Th2, and Th1 immune profiles. RESULTS: Th1/Th2 response is the most efficient to block reproductive activity in vaccinated animals, reducing the number of luteal bodies and pre-ovulatory follicles. Th2 and Th1/Th2 responses induced an increase in GnRH-I at the hypothalamus. CONCLUSION: The immune profile induced by different adjuvants is essential on the effects over fertility, gonadal function, and hypothalamic GnRH-I expression in immunocastrated animals.


Assuntos
Hormônio Liberador de Gonadotropina/imunologia , Gônadas/fisiologia , Hipotálamo/metabolismo , Precursores de Proteínas/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Fertilidade , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/patologia , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Precursores de Proteínas/genética , Receptores LHRH/metabolismo , Equilíbrio Th1-Th2 , Vacinação
3.
BMC Cancer ; 16(1): 731, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27619675

RESUMO

BACKGROUND: For several decades now an antagonism between Trypanosoma cruzi infection and tumor development has been detected. The molecular basis of this phenomenon remained basically unknown until our proposal that T. cruzi Calreticulin (TcCRT), an endoplasmic reticulum-resident chaperone, translocated-externalized by the parasite, may mediate at least an important part of this effect. Thus, recombinant TcCRT (rTcCRT) has important in vivo antiangiogenic and antitumor activities. However, the relevant question whether the in vivo antitumor effect of T. cruzi infection is indeed mediated by the native chaperone (nTcCRT), remains open. Herein, by using specific modified anti-rTcCRT antibodies (Abs), we have neutralized the antitumor activity of T. cruzi infection and extracts thereof, thus identifying nTcCRT as a valid mediator of this effect. METHODS: Polyclonal anti-rTcCRT F(ab')2 Ab fragments were used to reverse the capacity of rTcCRT to inhibit EAhy926 endothelial cell (EC) proliferation, as detected by BrdU uptake. Using these F(ab')2 fragments, we also challenged the capacity of nTcCRT, during T. cruzi infection, to inhibit the growth of an aggressive mammary adenocarcinoma cell line (TA3-MTXR) in mice. Moreover, we determined the capacity of anti-rTcCRT Abs to reverse the antitumor effect of an epimastigote extract (EE). Finally, the effects of these treatments on tumor histology were evaluated. RESULTS: The rTcCRT capacity to inhibit ECs proliferation was reversed by anti-rTcCRT F(ab')2 Ab fragments, thus defining them as valid probes to interfere in vivo with this important TcCRT function. Consequently, during infection, these Ab fragments also reversed the in vivo experimental mammary tumor growth. Moreover, anti-rTcCRT Abs also neutralized the antitumor effect of an EE, again identifying the chaperone protein as an important mediator of this anti mammary tumor effect. Finally, as determined by conventional histological parameters, in infected animals and in those treated with EE, less invasive tumors were observed while, as expected, treatment with F(ab')2 Ab fragments increased malignancy. CONCLUSION: We have identified translocated/externalized nTcCRT as responsible for at least an important part of the anti mammary tumor effect of the chaperone observed during experimental infections with T. cruzi.


Assuntos
Calreticulina/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Trypanosoma cruzi/metabolismo , Tripanossomíase/parasitologia , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/farmacologia , Calreticulina/antagonistas & inibidores , Calreticulina/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Feminino , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/farmacologia
4.
Am J Trop Med Hyg ; 92(5): 887-97, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25758653

RESUMO

Trypanosoma cruzi calreticulin (TcCRT), a 47-kDa chaperone, translocates from the endoplasmic reticulum to the area of flagellum emergence. There, it binds to complement components C1 and mannan-binding lectin (MBL), thus acting as a main virulence factor, and inhibits the classical and lectin pathways. The localization and functions of TcCRT, once the parasite is inside the host cell, are unknown. In parasites infecting murine macrophages, polyclonal anti-TcCRT antibodies detected TcCRT mainly in the parasite nucleus and kinetoplast. However, with a monoclonal antibody (E2G7), the resolution and specificity of the label markedly improved, and TcCRT was detected mainly in the parasite kinetoplast. Gold particles, bound to the respective antibodies, were used as probes in electron microscopy. This organelle may represent a stopover and accumulation site for TcCRT, previous its translocation to the area of flagellum emergence. Finally, early during T. cruzi infection and by unknown mechanisms, an important decrease in the number of MHC-I positive host cells was observed.


Assuntos
Calreticulina/metabolismo , Doença de Chagas/parasitologia , Macrófagos/parasitologia , Trypanosoma cruzi/metabolismo , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Calreticulina/imunologia , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Complemento C1/metabolismo , Interações Hospedeiro-Parasita , Humanos , Macrófagos/metabolismo , Lectina de Ligação a Manose/metabolismo , Camundongos , Modelos Biológicos , Chaperonas Moleculares/imunologia , Chaperonas Moleculares/metabolismo , Organelas/metabolismo , Organelas/ultraestrutura , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Coelhos , Proteínas Recombinantes , Trypanosoma cruzi/imunologia , Trypanosoma cruzi/ultraestrutura , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo
5.
PLoS Negl Trop Dis ; 8(2): e2696, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24551259

RESUMO

Trypanosoma cruzi calreticulin (TcCRT) is a virulence factor that binds complement C1, thus inhibiting the activation of the classical complement pathway and generating pro-phagocytic signals that increase parasite infectivity. In a previous work, we characterized a clonal cell line lacking one TcCRT allele (TcCRT+/-) and another overexpressing it (TcCRT+), both derived from the attenuated TCC T. cruzi strain. The TcCRT+/- mutant was highly susceptible to killing by the complement machinery and presented a remarkable reduced propagation and differentiation rate both in vitro and in vivo. In this report, we have extended these studies to assess, in a mouse model of disease, the virulence, immunogenicity and safety of the mutant as an experimental vaccine. Balb/c mice were inoculated with TcCRT+/- parasites and followed-up during a 6-month period. Mutant parasites were not detected by sensitive techniques, even after mice immune suppression. Total anti-T. cruzi IgG levels were undetectable in TcCRT+/- inoculated mice and the genetic alteration was stable after long-term infection and it did not revert back to wild type form. Most importantly, immunization with TcCRT+/- parasites induces a highly protective response after challenge with a virulent T. cruzi strain, as evidenced by lower parasite density, mortality, spleen index and tissue inflammatory response. TcCRT+/- clones are restricted in two important properties conferred by TcCRT and indirectly by C1q: their ability to evade the host immune response and their virulence. Therefore, deletion of one copy of the TcCRT gene in the attenuated TCC strain generated a safe and irreversibly gene-deleted live attenuated parasite with high immunoprotective properties. Our results also contribute to endorse the important role of TcCRT as a T. cruzi virulence factor.


Assuntos
Calreticulina/genética , Proteínas de Protozoários/genética , Trypanosoma cruzi/genética , Trypanosoma cruzi/patogenicidade , Animais , Calreticulina/metabolismo , Deleção de Genes , Interações Hospedeiro-Parasita/genética , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Protozoários/metabolismo , Trypanosoma cruzi/fisiologia , Virulência/genética
6.
PLoS Negl Trop Dis ; 7(8): e2376, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991234

RESUMO

BACKGROUND: 9 million people are infected with Trypanosoma cruzi in Latin America, plus more than 300,000 in the United States, Canada, Europe, Australia, and Japan. Approximately 30% of infected individuals develop circulatory or digestive pathology. While in underdeveloped countries transmission is mainly through hematophagous arthropods, transplacental infection prevails in developed ones. METHODOLOGY/PRINCIPAL FINDINGS: During infection, T. cruzi calreticulin (TcCRT) translocates from the endoplasmic reticulum to the area of flagellum emergence. There, TcCRT acts as virulence factor since it binds maternal classical complement component C1q that recognizes human calreticulin (HuCRT) in placenta, with increased parasite infectivity. As measured ex vivo by quantitative PCR in human placenta chorionic villi explants (HPCVE) (the closest available correlate of human congenital T. cruzi infection), C1q mediated up to a 3-5-fold increase in parasite load. Because anti-TcCRT and anti-HuCRT F(ab')2 antibody fragments are devoid of their Fc-dependent capacity to recruit C1q, they reverted the C1q-mediated increase in parasite load by respectively preventing its interaction with cell-bound CRTs from both parasite and HPCVE origins. The use of competing fluid-phase recombinant HuCRT and F(ab')2 antibody fragments anti-TcCRT corroborated this. These results are consistent with a high expression of fetal CRT on placental free chorionic villi. Increased C1q-mediated infection is paralleled by placental tissue damage, as evidenced by histopathology, a damage that is ameliorated by anti-TcCRT F(ab')2 antibody fragments or fluid-phase HuCRT. CONCLUSIONS/SIGNIFICANCE: T. cruzi infection of HPCVE is importantly mediated by human and parasite CRTs and C1q. Most likely, C1q bridges CRT on the parasite surface with its receptor orthologue on human placental cells, thus facilitating the first encounter between the parasite and the fetal derived placental tissue. The results presented here have several potential translational medicine aspects, specifically related with the capacity of antibody fragments to inhibit the C1q/CRT interactions and thus T. cruzi infectivity.


Assuntos
Calreticulina/imunologia , Calreticulina/metabolismo , Doença de Chagas/imunologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Placenta/imunologia , Trypanosoma cruzi/imunologia , Doença de Chagas/parasitologia , Feminino , Humanos , Placenta/parasitologia , Gravidez , Ligação Proteica
7.
J Immunol ; 190(12): 6457-67, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23677468

RESUMO

Elevated numbers of activated platelets circulate in patients with chronic inflammatory diseases, including atherosclerosis and coronary disease. Activated platelets can activate the complement system. Although complement activation is essential for immune responses and removal of spent cells from circulation, it also contributes to inflammation and thrombosis, especially in patients with defective complement regulation. Proinflammatory activated leukocytes, which interact directly with platelets in response to vascular injury, are among the main sources of properdin, a positive regulator of the alternative pathway. The role of properdin in complement activation on stimulated platelets is unknown. Our data show that physiological forms of human properdin bind directly to human platelets after activation by strong agonists in the absence of C3, and bind nonproportionally to surface CD62P expression. Activation of the alternative pathway on activated platelets occurs when properdin is on the surface and recruits C3b or C3(H2O) to form C3b,Bb or a novel cell-bound C3 convertase [C3(H2O),Bb], which normally is present only in the fluid phase. Alternatively, properdin can be recruited by C3(H2O) on the platelet surface, promoting complement activation. Inhibition of factor H-mediated cell surface complement regulation significantly increases complement deposition on activated platelets with surface properdin. Finally, properdin released by activated neutrophils binds to activated platelets. Altogether, these data suggest novel molecular mechanisms for alternative pathway activation on stimulated platelets that may contribute to localization of inflammation at sites of vascular injury and thrombosis.


Assuntos
Complemento C3/imunologia , Via Alternativa do Complemento/fisiologia , Ativação Plaquetária/fisiologia , Properdina/imunologia , Plaquetas/imunologia , Plaquetas/metabolismo , Complemento C3/metabolismo , Humanos , Properdina/metabolismo
8.
Mol Immunol ; 52(3-4): 133-40, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22673211

RESUMO

In Latin America, there are about 10-12 million people infected with Trypanosoma cruzi, the agent of Chagas' disease, one of the most important neglected tropical parasitism. Identification of molecular targets, specific for the aggressor or host cells or both, may be useful in the development of pharmacological and/or immunological therapeutic tools. Classic efforts in Chagas' disease explore those strategies. Although the immune system frequently controls parasite aggressions, sterile immunity is seldom achieved and chronic interactions are thus established. However, laboratory-modified immunologic probes aimed at selected parasite targets, may be more effective than their unmodified counterparts. Calreticulin (CRT) from vertebrates is a calcium binding protein, present mainly in the endoplasmic reticulum (ER), where it directs the conformation of proteins and controls calcium levels. We have isolated, gene-cloned, expressed and characterized T. cruzi calreticulin (TcCRT). Upon infection, the parasite can translocate this molecule from the ER to the surface, where it inhibits both the classical and lectin complement pathways. Moreover, by virtue of its capacity to bind and inactivate first complement component C1, it promotes parasite infectivity. These two related properties reside in the central domain of this molecule. A different domain, amino terminal, binds to endothelial cells, thus inhibiting their angiogenic capacity. Since tumor growth depends, to a large extent on angiogenesis, their growth is also inhibited.


Assuntos
Calreticulina/metabolismo , Doença de Chagas/imunologia , Doença de Chagas/parasitologia , Interações Hospedeiro-Parasita , Neoplasias/patologia , Trypanosoma cruzi/fisiologia , Inibidores da Angiogênese , Animais , Proliferação de Células , Doença de Chagas/patologia , Humanos , Neoplasias/metabolismo , Trypanosoma cruzi/imunologia , Trypanosoma cruzi/patogenicidade
9.
Trends Parasitol ; 27(3): 115-22, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21288773

RESUMO

Calreticulin (CRT) from vertebrates is a calcium-binding protein present mainly in the endoplasmic reticulum (ER). There, it directs the conformation of proteins and controls calcium levels. This review will focus on several extracellular roles of Trypanosoma cruzi CRT (TcCRT) in relation to its capacity to inhibit the complement system, mediate parasite infectivity, interfere with angiogenesis and, as a possible consequence, with tumor growth. The TcCRT antiangiogenic effect parallels with the capacity of T. cruzi infection to inhibit tumor development in vivo. Thus, the TcCRT, complement, and endothelial cell interactions seem to be an evolutionary adaptation to promote prolonged parasite-host relationships.


Assuntos
Adaptação Fisiológica/fisiologia , Calreticulina/metabolismo , Interações Hospedeiro-Parasita , Trypanosoma cruzi/patogenicidade , Animais , Trypanosoma cruzi/metabolismo
10.
Immunobiology ; 216(1-2): 265-73, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20472323

RESUMO

In Trypanosoma cruzi, calreticulin (TcCRT) translocates from the endoplasmic reticulum (ER) to the area of flagellum emergence. We propose herein that the parasite uses this molecule to capture complement C1, in an infective apoptotic mimicry strategy. Thus, TcCRT/C1 interactions, besides inhibiting the classical pathway of complement activation as previously shown in our laboratories, will also promote infectivity. This fact correlates with significant increases in TcCRT mRNA levels during early infection stages of a VERO cell line. In vitro, the collagenous and globular C1q domains simultaneously bind TcCRT and antigen aggregated Igs, respectively. Accordingly, mouse immunizations with TcCRT induced humoral responses that, after challenge, correlated with increased parasitemia. Thus, on the parasite surface, whole Igs anti-TcCRT promote C1 deposits on trypomastigotes while, as expected, F(ab')2 fragments decrease it. Likewise, pretreatment of the parasites with whole anti-TcCRT antibodies augmented parasitemia and mortality in mice. In contrast, pretreatment with F(ab')2 fragments anti-TcCRT, devoid of their capacity to provide additional C1q binding sites, was protective. Most important, while pretreatment of trypomastigotes with C1q increased infectivity in the RAW murine cell line, as well as mice mortality and parasitemia, the F(ab')2 fragments significantly interfered with the C1q-dependent infectivity. Differently from other surface molecules involved in infectivity, TcCRT uses C1 as an adaptor molecule to recognize host cells. As expected, since TcCRT is one of several cell surface parasite molecules participating in infectivity, attempts to interfere with the C1/TcCRT interactions with F(ab')2 fragments, were moderately but significantly effective, both in vitro and in vivo.


Assuntos
Complemento C1/metabolismo , Macrófagos/metabolismo , Trypanosoma cruzi/fisiologia , Animais , Complexo Antígeno-Anticorpo/metabolismo , Antígenos de Protozoários/imunologia , Calreticulina/imunologia , Linhagem Celular , Doença de Chagas , Complemento C1/imunologia , Imunidade Humoral , Imunização , Macrófagos/imunologia , Macrófagos/parasitologia , Macrófagos/patologia , Camundongos , Parasitemia , Ligação Proteica/imunologia , Trypanosoma cruzi/patogenicidade , Virulência
11.
PLoS Negl Trop Dis ; 4(7): e730, 2010 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-20625551

RESUMO

BACKGROUND: In Latin America, 18 million people are infected with Trypanosoma cruzi, the agent of Chagas' disease, with the greatest economic burden. Vertebrate calreticulins (CRT) are multifunctional, intra- and extracellular proteins. In the endoplasmic reticulum (ER) they bind calcium and act as chaperones. Since human CRT (HuCRT) is antiangiogenic and suppresses tumor growth, the presence of these functions in the parasite orthologue may have consequences in the host/parasite interaction. Previously, we have cloned and expressed T. cruzi calreticulin (TcCRT) and shown that TcCRT, translocated from the ER to the area of trypomastigote flagellum emergence, promotes infectivity, inactivates the complement system and inhibits angiogenesis in the chorioallantoid chicken egg membrane. Most likely, derived from these properties, TcCRT displays in vivo inhibitory effects against an experimental mammary tumor. METHODOLOGY AND PRINCIPAL FINDINGS: TcCRT (or its N-terminal vasostatin-like domain, N-TcCRT) a) Abrogates capillary growth in the ex vivo rat aortic ring assay, b) Inhibits capillary morphogenesis in a human umbilical vein endothelial cell (HUVEC) assay, c) Inhibits migration and proliferation of HUVECs and the human endothelial cell line Eahy926. In these assays TcCRT was more effective, in molar terms, than HuCRT: d) In confocal microscopy, live HUVECs and EAhy926 cells, are recognized by FITC-TcCRT, followed by its internalization and accumulation around the host cell nuclei, a phenomenon that is abrogated by Fucoidin, a specific scavenger receptor ligand and, e) Inhibits in vivo the growth of the murine mammary TA3 MTXR tumor cell line. CONCLUSIONS/SIGNIFICANCE: We describe herein antiangiogenic and antitumor properties of a parasite chaperone molecule, specifically TcCRT. Perhaps, by virtue of its capacity to inhibit angiogenesis (and the complement system), TcCRT is anti-inflammatory, thus impairing the antiparasite immune response. The TcCRT antiangiogenic effect could also explain, at least partially, the in vivo antitumor effects reported herein and the reports proposing antitumor properties for T. cruzi infection.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Calreticulina/farmacologia , Trypanosoma cruzi/química , Inibidores da Angiogênese/isolamento & purificação , Animais , Antineoplásicos/isolamento & purificação , Calreticulina/isolamento & purificação , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Humanos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
12.
Mol Immunol ; 47(7-8): 1516-21, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20153898

RESUMO

Trypanosoma cruzi (T. cruzi), the agent of Chagas' disease, the sixth most important neglected tropical disease worldwide, causes 50,000 deaths per year in Latin America. T. cruzi calreticulin (TcCRT), a highly pleiotropic chaperone molecule, plays important roles in several host/parasite interactions. Among other functions, we have previously shown that TcCRT, translocated from the endoplasmic reticulum to the area of flagellar emergence, binds human C1q and inhibits activation of the classical pathway in vitro. Based on a series of in vitro experiments, we propose here two mechanisms to explain how TcCRT inhibits the classical pathway at the initial stages of C1 (q, r, s) activation. First, TcCRT interacts in vitro with both solid phase bound active C1s and C1, but impairment of C4 activating capacity is evident only when the serine proteases are within the structural context of the macromolecular first component. Although C1s activity, in this context, is inhibited by TcCRT, the serine protease is not displaced from the C1 complex. Second, TcCRT prevents C1 formation, by interfering with the ability of the (C1r-C1s)(2) tetramer to bind C1q. These complement inhibitory effects are better explained by direct interaction of the parasite protein with C1, rather than by the TcCRT capacity to bind calcium, an essential element for the functional integrity of C1.


Assuntos
Calreticulina/imunologia , Proteínas do Sistema Complemento/imunologia , Proteínas de Protozoários/imunologia , Trypanosoma cruzi/imunologia , Animais , Cálcio/metabolismo , Calreticulina/metabolismo , Ativação do Complemento , Proteínas do Sistema Complemento/metabolismo , Humanos , Ligação Proteica , Proteínas de Protozoários/metabolismo , Serina Proteases/metabolismo , Trypanosoma cruzi/metabolismo
13.
Biol Res ; 43(3): 287-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21249299

RESUMO

Angiogenesis is a complex multi-step process of neovascularization arising from preexisting blood vessels whose generation is regulated by pro- and anti-angiogenic factors. Both Trypanosoma cruzi calreticulin (TcCRT) and its human counterpart (HuCRT) are antiangiogenic. This is the first report where the TcCRT and HuCRT anti-angiogenic properties are compared in vivo. In the chick embryonic chorioallantoid membrane assay (CAM) and at equimolar concentrations, TcCRT displayed significantly higher antiangiogenic activities than its human counterpart. LPS had marginal effects at the concentrations present in the recombinant protein preparations and the TcCRT antiangiogenic effects were largely inhibited by specific polyclonal antibodies, thus, reinforcing the fact that the observed TcCRT effects can be attributed to the parasite-derived molecule and not to the endotoxin. The antiangiogenic TcCRT effects correlate with its anti-tumor in vivo effects, as recently shown in our laboratory.


Assuntos
Inibidores da Angiogênese/farmacologia , Calreticulina/farmacologia , Trypanosoma cruzi/química , Inibidores da Angiogênese/isolamento & purificação , Animais , Calreticulina/isolamento & purificação , Embrião de Galinha , Humanos , Neovascularização Patológica
14.
Biol. Res ; 43(3): 287-289, 2010. graf
Artigo em Inglês | LILACS | ID: lil-571989

RESUMO

Angiogenesis is a complex multi-step process of neovascularization arising from preexisting blood vessels whose generation is regulated by pro- and anti-angiogenic factors. Both Trypanosoma cruzi calreticulin (TcCRT) and its human counterpart (HuCRT) are antiangiogenic. This is the first report where the TcCRT and HuCRT anti-angiogenic properties are compared in vivo. In the chick embryonic chorioallantoid membrane assay (CAM) and at equimolar concentrations, TcCRT displayed significantly higher antiangiogenic activities than its human counterpart. LPS had marginal effects at the concentrations present in the recombinant protein preparations and the TcCRT antiangiogenic effects were largely inhibited by specific polyclonal antibodies, thus, reinforcing the fact that the observed TcCRT effects can be attributed to the parasite-derived molecule and not to the endotoxin. The antiangiogenic TcCRT effects correlate with its anti-tumor in vivo effects, as recently shown in our laboratory.


Assuntos
Animais , Embrião de Galinha , Humanos , Inibidores da Angiogênese/farmacologia , Calreticulina/farmacologia , Trypanosoma cruzi/química , Inibidores da Angiogênese/isolamento & purificação , Calreticulina/isolamento & purificação , Neovascularização Patológica
15.
Mol Immunol ; 46(6): 1092-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19108895

RESUMO

Trypanosoma cruzi (T. cruzi) is the causative agent of Chagas' disease, an endemic and chronic illness that affects 18 million people in Latin America. The mechanisms underlying its pathogenesis are controversial. There is a growing body of evidence supporting the view that T. cruzi infection elicits severe autoimmune responses in the host, which significantly contribute to the pathogenesis of Chagas' disease, and several recent studies have reported the presence of autoantibodies and effector T lymphocytes against parasite and self antigens in infected patients and experimentally infected animals. T. cruzi calreticulin (TcCRT) is a 45kDa protein, immunogenic in humans, rabbits and mice. It has a high degree of homology with human (HuCRT) and mouse calreticulin (MoCRT), which would explain why an immune response to TcCRT could contribute to autoimmune reactions in Chagas' disease. Anti-TcCRT antibodies generated in A/J mice immunized with recombinant TcCRT (rTcCRT) reacted with rHuCRT and bound to neonatal and adult isogenic cardiomyocytes cultured in vitro. Interestingly, histological alterations, such as edema formation and cell infiltrates, which include CD3(+) cells, were detected in heart sections from immunized animals. Therefore, in rTcCRT-immunized mice, an autoimmune reaction against host CRT, paralleled by histological cardiac alterations, suggests a role of the parasite molecule in the induction of immunologically mediated heart tissue damage. The data presented here propose that TcCRT participates in the induction of cardiac autoimmunity in Chagas' disease.


Assuntos
Autoimunidade , Calreticulina/imunologia , Doença de Chagas/imunologia , Proteínas de Protozoários/imunologia , Trypanosoma cruzi/imunologia , Animais , Autoanticorpos/imunologia , Complexo CD3/imunologia , Calreticulina/genética , Células Cultivadas , Doença de Chagas/patologia , Feminino , Humanos , Camundongos , Miocárdio/patologia , Miócitos Cardíacos/patologia , Proteínas Recombinantes/imunologia
16.
Biol Res ; 38(2-3): 187-95, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16238097

RESUMO

Trypanosoma cruzi calreticulin (TcCRT), described in our laboratory, retains several important functional features from its vertebrate homologues. We have shown that recombinant TcCRT inhibits the human complement system when it binds to the collagenous portion of C1q. The generation of classical pathway convertases and membrane attack complexes is thus strongly inhibited. In most T. cruzi-infected individuals, TcCRT is immunogenic and mediates the generation of specific antibodies. By reverting the C1q / TcCRT interaction, a parasite immune evasion strategy, these antibodies contribute to the host/parasite equilibrium. In an in vitro correlate of this situation, we show that the Clq/TcCRT interaction is inhibited by F(ab')2 polyclonal anti-TcCRT IgG fragments. It is therefore feasible that in infected humans anti-TcCRT antibodies participate in reverting an important parasite strategy aimed at inhibiting the classical complement pathway. Thus, membrane-bound TcCRT interacts with the collagenous portion Clq, and this Clq is recognized by the CD91-bound host cell CRT, thus facilitating parasite internalization. Based on our in vitro results, it could be proposed that the in vivo interaction between TcCRT and vertebrate Clq could be inhibited by F(ab')2 fragments anti-rTcCRT or against its S functional domain, thus interfering with the internalization process.


Assuntos
Anticorpos Antiprotozoários/imunologia , Calreticulina/fisiologia , Complemento C1q/antagonistas & inibidores , Fragmentos Fab das Imunoglobulinas/fisiologia , Trypanosoma cruzi/imunologia , Animais , Feminino , Interações Hospedeiro-Parasita/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Coelhos
17.
Mol Biochem Parasitol ; 140(2): 133-40, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15760653

RESUMO

Angiogenesis leads to neovascularization from existing blood vessels. It is associated with tumor growth and metastasis and is regulated by pro- and antiangiogenic molecules, some of them currently under clinical trials for cancer treatment. During the last few years we have cloned, sequenced and expressed a Trypanosoma cruzi calreticulin gene (TcCRT). Its product, TcCRT, a 45 kDa protein, is more than 50% identical to human CRT (HuCRT). TcCRT, present on the surface of trypomastigotes, binds both C1q and mannan binding lectin and inhibits the classical activation pathway of human complement. Since TcCRT is highly homologous to a functional antiangiogenic fragment from HuCRT (aa 120-180), recombinant (r) and native (n) TcCRT were tested in their antiangiogenic effects, in the chick embryonic chorioallantoid membrane (CAM) assay. Both proteins mediated highly significant antiangiogenic effects in the in vivo CAM assay. This effect was further substantiated in experiments showing that the plasmid construct pSecTag/TcCRT also displayed significant antiangiogenic properties, as compared to the empty vector. Most likely, the fact that antiangiogenic substances act preferentially on growing neoplasic tissues, but not on already established tumors, is due to their effects on emerging blood vessels. The results shown here indicate that TcCRT, like its human counterpart, has antiangiogenic properties. These properties may explain, at least partly, the reported antineoplasic effect of experimental T. cruzi infection.


Assuntos
Inibidores da Angiogênese/farmacologia , Calreticulina/farmacologia , Animais , Antineoplásicos/farmacologia , Calreticulina/biossíntese , Calreticulina/genética , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/efeitos dos fármacos , Relação Dose-Resposta a Droga , Escherichia coli/genética , Escherichia coli/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/prevenção & controle , Plasmídeos , Proteínas Recombinantes/farmacologia , Trypanosoma cruzi/genética
18.
Mol Immunol ; 40(17): 1279-91, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15128045

RESUMO

Although parasites range from protozoan to complex, evolutionary advanced arthropods, in general, a hallmark of parasite life cycles is their ability to adapt to changes in temperature, pH and host defense strategies. Calreticulin, a calcium-binding protein, highly conserved and multifunctional, is present in every cell of higher organisms, except erythrocytes. The surprising array of calreticulin-associated functions include lectin-like chaperoning, calcium storage and signaling, modulation of gene expression, cell adhesion, enhancement of phagocytosis of C1q or collectin opsonized apoptotic cells, inhibition of angiogenesis and tumoral growth, inhibition of perforin pore formation in T and NK cells, and inhibition of C1q-dependent complement activation. Likewise, calreticulin is present in a wide spectrum of sub cellular compartments. Parasite calreticulin shows a surprisingly high degree of conservation within the framework of its functional domains. Its role within the parasite/host relationship needs to be assessed further, in particular with regard to its impact on parasite infectivity, by helping to evade from its hosts' immune response. With special emphasis on calreticulin from Trypanosoma cruzi, the intracellular protozoan agent of American trypanosomiasis (Chagas' disease), we wish to exemplify and highlight the various implications of parasite calreticulin, within the pathophysiology of parasite-mediated human and animal disease.


Assuntos
Cálcio/metabolismo , Calreticulina/metabolismo , Doença de Chagas/metabolismo , Interações Hospedeiro-Parasita , Trypanosoma cruzi/metabolismo , Animais , Calreticulina/genética , Proteínas do Sistema Complemento/metabolismo , Interações Hospedeiro-Parasita/fisiologia , Humanos , Chaperonas Moleculares/metabolismo , Vertebrados/parasitologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...