Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Andes Pediatr ; 94(2): 134-143, 2023 Apr.
Artigo em Espanhol | MEDLINE | ID: mdl-37358106

RESUMO

Since the American Academy of Pediatrics recommended the supine sleeping position for infants to prevent SIDS, positional plagiocephaly (PP) prevalence has increased. There are great controversies regarding the possible consequences of PP and the degree of severity required for them to manifest. There is no consensus on the efficacy of PP therapies, such as positioning, kinesiology, and cranial orthoses. This review aims to analyze the existing literature to update the causes, main characteristics, and evidence on the treatment of PP. Intervention from the newborn period is important, encompassing both prevention and management education, as well as early screening, evaluating the possible presence of congenital muscular torticollis, to start early treatment. The presence of PP can be a risk marker for psychomotor development.


Assuntos
Plagiocefalia não Sinostótica , Torcicolo , Lactente , Recém-Nascido , Humanos , Criança , Plagiocefalia não Sinostótica/diagnóstico , Plagiocefalia não Sinostótica/etiologia , Plagiocefalia não Sinostótica/terapia , Sono , Torcicolo/complicações , Prevalência
2.
Front Immunol ; 12: 789145, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975884

RESUMO

Chagas' disease is a zoonotic parasitic ailment now affecting more than 6 million people, mainly in Latin America. Its agent, the protozoan Trypanosoma cruzi, is primarily transmitted by endemic hematophagous triatomine insects. Transplacental transmission is also important and a main source for the emerging global expansion of this disease. In the host, the parasite undergoes intra (amastigotes) and extracellular infective (trypomastigotes) stages, both eliciting complex immune responses that, in about 70% of the cases, culminate in permanent immunity, concomitant with the asymptomatic presence of the parasite. The remaining 30% of those infected individuals will develop a syndrome, with variable pathological effects on the circulatory, nervous, and digestive systems. Herein, we review an important number of T. cruzi molecules, mainly located on its surface, that have been characterized as immunogenic and protective in various experimental setups. We also discuss a variety of parasite strategies to evade the complement system - mediated immune responses. Within this context, we also discuss the capacity of the T. cruzi infective trypomastigote to translocate the ER-resident chaperone calreticulin to its surface as a key evasive strategy. Herein, it is described that T. cruzi calreticulin inhibits the initial stages of activation of the host complement system, with obvious benefits for the parasite. Finally, we speculate on the possibility to experimentally intervene in the interaction of calreticulin and other T. cruzi molecules that interact with the complement system; thus resulting in significant inhibition of T. cruzi infectivity.


Assuntos
Antiprotozoários/farmacologia , Doença de Chagas/tratamento farmacológico , Interações Hospedeiro-Parasita/imunologia , Evasão da Resposta Imune/efeitos dos fármacos , Trypanosoma cruzi/imunologia , Antiprotozoários/uso terapêutico , Calreticulina/metabolismo , Doença de Chagas/imunologia , Doença de Chagas/parasitologia , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Humanos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/imunologia , Proteínas de Protozoários/metabolismo , Trypanosoma cruzi/metabolismo
3.
Front Immunol ; 11: 1561, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793217

RESUMO

Because of its capacity to increase a physiologic inflammatory response, to stimulate phagocytosis, to promote cell lysis and to enhance pathogen immunogenicity, the complement system is a crucial component of both the innate and adaptive immune responses. However, many infectious agents resist the activation of this system by expressing or secreting proteins with a role as complement regulatory, mainly inhibitory, proteins. Trypanosoma cruzi, the causal agent of Chagas disease, a reemerging microbial ailment, possesses several virulence factors with capacity to inhibit complement at different stages of activation. T. cruzi calreticulin (TcCalr) is a highly-conserved, endoplasmic reticulum-resident chaperone that the parasite translocates to the extracellular environment, where it exerts a variety of functions. Among these functions, TcCalr binds C1, MBL and ficolins, thus inhibiting the classical and lectin pathways of complement at their earliest stages of activation. Moreover, the TcCalr/C1 interaction also mediates infectivity by mimicking a strategy used by apoptotic cells for their removal. More recently, it has been determined that these Calr strategies are also used by a variety of other parasites. In addition, as reviewed elsewhere, TcCalr inhibits angiogenesis, promotes wound healing and reduces tumor growth. Complement C1 is also involved in some of these properties. Knowledge on the role of virulence factors, such as TcCalr, and their interactions with complement components in host-parasite interactions, may lead toward the description of new anti-parasite therapies and prophylaxis.


Assuntos
Calreticulina/imunologia , Complemento C1/imunologia , Interações Hospedeiro-Parasita/imunologia , Parasitos/patogenicidade , Animais , Ativação do Complemento , Humanos , Evasão da Resposta Imune , Parasitos/imunologia , Trypanosoma cruzi/imunologia , Trypanosoma cruzi/patogenicidade , Fatores de Virulência/imunologia
4.
Mol Immunol ; 124: 51-60, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32526557

RESUMO

American Trypanosomiasis, a parasitic disease produced by Trypanosoma cruzi (T. cruzi), endemic in Latin America, infects about 6 million people. During the chronic stage of the infection, approximately 30% of infected people will develop Chagas Disease, the clinical manifestation. Few decades ago it was reported that, during the chronic stage, the parasite interferes with the development of solid tumors. However, the identification of parasite molecules responsible for such effects remained elusive. Years later, we described T.cruzi Calreticulin (TcCalr), an endoplasmic reticulum resident chaperone that infective trypomastigotes translocate to the parasite exterior, where it displays anticomplement activities. Most likely, at least some of these activities are related with the antitumor properties of TcCalr, as shown in in vitro, ex vivo, in ovum, and in vivo models. In this context we, we have seen that in vivo subcutaneous peritumoral inoculation of rTcCalr enhances local infiltration of T cells and slows tumor development. Based on these precedents, we propose that in vitro treatment of a mammary adenocarcinoma (TA3 cell line) with rTcCalr, will enhance tumor immunogenicity. In agreement with this proposal, we have shown that: i). rTcCalr binds to TA3 cells in a concentration-dependent fashion, ii). C1q binds to TA3 cells in an rTcCalr-dependent fashion, confirmed by the reversion attained using anti-TcS (a central TcCalr domain that binds C1) F(ab')2 antibody fragments, iii). incubation of TA3 cells with rTcCalr, promotes cell phagocytosis by murine macrophages and, iv). rTcCalr decreases the membrane expression of MHC class II, m-Dectin-1, Galectin-9 and PD-L1, while increasing the expression of Rae-1γ. In synthesis, herein we show that in vitro treatment of a murine mammary adenocarcinoma with rTcCalr enhances phagocytosis and modulates the expression of a variety of membrane molecules that correlates with increased tumor immunogenicity.


Assuntos
Adenocarcinoma/imunologia , Antígenos de Protozoários/imunologia , Calreticulina/imunologia , Neoplasias Mamárias Experimentais/imunologia , Animais , Linhagem Celular Tumoral , Camundongos , Fagocitose/imunologia , Trypanosoma cruzi
5.
Trends Parasitol ; 36(4): 368-381, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32191851

RESUMO

To successfully infect, Trypanosoma cruzi evades and modulates the host immune response. T. cruzi calreticulin (TcCalr) is a multifunctional, endoplasmic reticulum (ER)-resident chaperone that, translocated to the external microenvironment, mediates crucial host-parasite interactions. TcCalr binds and inactivates C1 and mannose-binding lectin (MBL)/ficolins, important pattern- recognition receptors (PRRs) of the complement system. Using an apoptotic mimicry strategy, the C1-TcCalr association facilitates the infection of target cells. T. cruzi infection also seems to confer protection against tumorigenesis. Thus, recombinant TcCalr has important antiangiogenic properties, detected in vitro, ex vivo, and in ovum, most likely contributing at least in part, to its antitumor properties. Consequently, TcCalr is useful for investigating key issues of host-parasite interactions and possible new immunological/pharmacological interventions in the areas of Chagas' disease and experimental cancer.


Assuntos
Calreticulina/imunologia , Carcinogênese/imunologia , Doença de Chagas/complicações , Doença de Chagas/imunologia , Interações Hospedeiro-Parasita/imunologia , Neoplasias/etiologia , Trypanosoma cruzi/patogenicidade , Animais , Doença de Chagas/parasitologia , Doença de Chagas/patologia , Humanos , Evasão da Resposta Imune/imunologia , Neoplasias/imunologia , Trypanosoma cruzi/fisiologia , Fatores de Virulência/imunologia
6.
Immunobiology ; 225(1): 151863, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31732192

RESUMO

Microbes have developed mechanisms to resist the host immune defenses and some elicit antitumor immune responses. About 6 million people are infected with Trypanosoma cruzi, the protozoan agent of Chagas' disease, the sixth neglected tropical disease worldwide. Eighty years ago, G. Roskin and N. Klyuyeva proposed that T. cruzi infection mediates an anti-cancer activity. This observation has been reproduced by several other laboratories, but no molecular basis has been proposed. We have shown that the highly pleiotropic chaperone calreticulin (TcCalr, formerly known as TcCRT), translocates from the parasite ER to the exterior, where it mediates infection. Similar to its human counterpart HuCALR (formerly known as HuCRT), TcCalr inhibits C1 in its capacity to initiate the classical pathway of complement activation. We have also proposed that TcCalr inhibits angiogenesis and it is a likely mediator of antitumor effects. We have generated several in silico structural TcCalr models to delimit a peptide (VC-TcCalr) at the TcCalr N-domain. Chemically synthesized VC-TcCalr did bind to C1q and was anti-angiogenic in Gallus gallus chorioallantoic membrane assays. These properties were associated with structural features, as determined in silico. VC-TcCalr, a strong dipole, interacts with charged proteins such as collagen-like tails and scavenger receptors. Comparatively, HuCALR has less polarity and spatial stability, probably due to at least substitutions of Gln for Gly, Arg for Lys, Arg for Asp and Ser for Arg that hinder protein-protein interactions. These differences can explain, at least in part, how TcCalr inhibits the complement activation pathway and has higher efficiency as an antiangiogenic and antitumor agent than HuCALR.


Assuntos
Moduladores da Angiogênese/metabolismo , Antineoplásicos/metabolismo , Calreticulina/metabolismo , Doença de Chagas/imunologia , Complemento C1q/metabolismo , Proteínas de Protozoários/metabolismo , Trypanosoma cruzi/fisiologia , Moduladores da Angiogênese/química , Animais , Antineoplásicos/química , Calreticulina/química , Células Cultivadas , Doença de Chagas/parasitologia , Embrião de Galinha , Ativação do Complemento , Interações Hospedeiro-Parasita , Humanos , Simulação de Dinâmica Molecular , Estrutura Molecular , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/química , Alinhamento de Sequência
7.
Artigo em Inglês | MEDLINE | ID: mdl-30711049

RESUMO

Brucella canis is a small intracellular Gram-negative bacterium that frequently leads to chronic infections highly resistant to antibiotic therapy in dogs. Also, it causes mild human brucellosis compared to other zoonotic Brucella spp. Herein we characterize the cellular immune response elicited by B. canis by analysing human and canine CD4+ T cells after stimulation with autologous monocyte-derived dendritic cells (MoDCs). Human and canine B. canis-primed MoDCs stimulated autologous CD4+ T cells; however, a Th1 response was triggered by human MoDCs, whereas canine MoDCs induced Th1/Th17 responses, with increased CD4+ T cells producing IFN-γ and IL-17A simultaneously. Each pattern of cellular response may contribute to host susceptibility, helping to understand the differences in B. canis virulence between these two hosts. In addition, other aspects of canine immunology are unveiled by highlighting the participation of IL-17A-producing canine MoDCs and CD4+ T cells producing IFN-γ and IL-17A.


Assuntos
Brucella canis/imunologia , Linfócitos T CD4-Positivos/fisiologia , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Th1/fisiologia , Células Th17/fisiologia , Animais , Biomarcadores , Brucelose/veterinária , Comunicação Celular/imunologia , Células Dendríticas/metabolismo , Doenças do Cão/imunologia , Doenças do Cão/metabolismo , Doenças do Cão/microbiologia , Cães , Humanos , Imunofenotipagem , Ativação Linfocitária
8.
Arch Dermatol Res ; 310(8): 639-650, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30099574

RESUMO

Calreticulin is an endoplasmic reticulum-resident, calcium-binding, stress-produced, chaperone protein that serves multiple functions and is widely distributed in eukaryotic cells. Exogenously applied recombinant calreticulin solution, markedly enhanced the rate and quality of skin wound healing. These modulatory effects are more efficient than commercially available topic platelet-derived growth factor ointments (Regranex®). Trypanosoma cruzi calreticulin is more effective in equimolar terms to human counterpart in accelerating skin wound healing. While the effect of externally added recombinant parasite calreticulin on wound healing has been reported, the domains responsible for these modulatory effects have not yet been established. Here, recombinant parasite calreticulin and some of its domains were tested to assess their influence in increasing proliferation and migration of fibroblasts in vitro and rat skin wound healing in vivo. Herein, we propose that Trypanosoma cruzi whole calreticulin or some of its domains are differentially involved in the modulation of wound-healing cell migration and proliferation, and cosmetic outcome. Therefore, precise combination of the parasite protein and its domains could allow us to tailor-specific desired effects during the skin wound-healing process.


Assuntos
Calreticulina/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Proteínas de Protozoários/farmacologia , Pele/efeitos dos fármacos , Trypanosoma cruzi/metabolismo , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico , Animais , Calreticulina/genética , Calreticulina/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Masculino , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Domínios Proteicos , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Pele/lesões , Pele/metabolismo , Pele/patologia , Trypanosoma cruzi/genética , Ferimentos Penetrantes/metabolismo , Ferimentos Penetrantes/patologia
9.
Immunobiology ; 223(12): 802-806, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30131175

RESUMO

Trypanosoma cruzi calreticulin (TcCalr, formerly known as TcCRT), upon binding to Complement (C) C1 and ficolins, inhibits the classical and lectin pathways and promotes infectivity. This virulence correlates with the expression of TcCalr. The TcCalr C inhibitory capacity was shown in a previous work using a clonal epimastigote cell line from the TCC T. cruzi strain, lacking one TcCalr allele (TcCalr+/-) or over expressing it (TcCalr+). In this work, we detected atypical morphology in TcCalr+/- and in TcCalr+ parasites, as compared to the wild-type (WT) strain. Polyclonal anti-TcCalr antibodies detected TcCalr presence mainly in the parasite nucleus. The number of TcCalr indicator gold particles, detected in electron microscopy and quantified in silico, correlated with the number of TcCalr coding genes. Both TcCalr+ and TcCalr +/- epimastigotes presented morphological alterations.


Assuntos
Calreticulina/genética , Doença de Chagas/parasitologia , Dosagem de Genes , Genes de Protozoários , Trypanosoma cruzi/genética , Alelos , Animais , Animais Geneticamente Modificados , Calreticulina/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Genótipo , Humanos , Imuno-Histoquímica , Trypanosoma cruzi/citologia , Trypanosoma cruzi/ultraestrutura
10.
Vet Res ; 48(1): 72, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29096717

RESUMO

Brucella canis is a small intracellular Gram-negative bacterium whose primary host is the dog, but it also can cause mild human brucellosis. One of the main causes of an inefficient immune response against other species of Brucella is their interaction with dendritic cells (DCs), which affects antigen presentation and impairs the development of an effective Th1 immune response. This study analysed the cytokine pattern production, by RT-qPCR and ELISA, in human and canine DCs against whole B. canis or its purified LPS. Human and canine DCs produced different patterns of cytokines after stimulation with B. canis. In particular, while human DCs produced a Th1-pattern of cytokines (IL-1ß, IL-12, and TNF-α), canine cells produced both Th1 and Th17-related cytokines (IL-6, IL-12, IL-17, and IFN-γ). Thus, differences in susceptibility and pathogenicity between these two hosts could be explained, at least partly, by the distinct cytokine patterns observed in this study, where we propose that human DCs induce an effective Th1 immune response to control the infection, while canine DCs lead to a less effective immune response, with the activation of Th17-related response ineffective to control the B. canis infection.


Assuntos
Brucella canis/fisiologia , Brucelose/imunologia , Citocinas/genética , Células Dendríticas/imunologia , Animais , Brucelose/microbiologia , Citocinas/metabolismo , Células Dendríticas/microbiologia , Cães , Feminino , Humanos , Masculino
11.
Front Microbiol ; 8: 1667, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28919885

RESUMO

American Trypanosomiasis is an important neglected reemerging tropical parasitism, infecting about 8 million people worldwide. Its agent, Trypanosoma cruzi, exhibits multiple mechanisms to evade the host immune response and infect host cells. An important immune evasion strategy of T. cruzi infective stages is its capacity to inhibit the complement system activation on the parasite surface, avoiding opsonizing, immune stimulating and lytic effects. Epimastigotes, the non-infective form of the parasite, present in triatomine arthropod vectors, are highly susceptible to complement-mediated lysis while trypomastigotes, the infective form, present in host bloodstream, are resistant. Thus T. cruzi susceptibility to complement varies depending on the parasite stage (amastigote, trypomastigotes or epimastigote) and on the T. cruzi strain. To avoid complement-mediated lysis, T. cruzi trypomastigotes express on the parasite surface a variety of complement regulatory proteins, such as glycoprotein 58/68 (gp58/68), T. cruzi complement regulatory protein (TcCRP), trypomastigote decay-accelerating factor (T-DAF), C2 receptor inhibitor trispanning (CRIT) and T. cruzi calreticulin (TcCRT). Alternatively, or concomitantly, the parasite captures components with complement regulatory activity from the host bloodstream, such as factor H (FH) and plasma membrane-derived vesicles (PMVs). All these proteins inhibit different steps of the classical (CP), alternative (AP) or lectin pathways (LP). Thus, TcCRP inhibits the CP C3 convertase assembling, gp58/68 inhibits the AP C3 convertase, T-DAF interferes with the CP and AP convertases assembling, TcCRT inhibits the CP and LP, CRIT confers ability to resist the CP and LP, FH is used by trypomastigotes to inhibit the AP convertases and PMVs inhibit the CP and LP C3 convertases. Many of these proteins have similar molecular inhibitory mechanisms. Our laboratory has contributed to elucidate the role of TcCRT in the host-parasite interplay. Thus, we have proposed that TcCRT is a pleiotropic molecule, present not only in the parasite endoplasmic reticulum, but also on the trypomastigote surface, participating in key processes to establish T. cruzi infection, such as inhibition of the complement system and serving as an important virulence factor. Additionally, TcCRT interaction with key complement components, participates as an anti-angiogenic and anti-tumor molecule, inhibiting at least in important part, tumor growth in infected animals.

12.
Int J Oncol ; 50(4): 1261-1270, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28260038

RESUMO

Ovarian cancer is the seventh most common cancer among women worldwide, causing approximately 120,000 deaths every year. Immunotherapy, designed to boost the body's natural defenses against cancer, appears to be a promising option against ovarian cancer. Calreticulin (CRT) is an endoplasmic reticulum (ER) resident chaperone that, translocated to the cell membrane after ER stress, allows cancer cells to be recognized by the immune system. The nerve growth factor (NGF) is a pro-angiogenic molecule overexpressed in this cancer. In the present study, we aimed to determine weather NGF has an effect in CRT translocation induced by cytotoxic and ER stress. We treated A2780 ovarian cancer cells with NGF, thapsigargin (Tg), an ER stress inducer and mitoxantrone (Mtx), a chemotherapeutic drug; CRT subcellular localization was analyzed by immunofluorescence followed by confocal microscopy. In order to determine NGF effect on Mtx and Tg-induced CRT translocation from the ER to the cell membrane, cells were preincubated with NGF prior to Mtx or Tg treatment and CRT translocation to the cell surface was determined by flow cytometry. In addition, by western blot analyses, we evaluated proteins associated with the CRT translocation pathway, both in A2780 cells and human ovarian samples. We also measured NGF effect on cell apoptosis induced by Mtx. Our results indicate that Mtx and Tg, but not NGF, induce CRT translocation to the cell membrane. NGF, however, inhibited CRT translocation induced by Mtx, while it had no effect on Tg-induced CRT exposure. NGF also diminished cell death induced by Mtx. NGF effect on CRT translocation could have consequences in immunotherapy, potentially lessening the effectiveness of this type of treatment.


Assuntos
Calreticulina/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Neoplasias Epiteliais e Glandulares/imunologia , Fator de Crescimento Neural/metabolismo , Neoplasias Ovarianas/imunologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Calreticulina/imunologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Citometria de Fluxo , Humanos , Imunoterapia , Mitoxantrona/farmacologia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais , Tapsigargina/farmacologia
13.
Front Immunol ; 8: 188, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28286504

RESUMO

Molluskan hemocyanins are enormous oxygen-carrier glycoproteins that show remarkable immunostimulatory properties when inoculated in mammals, such as the generation of high levels of antibodies, a strong cellular reaction, and generation of non-specific antitumor immune responses in some types of cancer, particularly for superficial bladder cancer. These proteins have the ability to bias the immune response toward a Th1 phenotype. However, despite all their current uses with beneficial clinical outcomes, a clear mechanism explaining these properties is not available. Taking into account reports of natural antibodies against the hemocyanin of the gastropod Megathura crenulata [keyhole limpet hemocyanin (KLH)] in humans as well as other vertebrate species, we report here for the first time, the presence, in sera from unimmunized healthy donors, of antibodies recognizing, in addition to KLH, two other hemocyanins from gastropods with documented immunomodulatory capacities: Fisurella latimarginata hemocyanin (FLH) and Concholepas concholepas hemocyanin (CCH). Through an ELISA screening, we found IgM and IgG antibodies reactive with these hemocyanins. When the capacity of these antibodies to bind deglycosylated hemocyanins was studied, no decreased interaction was detected. Moreover, in the case of FLH, deglycosylation increased antibody binding. We evaluated through an in vitro complement deposition assay whether these antibodies activated the classical pathway of the human complement system. The results showed that all three hemocyanins and their deglycosylated counterparts elicited this activation, mediated by C1 binding to immunoglobulins. Thus, this work contributes to the understanding on how the complement system could participate in the immunostimulatory properties of hemocyanins, through natural, complement-activating antibodies reacting with these proteins. Although a role for carbohydrates cannot be completely ruled out, in our experimental setting, glycosylation status had a limited effect. Finally, our data open possibilities for further studies leading to the design of improved hemocyanin-based research tools for diagnosis and immunotherapy.

14.
Immunobiology ; 222(3): 529-535, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27839837

RESUMO

Chagas disease is an endemic pathology in Latin America, now emerging in developed countries, caused by the intracellular protozoan Trypanosoma cruzi, whose life cycle involves three stages: amastigotes, epimastigotes, and trypomastigotes. T. cruzi Calreticulin (TcCRT), an endoplasmic reticulum resident chaperone, translocates to the external cellular membrane, where it captures complement component C1, ficolins and MBL, thus inactivating the classical and lectin pathways. Trypomastigote-bound C1 is detected as an "eat me" signal by macrophages and promotes the infective process. Unlike infective trypomastigotes, non-infective epimastigotes either do not express or express only marginal levels of TcCRT on their external membrane. We show that epimastigotes bind exogenous rTcCRT to their cellular membrane and, in the presence of C1q, this parasite form is internalized into normal fibroblasts. On the other hand, Calreticulin (CRT)-deficient fibroblasts show impaired parasite internalization. In synthesis, CRT from both parasite and host cell origin is important in the establishment of C1q-dependent first contacts between parasites and host cells.


Assuntos
Calreticulina/imunologia , Endocitose/imunologia , Interações Hospedeiro-Parasita/imunologia , Trypanosoma cruzi/imunologia , Adjuvantes Imunológicos , Animais , Calreticulina/genética , Calreticulina/metabolismo , Membrana Celular/imunologia , Membrana Celular/metabolismo , Doença de Chagas/imunologia , Doença de Chagas/parasitologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Fibroblastos/metabolismo , Fibroblastos/parasitologia , Técnicas de Inativação de Genes , Camundongos , Ligação Proteica , Trypanosoma cruzi/metabolismo , Trypanosoma cruzi/patogenicidade , Fatores de Virulência/imunologia
15.
Am J Trop Med Hyg ; 96(2): 295-303, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-27895277

RESUMO

Triatoma infestans is an important hematophagous vector of Chagas disease, a neglected chronic illness affecting approximately 6 million people in Latin America. Hematophagous insects possess several molecules in their saliva that counteract host defensive responses. Calreticulin (CRT), a multifunctional protein secreted in saliva, contributes to the feeding process in some insects. Human CRT (HuCRT) and Trypanosoma cruzi CRT (TcCRT) inhibit the classical pathway of complement activation, mainly by interacting through their central S domain with complement component C1. In previous studies, we have detected CRT in salivary gland extracts from T. infestans We have called this molecule TiCRT. Given that the S domain is responsible for C1 binding, we have tested its role in the classical pathway of complement activation in vertebrate blood. We have cloned and characterized the complete nucleotide sequence of CRT from T. infestans, and expressed its S domain. As expected, this S domain binds to human C1 and, as a consequence, it inhibits the classical pathway of complement, at its earliest stage of activation, namely the generation of C4b. Possibly, the presence of TiCRT in the salivary gland represents an evolutionary adaptation in hematophagous insects to control a potential activation of complement proteins, present in the massive blood meal that they ingest, with deleterious consequences at least on the anterior digestive tract of these insects.


Assuntos
Calreticulina/genética , Proteínas do Sistema Complemento/imunologia , Interações Hospedeiro-Parasita/genética , Triatoma/genética , Animais , Galinhas/parasitologia , Clonagem Molecular , Complemento C1/imunologia , Expressão Gênica , Alinhamento de Sequência , Análise de Sequência de DNA
16.
IUCrJ ; 3(Pt 6): 408-419, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27840680

RESUMO

Calreticulin (CRT) is a multifaceted protein, initially discovered as an endoplasmic reticulum (ER) chaperone protein, that is essential in calcium metabolism. Various implications in cancer, early development and immunology have been discovered more recently for CRT, as well as its role as a dominant 'eat-me' prophagocytic signal. Intriguingly, cell-surface exposure/secretion of CRT is among the infective strategies used by parasites such as Trypanosoma cruzi, Entamoeba histolytica, Taenia solium, Leishmania donovani and Schistosoma mansoni. Because of the inherent flexibility of CRTs, their analysis by X-ray crystallography requires the design of recombinant constructs suitable for crystallization, and thus only the structures of two very similar mammalian CRT lectin domains are known. With the X-ray structures of two distant parasite CRTs, insights into species structural determinants that might be harnessed to fight against the parasites without affecting the functions of the host CRT are now provided. Moreover, although the hypothesis that CRT can exhibit both open and closed conformations has been proposed in relation to its chaperone function, only the open conformation has so far been observed in crystal structures. The first evidence is now provided of a complex conformational transition with the junction reoriented towards P-domain closure. SAXS experiments also provided additional information about the flexibility of T. cruzi CRT in solution, thus complementing crystallographic data on the open conformation. Finally, regarding the conserved lectin-domain structure and chaperone function, evidence is provided of its dual carbohydrate/protein specificity and a new scheme is proposed to interpret such unusual substrate-binding properties. These fascinating features are fully consistent with previous experimental observations, as discussed considering the broad spectrum of CRT sequence conservations and differences.

17.
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
18.
Front Immunol ; 7: 268, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27462315

RESUMO

Eight to 10 million people in 21 endemic countries are infected with Trypanosoma cruzi. However, only 30% of those infected develop symptoms of Chagas' disease, a chronic, neglected tropical disease worldwide. Similar to other pathogens, T. cruzi has evolved to resist the host immune response. Studies, performed 80 years ago in the Soviet Union, proposed that T. cruzi infects tumor cells with similar capacity to that displayed for target tissues such as cardiac, aortic, or digestive. An antagonistic relationship between T. cruzi infection and cancer development was also proposed, but the molecular mechanisms involved have remained largely unknown. Probably, a variety of T. cruzi molecules is involved. This review focuses on how T. cruzi calreticulin (TcCRT), exteriorized from the endoplasmic reticulum, targets the first classical complement component C1 and negatively regulates the classical complement activation cascade, promoting parasite infectivity. We propose that this C1-dependent TcCRT-mediated virulence is critical to explain, at least an important part, of the parasite capacity to inhibit tumor development. We will discuss how TcCRT, by directly interacting with venous and arterial endothelial cells, inhibits angiogenesis and tumor growth. Thus, these TcCRT functions not only illustrate T. cruzi interactions with the host immune defensive strategies, but also illustrate a possible co-evolutionary adaptation to privilege a prolonged interaction with its host.

19.
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
20.
J Tissue Eng Regen Med ; 9(1): 41-54, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23109509

RESUMO

In orthopaedics, the use of factors that enhance granulation tissue formation and prevent or delay new bone regeneration is sometimes desirable. Calreticulin (CRT), a unique endoplasmic reticulum luminal Ca(2+) -binding chaperone widely distributed in eukaryotic cells, is involved in many cellular functions. Among them, CRT has an important influence in cutaneous wound healing and diverse processes associated with cutaneous repair, inhibition of angiogenesis, promotion of cell adhesion and antitumour effect. One of the molecules involved in several aspects of the host-parasite interplay is Trypanosoma cruzi calreticulin (TcCRT), which is highly homologous to human calreticulin (HuCRT). Here, recombinant (r)HuCRT and rTcCRT are compared on their abilities to affect fibroblast behaviour in a scratch plate assay, and wound healing in in vivo skin rat models. In molar terms, rTcCRT is three orders of magnitude more efficient than rHuCRT in increasing proliferation and migration of human fibroblasts in vitro. A similar effect was observed in vivo on rat skin wounds and inhibition of bone gap bridging in rabbit unicortical bone osteotomies.


Assuntos
Calreticulina/metabolismo , Trypanosoma cruzi/metabolismo , Cicatrização , Animais , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Cálcio/química , Adesão Celular , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Quitosana/química , Reagentes de Ligações Cruzadas/química , Derme/metabolismo , Retículo Endoplasmático/metabolismo , Fibroblastos/metabolismo , Glutaral/química , Tecido de Granulação/patologia , Humanos , Masculino , Chaperonas Moleculares/química , Músculos/metabolismo , Neovascularização Patológica , Coelhos , Ratos , Proteínas Recombinantes/química , Regeneração , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...