Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Rev Med Virol ; 31(2): e2157, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32844538

RESUMO

Understanding Covid-19 pathophysiology is crucial for a better understanding of the disease and development of more effective treatments. Alpha-1-antitrypsin (A1AT) is a constitutive tissue protector with antiviral and anti-inflammatory properties. A1AT inhibits SARS-CoV-2 infection and two of the most important proteases in the pathophysiology of Covid-19: the transmembrane serine protease 2 (TMPRSS2) and the disintegrin and metalloproteinase 17 (ADAM17). It also inhibits the activity of inflammatory molecules, such as IL-8, TNF-α, and neutrophil elastase (NE). TMPRSS2 is essential for SARS-CoV-2-S protein priming and viral infection. ADAM17 mediates ACE2, IL-6R, and TNF-α shedding. ACE2 is the SARS-CoV-2 entry receptor and a key component for the balance of the renin-angiotensin system, inflammation, vascular permeability, and pulmonary homeostasis. In addition, clinical findings indicate that A1AT levels might be important in defining Covid-19 outcomes, potentially partially explaining associations with air pollution and with diabetes. In this review, we focused on the interplay between A1AT with TMPRSS2, ADAM17 and immune molecules, and the role of A1AT in the pathophysiology of Covid-19, opening new avenues for investigating effective treatments.


Assuntos
COVID-19/metabolismo , alfa 1-Antitripsina/metabolismo , Proteína ADAM17/metabolismo , Animais , Humanos , Fatores de Proteção , Serina Endopeptidases/metabolismo
2.
Cell Commun Signal ; 18(1): 190, 2020 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-33357215

RESUMO

The rapid ability of SARS-CoV-2 to spread among humans, along with the clinical complications of coronavirus disease 2019-COVID-19, have represented a significant challenge to the health management systems worldwide. The acute inflammation and coagulation abnormalities appear as the main causes for thousands of deaths worldwide. The intense inflammatory response could be involved with the formation of thrombi. For instance, the presence of uncleaved large multimers of von Willebrand (vWF), due to low ADAMTS13 activity in plasma could be explained by the inhibitory action of pro-inflammatory molecules such as IL-1ß and C reactive protein. In addition, the damage to endothelial cells after viral infection and/or activation of endothelium by pro-inflammatory cytokines, such as IL-1ß, IL-6, IFN-γ, IL-8, and TNF-α induces platelets and monocyte aggregation in the vascular wall and expression of tissue factor (TF). The TF expression may culminate in the formation of thrombi, and activation of cascade by the extrinsic pathway by association with factor VII. In this scenario, the phosphatidylserine-PtdSer exposure on the outer leaflet of the cell membrane as consequence of viral infection emerges as another possible underlying mechanism to acute immune inflammatory response and activation of coagulation cascade. The PtdSer exposure may be an important mechanism related to ADAM17-mediated ACE2, TNF-α, EGFR and IL-6R shedding, and the activation of TF on the surface of infected endothelial cells. In this review, we address the underlying mechanisms involved in the pathophysiology of inflammation and coagulation abnormalities. Moreover, we introduce key biochemical and pathophysiological concepts that support the possible participation of PtdSer exposure on the outer side of the SARS-CoV-2 infected cells membrane, in the pathophysiology of COVID-19. Video Abstract.


Assuntos
COVID-19/genética , Inflamação/genética , Fosfatidilserinas/genética , Trombose/genética , Proteína ADAM17/genética , Proteína ADAMTS13/genética , COVID-19/complicações , COVID-19/patologia , COVID-19/virologia , Células Endoteliais/virologia , Humanos , Inflamação/complicações , Inflamação/virologia , Fosfatidilserinas/metabolismo , Receptores de Interleucina-6/genética , SARS-CoV-2/patogenicidade , Trombose/patologia , Trombose/virologia , Fator de von Willebrand/genética
3.
Front Immunol ; 11: 576745, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117379

RESUMO

The Coronavirus Disease 2019 (COVID-19) has already caused hundreds of thousands of deaths worldwide in a few months. Cardiovascular disease, hypertension, diabetes and chronic lung disease have been identified as the main COVID-19 comorbidities. Moreover, despite similar infection rates between men and women, the most severe course of the disease is higher in elderly and co-morbid male patients. Therefore, the occurrence of specific comorbidities associated with renin-angiotensin system (RAS) imbalance mediated by the interaction between angiotensin-converting enzyme 2 (ACE2) and desintegrin and metalloproteinase domain 17 (ADAM17), along with specific genetic factors mainly associated with type II transmembrane serine protease (TMPRSS2) expression, could be decisive for the clinical outcome of COVID-19. Indeed, the exacerbated ADAM17-mediated ACE2, TNF-α, and IL-6R secretion emerges as a possible underlying mechanism for the acute inflammatory immune response and the activation of the coagulation cascade. Therefore, in this review, we focus on the main pathophysiological aspects of ACE2, ADAM17, and TMPRSS2 host proteins in COVID-19. Additionally, we discuss a possible mechanism to explain the deleterious effect of ADAM17 and TMPRSS2 over-activation in the COVID-19 outcome.


Assuntos
Proteína ADAM17/metabolismo , Infecções por Coronavirus/patologia , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/patologia , Serina Endopeptidases/metabolismo , Idoso , Envelhecimento , Enzima de Conversão de Angiotensina 2 , Betacoronavirus , COVID-19 , Comorbidade , Feminino , Humanos , Masculino , Pandemias , Receptores de Interleucina-6/metabolismo , Fatores de Risco , SARS-CoV-2 , Fator de Necrose Tumoral alfa/metabolismo
4.
J Med Virol ; 92(8): 1148-1157, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31825106

RESUMO

The progression of AIDS depends on the complex host and virus interactions. The most important disease progression hallmarks are immune activation and apoptosis. In this study, we address the prevalence of polymorphisms related to proinflammatory and apoptotic genes, such as IFNG (+874T/A), TNF (308G/A), IL6 (-174G/C), IL8 (-251A/T), FAS (-670A/G), and FASL (-124A/G) in 160 ethnically mixed HIV-1-infected patients from multicentre cohorts with different clinical outcomes (13 elite controllers [EC], 66 slow long-term non-progressors [LTNPs], and 81 progressors [P]). The genotyping was accomplished by TaqMan-qPCR. Among all the polymorphisms analyzed in the cytokines, the IL6 -174G/C polymorphism showed a higher frequency of GG genotype in the LTNP and LTNP+EC groups as compared to the P group. Moreover, there was a significantly higher frequency of the G allele in the LTNP and LTNP+EC groups as compared to the P group. On the other hand, the levels of CD4+ T lymphocytes were higher among individuals showing the AA and AG genotypes for the FASL -124A/G polymorphism as compared to the GG genotype. Furthermore, the AG and AA genotypes were more frequent, as compared to the GG genotype, in individuals showing a lower viral load. In contrast, for the FAS -670A/G polymorphism, a significantly higher viral load was observed in individuals with the AG genotype as compared to the GG genotype. In conclusion, we found three genetic allelic variants of the IL6 -174G/C, FASL -124A/G, and FAS -670A/G polymorphisms that were related to disease progression and immunological and virological markers in cohorts of HIV-1-positive ethnically mixed patients.


Assuntos
Proteína Ligante Fas/genética , Soropositividade para HIV/genética , Soropositividade para HIV/imunologia , Interleucina-6/genética , Receptor fas/genética , Adulto , Progressão da Doença , Etnicidade , Proteína Ligante Fas/imunologia , Feminino , Predisposição Genética para Doença , Genótipo , Soropositividade para HIV/etnologia , HIV-1/genética , HIV-1/imunologia , Humanos , Interleucina-6/imunologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Adulto Jovem , Receptor fas/imunologia
5.
Clin Immunol ; 209: 108266, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31669192

RESUMO

Host genetics affects both susceptibility and progression of HIV-1 infection. NLRP3 inflammasome provides a first-line defense in viral infections, and, accordingly, gain-of-function variants in NLRP3 have been associated with protection against HIV-1. Despite antiretroviral treatment (ART), HIV-infected patients continue to present systemic inflammation with a heterogeneous prognosis. As NLRP3 inflammasome is involved in several chronic diseases by amplifying "sterile" inflammation, its role in chronic phase of HIV infection has been postulated. Little is known about inflammasome genetics in HIV-infected patients and whether it may play a role in the different clinical outcomes. Therefore, we questioned whether NLRP3 inflammasome genetics could affect the clinical course of HIV-1 infection as it does in host/virus interaction. For this purpose, we analyzed selected single nucleotide polymorphisms (SNPs) in ART-treated HIV-infected patients (n = 300), in Long Term Non-Progressors/Elite Controllers and progressors (n = 133), and in HIV-infected individuals submitted to dendritic cell (DC)-based immunotherapy (n = 19). SNPs leading to increased activation of NLRP3 inflammasome are beneficial for patients, while SNPs that negatively affect NLRP3 activation or IL-18 production, detrimental. In contrast, gain-of-function variant in IL1B is also detrimental for patients, suggesting that while IL-1ß possible contributes to immune exhaustion, the axis NLRP3-inflammasome/IL-18 could act positively in chronic infection. Functional assays supported genetic results: NLRP3 variants associated with good quality HIV+ DC, and IL1B -511C > T with a poor one. Loss-of-function SNPs affect HIV+ T cells proliferation. These findings proposed for the first time that NLRP3 inflammasome, mainly through IL-18, play a protective role in chronic HIV infection.


Assuntos
Infecções por HIV/genética , Interleucina-18/genética , Interleucina-1beta/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Adulto , Progressão da Doença , Feminino , HIV-1/patogenicidade , Humanos , Inflamassomos/genética , Inflamação/genética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética
6.
Rev Med Virol ; 29(5): e2057, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31179598

RESUMO

During human immunodeficiency virus (HIV) infection, Nef viral protein plays a crucial role in viral pathogenesis and progression of acquired immunodeficiency syndrome. Nef is expressed in the early stages of infection and alters the cellular environment increasing infectivity, viral replication, and the evasion of host immune response through several mechanisms. Nef has numerous functional domains that allow it to interact with a number of proteins, interfering with intracellular traffic. Among these proteins, human peroxisomal thioesterase 8, ACOT8, has been shown to be an important cellular partner of Nef. It has been suggested that this interaction may be involved in Nef-dependent endocytosis and also in the modulation of lipid composition in membrane rafts. However, the actual role of this interaction, as well as the mechanisms involved, has not yet been fully elucidated. In this review, we focused on the interplay between Nef and ACOT8 proteins, highlighting the possible physiological relevance in HIV infection.


Assuntos
Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Palmitoil-CoA Hidrolase/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Biomarcadores , Humanos , Ligação Proteica
7.
Rev Med Virol ; 18(1): 5-18, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17764099

RESUMO

Life-prolonging antiretroviral therapy remarkably reduces viral load, but it does not eradicate the virus. An important obstacle preventing virus clearance is the presence of latent virion reservoirs in the host. However, new promising antiviral approaches are emerging, and a number of host cell factors involved in the disease progression and control of HIV-1 replication have been recently discovered. For instance, the RNA interference (RNAi) mechanism, besides many functions conserved throughout evolution, works as a defence mechanism against noxious transcripts which may provide a new tool to block viral replication. The recent definition of basic RNAi mechanisms, as well as the discovery of micro RNAs (microRNAs) encoded by the host cell genome and by HIV-1, also suggest that RNAi may be involved in the control of HIV replication.


Assuntos
Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/fisiologia , Interferência de RNA , RNA Interferente Pequeno , Regulação Viral da Expressão Gênica , Terapia Genética/métodos , Humanos , RNA Viral/genética , Latência Viral , Replicação Viral
8.
Virol J ; 4: 57, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17559673

RESUMO

HIV-1 Vpr is a viral accessory protein that activates ATR through the induction of DNA replication stress. ATR activation results in cell cycle arrest in G2 and induction of apoptosis. In the present study, we investigate the role of the ubiquitin/proteasome system (UPS) in the above activity of Vpr. We report that the general function of the UPS is required for Vpr to induce G2 checkpoint activation, as incubation of Vpr-expressing cells with proteasome inhibitors abolishes this effect. We further investigated in detail the specific E3 ubiquitin ligase subunits that Vpr manipulates. We found that Vpr binds to the DCAF1 subunit of a cullin 4a/DDB1 E3 ubiquitin ligase. The carboxy-terminal domain Vpr(R80A) mutant, which is able to bind DCAF1, is inactive in checkpoint activation and has dominant-negative character. In contrast, the mutation Q65R, in the leucine-rich domain of Vpr that mediates DCAF1 binding, results in an inactive Vpr devoid of dominant negative behavior. Thus, the interaction of Vpr with DCAF1 is required, but not sufficient, for Vpr to cause G2 arrest. We propose that Vpr recruits, through its carboxy terminal domain, an unknown cellular factor that is required for G2-to-M transition. Recruitment of this factor leads to its ubiquitination and degradation, resulting in failure to enter mitosis.


Assuntos
Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Linhagem Celular , Fase G2 , Produtos do Gene vpr/isolamento & purificação , HIV-1/efeitos dos fármacos , Células HeLa/citologia , Células HeLa/fisiologia , Células HeLa/virologia , Humanos , Rim , Oligopeptídeos/farmacologia , RNA Interferente Pequeno/genética , RNA Viral/genética , Transfecção , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
9.
Mem. Inst. Oswaldo Cruz ; 101(8): 833-843, Dec. 2006. ilus
Artigo em Inglês | LILACS | ID: lil-440569

RESUMO

The horizontal transfer of Trypanosoma cruzi mitochondrial minicircle DNA to the genomes of naturally infected humans may play an important role in the pathogenesis of Chagas disease. Minicircle integrations within LINE-1 elements create the potential for foreign DNA mobility within the host genome via the machinery associated with this retrotransposon. Here we document integration of minicircle DNA fragments in clonal human macrophage cell lines and their mobilization over time. The movement of an integration event in a clonal transfected cell line was tracked at three months and three years post-infection. The minicircle sequence integrated into a LINE-1 retrotransposon; one such foreign fragment subsequently relocated to another genomic location in association with associated LINE-1 elements. The p15 locus was altered at three years as a direct effect of minicircle/LINE-1 acquisition, resulting in elimination of p15 mRNA. Here we show for the first time a molecular pathology stemming from mobilization of a kDNA/LINE-1 mutation. These genomic changes and detected transcript variations are consistent with our hypothesis that minicircle integration is a causal component of parasite-independent, autoimmune-driven lesions seen in the heart and other target tissues associated with Chagas disease.


Assuntos
Humanos , Animais , DNA de Cinetoplasto/genética , Expressão Gênica/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Retroelementos/genética , Trypanosoma cruzi/genética , Linhagem Celular/parasitologia , Transferência Genética Horizontal , Interações Hospedeiro-Parasita/genética , Macrófagos/parasitologia , Trypanosoma cruzi/fisiologia
10.
Mem Inst Oswaldo Cruz ; 101(8): 833-43, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17293976

RESUMO

The horizontal transfer of Trypanosoma cruzi mitochondrial minicircle DNA to the genomes of naturally infected humans may play an important role in the pathogenesis of Chagas disease. Minicircle integrations within LINE-1 elements create the potential for foreign DNA mobility within the host genome via the machinery associated with this retrotransposon. Here we document integration of minicircle DNA fragments in clonal human macrophage cell lines and their mobilization over time. The movement of an integration event in a clonal transfected cell line was tracked at three months and three years post-infection. The minicircle sequence integrated into a LINE-1 retrotransposon; one such foreign fragment subsequently relocated to another genomic location in association with associated LINE-1 elements. The p15 locus was altered at three years as a direct effect of minicircle/LINE-1 acquisition, resulting in elimination of p15 mRNA. Here we show for the first time a molecular pathology stemming from mobilization of a kDNA/LINE-1 mutation. These genomic changes and detected transcript variations are consistent with our hypothesis that minicircle integration is a causal component of parasite-independent, autoimmune-driven lesions seen in the heart and other target tissues associated with Chagas disease.


Assuntos
DNA de Cinetoplasto/genética , Expressão Gênica/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Retroelementos/genética , Trypanosoma cruzi/genética , Animais , Sequência de Bases , Linhagem Celular/parasitologia , Transferência Genética Horizontal , Interações Hospedeiro-Parasita/genética , Humanos , Macrófagos/parasitologia , Dados de Sequência Molecular , Trypanosoma cruzi/fisiologia
11.
J Virol ; 78(23): 13072-81, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15542659

RESUMO

CD4 down-modulation is essential for the production of human immunodeficiency virus (HIV) infectious particles. Disease progression correlates with enhanced viral induced CD4 down-modulation, and a subset of long-term nonprogressors carry viruses defective in this function. Despite multiple pieces of evidence highlighting the importance of this function in viral pathogenesis in vivo, to date, HIV-induced CD4 down-modulation has not been used as a target for intervention. We describe here HIV-based vectors that deliver truncated CD4 molecules resistant to down-modulation by the viral products Nef and Vpu. Infection of cells previously transduced with these vectors proceeded normally, and viral particles were released in normal amounts. However, the infectivity of the released virions was reduced 1,000-fold. Lentiviral vectors expressing truncated CD4 molecules were efficient at blocking HIV-1 infectivity and replication in several cell lines and in CD4-positive primary lymphocytes. The findings presented here provide proof-of-principle that approaches targeting the virus-induced CD4 down-modulation may constitute the basis for novel anti-HIV therapies.


Assuntos
Antígenos CD4/análise , Vetores Genéticos/fisiologia , HIV-1/fisiologia , Linfócitos/virologia , Replicação Viral , Regulação para Baixo , Produtos do Gene nef/fisiologia , Proteínas do Vírus da Imunodeficiência Humana , Humanos , Proteínas Virais Reguladoras e Acessórias/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana
12.
J Biol Chem ; 278(36): 33912-9, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12816953

RESUMO

Three viral proteins participate in the down-modulation of CD4 in human immunodeficiency virus type 1 (HIV-1)-infected cells. The underlying mechanisms have been extensively investigated. However, the physiological relevance of this phenomenon remains poorly understood. To address the role of CD4 down-modulation in HIV-1 pathogenesis in vivo, we have characterized the functional properties of nef alleles isolated from seven HIV-1-infected patients at either the stage of AIDS (late alleles) or during the asymptomatic phase of infection (early alleles). HIV-1 variants carrying these nef alleles showed striking differences in CD4 down-modulation, virus infectivity, and replication properties. Infection of T cells with late strains resulted in production of viral particles with enhanced infectivity, as compared with variants carrying early nef alleles. These differences in infectivity were observed only when viruses were produced in cells with high levels of the viral receptor, suggesting a functional link between CD4 levels and the ability of Nef to down-modulate CD4 and to enhance viral infectivity. Similarly, late nef alleles were substantially more active than early nef genes in stimulating HIV-1 replication in high CD4-positive cells, including primary lymphocytes, but not in cells expressing low levels of the CD4 receptor. Single-round assays showed that differences in infectivity between late and early strains are largely reduced when evaluated in target cells with high levels of CD4, suggesting that the inhibitory effect occurs at the entry step. Supporting this, enhanced CD4 down-modulation by late nef alleles was associated with higher levels of envelope incorporation into viral particles, a phenomenon that likely accounted for the augmented infectivity. Our data suggest a mechanistic link between the Nef-mediated CD4 down-modulation and the enhancement of replication in CD4-positive lymphocytes. As progression to disease occurs, HIV-1 Nef variants with enhanced ability to down-modulate CD4 are selected. These strains efficiently overcome the deleterious effects of CD4 and replicate more aggressively in CD4-positive primary lymphocytes. These results highlight the importance of the virus-induced CD4 down-modulation in HIV-1 pathogenesis.


Assuntos
Antígenos CD4/fisiologia , Regulação para Baixo , Produtos do Gene env/metabolismo , Produtos do Gene nef/metabolismo , Alelos , Antígenos CD4/biossíntese , Antígenos CD4/metabolismo , Linhagem Celular , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Produtos do Gene nef/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/metabolismo , Humanos , Células Jurkat , Leucócitos Mononucleares/metabolismo , Linfócitos/virologia , Fatores de Tempo , Transfecção , Produtos do Gene nef do Vírus da Imunodeficiência Humana
13.
Rev. Inst. Med. Trop. Säo Paulo ; 43(5): 271-276, Sept.-Oct. 2001. ilus, tab
Artigo em Inglês | LILACS | ID: lil-308001

RESUMO

Trypanosoma cruzi (Schyzotrypanum, Chagas, 1909), and Chagas disease are endemic in captive-reared baboons at the Southwest Foundation for Biomedical Research, San Antonio, Texas. We obtained PCR amplification products from DNA extracted from sucking lice collected from the hair and skin of T. cruzi-infected baboons, with specific nested sets of primers for the protozoan kinetoplast DNA, and nuclear DNA. These products were hybridized to their complementary internal sequences. Selected sequences were cloned and sequencing established the presence of T. cruzi nuclear DNA, and minicircle kDNA. Competitive PCR with a kDNA set of primers determined the quantity of approximately 23.9 18.2 T. cruzi per louse. This finding suggests that the louse may be a vector incidentally contributing to the dissemination of T. cruzi infection in the baboon colony


Assuntos
Animais , Vetores de Doenças , Infestações por Piolhos , Papio , Ftirápteros , Trypanosoma cruzi , Primers do DNA , DNA de Protozoário , Reação em Cadeia da Polimerase , Trypanosoma cruzi
14.
Rev. Inst. Med. Trop. Säo Paulo ; 42(3): 157-61, May-Jun. 2000. ilus, tab
Artigo em Inglês | LILACS | ID: lil-262695

RESUMO

We used a molecular method and demonstrated that treatment of the chronic human Trypanosoma cruzi infections with nitroderivatives did not lead to parasitological cure. Seventeen treated and 17 untreated chronic Chagas' disease patients, with at least two out of three positive serologic assays for the infection, and 17 control subjects formed the study groups. PCR assays with nested sets of T. cruzi DNA primers monitored the efficacy of treatment. The amplification products were hybridized to their complementary internal sequences. Untreated and treated Chagas' disease patients yielded PCR amplification products with T. cruzi nuclear DNA primers. Competitive PCR was conducted to determine the quantity of parasites in the blood and revealed < 1 to 75 T. cruzi/ml in untreated (means 25.83 +/- 26.32) and < 1 to 36 T. cruzi/ml in treated (means 6.45 +/- 9.28) Chagas' disease patients. The difference between the means was not statistically significant. These findings reveal a need for precise definition of the role of treatment of chronic Chagas'disease patients with nitrofuran and nitroimidazole compounds.


Assuntos
Humanos , Masculino , Doença de Chagas/tratamento farmacológico , Nifurtimox/uso terapêutico , Nitroimidazóis/uso terapêutico , Tripanossomicidas/uso terapêutico , Trypanosoma cruzi/isolamento & purificação , Doença de Chagas/sangue , Doença Crônica , Primers do DNA , Hibridização Genética , Reação em Cadeia da Polimerase/métodos , Resultado do Tratamento , Trypanosoma cruzi/genética
15.
Mem. Inst. Oswaldo Cruz ; 95(supl.1): 123-31, 2000.
Artigo em Inglês | LILACS | ID: lil-274871

RESUMO

The development of biotechnology in the last three decades has generated the feeling that the newest scientific achievements will deliver high standard quality of life through abundance of food and means for successfully combating diseases. Where the new biotechnologies give access to genetic information, there is a common belief that physiological and pathological processes result from subtle modifications of gene expression. Trustfully, modern genetics has produced genetic maps, physical maps and complete nucleotide sequences from 141 viruses, 51 organelles, two eubacteria, one archeon and one eukaryote (Saccharomices cerevisiae). In addition, during the Centennial Commemoration of the Oswaldo Cruz Institute the nearly complete human genome map was proudly announced, whereas the latest Brazilian key stone contribution to science was the publication of the Shillela fastidiosa genomic sequence highlythed on a Nature cover issue. There exists a belief among the populace that further scientific accomplishments will rapidly lead to new drugs and methodological approaches to cure genetic diseases and other incurable ailments. Yet, much evidence has been accumulated, showing that a large information gap exists between the knowledge of genome sequence and our knowledge of genome function. Now that many genome maps are available, people wish to know what are we going to do with them. Certainly, all these scientific accomplishments will shed light on many more secrets of life. Nevertheless, parsimony in the weekly announcements of promising scientific achievements is necessary. We also need many more creative experimental biologists to discover new, as yet un-envisaged biotechnological approaches, and the basic resource needed for carrying out mile stone research necessary for leading us to that "promised land" often proclaimed by the mass media


Assuntos
Humanos , Animais , Biotecnologia/tendências , Genoma de Protozoário , Doenças Parasitárias/genética , Pesquisa/tendências , Mapeamento Cromossômico , Genoma , Interações Hospedeiro-Parasita/genética
16.
Mem. Inst. Oswaldo Cruz ; 94(suppl.1): 249-52, Sept. 1999. ilus
Artigo em Inglês | LILACS | ID: lil-245631

RESUMO

Integration of kDNA sequences within the genome of the host cell shown by PCR amplification with primers to the conserved Trypanosoma cruzi kDNA minicircle sequence was confirmed by Southern hybridization with specific probes. The cells containing the integrated kDNA sequences were then perpetuated as transfected macrophage subclonal lines. The kDNA transfected macrophages expressed membrane antigens that were recognized by antibodies in a panel of sera from ten patients with chronic Chagas disease. These antigens barely expressed in the membrane of uninfected, control macrophage clonal lines were recognized neither by factors in the control, non-chagasic subjects nor in the chagasic sera. This finding suggests the presence of an autoimmune antibody in the chagasic sera that recognizes auto-antigens in the membrane of T. cruzi kDNA transfected macrophage subclonal lines.


Assuntos
Humanos , Animais , Autoimunidade , Doença de Chagas/imunologia , DNA de Cinetoplasto , Trypanosoma cruzi , Sequência de Bases , Doença de Chagas/sangue , Genoma , Macrófagos , Trypanosoma cruzi/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA