Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
PLoS Pathog ; 14(4): e1006944, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29672607

RESUMO

Despite extensive genetic diversity of HIV-1 in chronic infection, a single or few maternal virus variants become the founders of an infant's infection. These transmitted/founder (T/F) variants are of particular interest, as a maternal or infant HIV vaccine should raise envelope (Env) specific IgG responses capable of blocking this group of viruses. However, the maternal or infant factors that contribute to selection of infant T/F viruses are not well understood. In this study, we amplified HIV-1 env genes by single genome amplification from 16 mother-infant transmitting pairs from the U.S. pre-antiretroviral era Women Infant Transmission Study (WITS). Infant T/F and representative maternal non-transmitted Env variants from plasma were identified and used to generate pseudoviruses for paired maternal plasma neutralization sensitivity analysis. Eighteen out of 21 (85%) infant T/F Env pseudoviruses were neutralization resistant to paired maternal plasma. Yet, all infant T/F viruses were neutralization sensitive to a panel of HIV-1 broadly neutralizing antibodies and variably sensitive to heterologous plasma neutralizing antibodies. Also, these infant T/F pseudoviruses were overall more neutralization resistant to paired maternal plasma in comparison to pseudoviruses from maternal non-transmitted variants (p = 0.012). Altogether, our findings suggest that autologous neutralization of circulating viruses by maternal plasma antibodies select for neutralization-resistant viruses that initiate peripartum transmission, raising the speculation that enhancement of this response at the end of pregnancy could further reduce infant HIV-1 infection risk.


Assuntos
Anticorpos Neutralizantes/imunologia , Infecções por HIV/transmissão , HIV-1/genética , Transmissão Vertical de Doenças Infecciosas , Plasma/metabolismo , Complicações Infecciosas na Gravidez/etiologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Feminino , Variação Genética , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Lactente , Testes de Neutralização , Período Periparto , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética
2.
J Virol ; 90(10): 4951-4965, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26937027

RESUMO

UNLABELLED: Maternal vaccination to induce anti-HIV immune factors in breast milk is a potential intervention to prevent postnatal HIV-1 mother-to-child transmission (MTCT). We previously demonstrated that immunization of lactating rhesus monkeys with a modified vaccinia Ankara (MVA) prime/intramuscular (i.m.) protein boost regimen induced functional IgG responses in milk, while MVA prime/intranasal (i.n.) boost induced robust milk Env-specific IgA responses. Yet, recent studies have suggested that prevention of postnatal MTCT may require both Env-specific IgA and functional IgG responses in milk. Thus, to investigate whether both responses could be elicited by a combined systemic/mucosal immunization strategy, animals previously immunized with the MVA prime/i.n. boost regimen received an i.n./i.m. combined C.1086 gp120 boost. Remarkably, high-magnitude Env-specific IgA responses were observed in milk, surpassing those in plasma. Furthermore, 29% of vaccine-elicited Env-specific B cells isolated from breast milk were IgA isotype, in stark contrast to the overwhelming predominance of IgG isotype Env-specific B cells in breast milk of chronically HIV-infected women. A clonal relationship was identified between Env-specific blood and breast milk B cells, suggesting trafficking of that cell population between the two compartments. Furthermore, IgA and IgG monoclonal antibodies isolated from Env-specific breast milk B cells demonstrated diverse Env epitope specificities and multiple effector functions, including tier 1 neutralization, antibody-dependent cellular cytotoxicity (ADCC), infected cell binding, and inhibition of viral attachment to epithelial cells. Thus, maternal i.n./i.m. combined immunization is a novel strategy to enhance protective Env-specific IgA in milk, which is subsequently transferred to the infant via breastfeeding. IMPORTANCE: Efforts to increase the availability of antiretroviral therapy to pregnant and breastfeeding women in resource-limited areas have proven remarkably successful at reducing HIV vertical transmission rates. However, more than 200,000 children are infected annually due to failures in therapy implementation, monitoring, and adherence, nearly half by postnatal HIV exposure via maternal breast milk. Intriguingly, in the absence of antiretroviral therapy, only 10% of breastfed infants born to HIV-infected mothers acquire the virus, suggesting the existence of naturally protective immune factors in milk. Enhancement of these protective immune factors through maternal vaccination will be a critical strategy to reduce the global pediatric AIDS epidemic. We have previously demonstrated that a high magnitude of HIV Env-specific IgA in milk correlates with reduced risk of infant HIV acquisition. In this study, we describe a novel HIV vaccine regimen that induces potent IgA responses in milk and therefore could potentially protect against breast milk HIV MTCT.


Assuntos
Vacinas contra a AIDS/imunologia , Linfócitos B/imunologia , Anticorpos Anti-HIV/análise , HIV-1/imunologia , Imunoglobulina A/análise , Lactação , Leite/imunologia , Vacinas contra a AIDS/administração & dosagem , Animais , Anticorpos Neutralizantes/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Feminino , Anticorpos Anti-HIV/sangue , Proteína gp120 do Envelope de HIV/administração & dosagem , Proteína gp120 do Envelope de HIV/imunologia , Humanos , Imunidade Materno-Adquirida , Imunidade nas Mucosas , Imunização Secundária , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Imunoglobulina G/análise , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lactente , Macaca mulatta , Gravidez
3.
J Virol ; 89(21): 10868-78, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292320

RESUMO

UNLABELLED: Despite the nutritional and health benefits of breast milk, breast milk can serve as a vector for mother-to-child HIV transmission. Most HIV-infected infants acquire HIV through breastfeeding. Paradoxically, most infants breastfed by HIV-positive women do not become infected. This is potentially attributed to anti-HIV factors in breast milk. Breast milk of HIV-negative women can inhibit HIV infection. However, the HIV-inhibitory activity of breast milk from HIV-positive mothers has not been evaluated. In addition, while significant differences in breast milk composition between transmitting and nontransmitting HIV-positive mothers have been correlated with transmission risk, the HIV-inhibitory activity of their breast milk has not been compared. This knowledge may significantly impact the design of prevention approaches in resource-limited settings that do not deny infants of HIV-positive women the health benefits of breast milk. Here, we utilized bone marrow/liver/thymus humanized mice to evaluate the in vivo HIV-inhibitory activity of breast milk obtained from HIV-positive transmitting and nontransmitting mothers. We also assessed the species specificity and biochemical characteristics of milk's in vivo HIV-inhibitory activity and its ability to inhibit other modes of HIV infection. Our results demonstrate that breast milk of HIV-positive mothers has potent HIV-inhibitory activity and indicate that breast milk can prevent multiple routes of infection. Most importantly, this activity is unique to human milk. Our results also suggest multiple factors in breast milk may contribute to its HIV-inhibitory activity. Collectively, our results support current recommendations that HIV-positive mothers in resource-limited settings exclusively breastfeed in combination with antiretroviral therapy. IMPORTANCE: Approximately 240,000 children become infected with HIV annually, the majority via breastfeeding. Despite daily exposure to virus in breast milk, most infants breastfed by HIV-positive women do not acquire HIV. The low risk of breastfeeding-associated HIV transmission is likely due to antiviral factors in breast milk. It is well documented that breast milk of HIV-negative women can inhibit HIV infection. Here, we demonstrate, for the first time, that breast milk of HIV-positive mothers (nontransmitters and transmitters) inhibits HIV transmission. We also demonstrate that breast milk can prevent multiple routes of HIV acquisition and that this activity is unique to human milk. Collectively, our results support current guidelines which recommend that HIV-positive women in resource-limited settings exclusively breastfeed in combination with infant or maternal antiretroviral therapy.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/transmissão , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Leite Humano/química , Animais , Western Blotting , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Transmissão Vertical de Doenças Infecciosas/estatística & dados numéricos , Macaca mulatta , Camundongos , Leite Humano/imunologia , Especificidade da Espécie , Zâmbia
4.
PLoS Pathog ; 10(12): e1004538, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25473946

RESUMO

In contrast to the ability of long-lived CD8(+) memory T cells to mediate protection against systemic viral infections, the relationship between CD4(+) T cell memory and acquired resistance against infectious pathogens remains poorly defined. This is especially true for T helper 1 (Th1) concomitant immunity, in which protection against reinfection coincides with a persisting primary infection. In these situations, pre-existing effector CD4 T cells generated by ongoing chronic infection, not memory cells, may be essential for protection against reinfection. We present a systematic study of the tissue homing properties, functionality, and life span of subsets of memory and effector CD4 T cells activated in the setting of chronic Leishmania major infection in resistant C57Bl/6 mice. We found that pre-existing, CD44(+)CD62L(-)T-bet(+)Ly6C+ effector (T(EFF)) cells that are short-lived in the absence of infection and are not derived from memory cells reactivated by secondary challenge, mediate concomitant immunity. Upon adoptive transfer and challenge, non-dividing Ly6C(+) T(EFF) cells preferentially homed to the skin, released IFN-γ, and conferred protection as compared to CD44(+)CD62L(-)Ly6C(-) effector memory or CD44(+)CD62L(+)Ly6C(-) central memory cells. During chronic infection, Ly6C(+) T(EFF) cells were maintained at high frequencies via reactivation of T(CM) and the T(EFF) themselves. The lack of effective vaccines for many chronic diseases may be because protection against infectious challenge requires the maintenance of pre-existing T(EFF) cells, and is therefore not amenable to conventional, memory inducing, vaccination strategies.


Assuntos
Antígenos Ly/imunologia , Linfócitos T CD4-Positivos/imunologia , Memória Imunológica , Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Animais , Antígenos Ly/genética , Doença Crônica , Feminino , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Interferon gama/genética , Interferon gama/imunologia , Selectina L/genética , Selectina L/imunologia , Leishmania major/genética , Leishmaniose Cutânea/genética , Camundongos
5.
Proc Natl Acad Sci U S A ; 110(45): 18220-5, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24145401

RESUMO

Achieving an AIDS-free generation will require elimination of postnatal transmission of HIV-1 while maintaining the nutritional and immunologic benefits of breastfeeding for infants in developing regions. Maternal/infant antiretroviral prophylaxis can reduce postnatal HIV-1 transmission, yet toxicities and the development of drug-resistant viral strains may limit the effectiveness of this strategy. Interestingly, in the absence of antiretroviral prophylaxis, greater than 90% of infants exposed to HIV-1 via breastfeeding remain uninfected, despite daily mucosal exposure to the virus for up to 2 y. Moreover, milk of uninfected women inherently neutralizes HIV-1 and prevents virus transmission in animal models, yet the factor(s) responsible for this anti-HIV activity is not well-defined. In this report, we identify a primary HIV-1-neutralizing protein in breast milk, Tenascin-C (TNC). TNC is an extracellular matrix protein important in fetal development and wound healing, yet its antimicrobial properties have not previously been established. Purified TNC captured and neutralized multiclade chronic and transmitted/founder HIV-1 variants, and depletion of TNC abolished the HIV-1-neutralizing activity of milk. TNC bound the HIV-1 Envelope protein at a site that is induced upon engagement of its primary receptor, CD4, and is blocked by V3 loop- (19B and F39F) and chemokine coreceptor binding site-directed (17B) monoclonal antibodies. Our results demonstrate the ability of an innate mucosal host protein found in milk to neutralize HIV-1 via binding to the chemokine coreceptor site, potentially explaining why the majority of HIV-1-exposed breastfed infants are protected against mucosal HIV-1 transmission.


Assuntos
Síndrome da Imunodeficiência Adquirida/transmissão , HIV-1/efeitos dos fármacos , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Leite Humano/química , Tenascina/farmacologia , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Western Blotting , Linhagem Celular , Cromatografia por Troca Iônica , Relação Dose-Resposta a Droga , Feminino , Humanos , Imunoprecipitação , Concentração Inibidora 50 , Espectrometria de Massas , Tenascina/metabolismo , Proteínas do Envelope Viral/metabolismo
6.
J Immunol ; 189(10): 4832-41, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23045616

RESUMO

Numerous experimental Leishmania vaccines have been developed to prevent the visceral and cutaneous forms of Leishmaniasis, which occur after exposure to the bite of an infected sand fly, yet only one is under evaluation in humans. KSAC and L110f, recombinant Leishmania polyproteins delivered in a stable emulsion (SE) with the TLR4 agonists monophosphoryl lipid A or glucopyranosyl lipid A (GLA) have shown protection in animal models. KSAC+GLA-SE protected against cutaneous disease following sand fly transmission of Leishmania major in susceptible BALB/c mice. Similar polyprotein adjuvant combinations are the vaccine candidates most likely to see clinical evaluation. We assessed immunity generated by KSAC or L110f vaccination with GLA-SE following challenge with L. major by needle or infected sand fly bite in resistant C57BL/6 mice. Polyprotein-vaccinated mice had a 60-fold increase in CD4(+)IFN-γ(+) T cell numbers versus control animals at 2 wk post-needle inoculation of L. major, and this correlated with a 100-fold reduction in parasite load. Immunity did not, however, reach levels observed in mice with a healed primary infection. Following challenge by infected sand fly bite, polyprotein-vaccinated animals had comparable parasite loads, greater numbers of neutrophils at the challenge site, and reduced CD4(+)IFN-γ(+)/IL-17(+) ratios versus nonvaccinated controls. In contrast, healed animals had significantly reduced parasite loads and higher CD4(+)IFN-γ(+)/IL-17(+) ratios. These observations demonstrate that vaccine-induced protection against needle challenge does not necessarily translate to protection following challenge by infected sand fly bite.


Assuntos
Adjuvantes Imunológicos/farmacologia , Leishmania major/imunologia , Vacinas contra Leishmaniose/farmacologia , Leishmaniose Cutânea/prevenção & controle , Lipídeo A/análogos & derivados , Proteínas de Protozoários/farmacologia , Psychodidae , Animais , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Emulsões , Interferon gama/imunologia , Interleucina-17/imunologia , Leishmania major/genética , Vacinas contra Leishmaniose/genética , Vacinas contra Leishmaniose/imunologia , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/transmissão , Lipídeo A/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia
7.
Mucosal Immunol ; 12(4): 1004-1012, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30976088

RESUMO

Interactions between innate antiviral factors at mucosal surfaces and HIV-1 virions contribute to the natural inefficiency of HIV-1 transmission and are a platform to inform the development of vaccine and nonvaccine strategies to block mucosal HIV-1 transmission. Tenascin-C (TNC) is a large, hexameric extracellular matrix glycoprotein identified in breast milk and genital fluids that broadly neutralizes HIV-1 via interaction with the HIV-1 Envelope (Env) variable 3 (V3) loop. In this report, we characterize the specific determinants of the interaction between TNC and the HIV-1 Env. We observed that TNC binding and neutralization of HIV-1 is dependent on the TNC fibrinogen-like globe (fbg) and fibronectin-type III (fn) domains, oligomerization, and its newly-mapped glycan structure. Moreover, we observed that TNC-mediated neutralization is also dependent on Env V3 residues 321/322 and 326/327, which surround the IGDIR motif of the V3 loop, as well the N332 glycan, which is critical to the broadly neutralizing activity of glycan-dependent V3-specific antibodies such as PGT128. Our results demonstrate a striking parallel between innate and adaptive immune mechanisms of broad HIV neutralization and provide further insight into the host protein-virus interactions responsible for the natural inefficiency of mucosal HIV-1 transmission.


Assuntos
HIV-1/metabolismo , Tenascina/química , Tenascina/metabolismo , Produtos do Gene env do Vírus da Imunodeficiência Humana/química , Produtos do Gene env do Vírus da Imunodeficiência Humana/metabolismo , Sequência de Aminoácidos , Aminoácidos , Mapeamento de Epitopos , Epitopos/química , Epitopos/imunologia , Glicosilação , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/imunologia , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/imunologia , Humanos , Modelos Moleculares , Testes de Neutralização , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia
9.
JCI Insight ; 2(13)2017 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-28679960

RESUMO

Human cytomegalovirus (HCMV) is the most common congenital infection and a known cause of microcephaly, sensorineural hearing loss, and cognitive impairment among newborns worldwide. Natural maternal HCMV immunity reduces the incidence of congenital infection, but does not prevent the disease altogether. We employed a nonhuman primate model of congenital CMV infection to investigate the ability of preexisting antibodies to protect against placental CMV transmission in the setting of primary maternal infection and subsequent viremia, which is required for placental virus exposure. Pregnant, CD4+ T cell-depleted, rhesus CMV-seronegative (RhCMV-seronegative) rhesus monkeys were treated with either standardly produced hyperimmune globulin (HIG) from RhCMV-seropositive macaques or dose-optimized, potently RhCMV-neutralizing HIG prior to intravenous challenge with an RhCMV mixture. HIG passive infusion provided complete protection against fetal loss in both groups. The dose-optimized, RhCMV-neutralizing HIG additionally inhibited placental transmission of RhCMV and reduced viral replication and diversity. Our findings suggest that the presence of durable and potently neutralizing antibodies at the time of primary infection can prevent transmission of systemically replicating maternal RhCMV to the developing fetus, and therefore should be a primary target of vaccines to eliminate this neonatal infection.

10.
PLoS One ; 11(5): e0155261, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27182834

RESUMO

Tenascin-C (TNC) is a newly identified innate HIV-1-neutralizing protein present in breast milk, yet its presence and potential HIV-inhibitory function in other mucosal fluids is unknown. In this study, we identified TNC as a component of semen and cervical fluid of HIV-1-infected and uninfected individuals, although it is present at a significantly lower concentration and frequency compared to that of colostrum and mature breast milk, potentially due to genital fluid protease degradation. However, TNC was able to neutralize HIV-1 after exposure to low pH, suggesting that TNC could be active at low pH in the vaginal compartment. As mucosal fluids are complex and contain a number of proteins known to interact with the HIV-1 envelope, we further studied the relationship between the concentration of TNC and neutralizing activity in breast milk. The amount of TNC correlated only weakly with the overall innate HIV-1-neutralizing activity of breast milk of uninfected women and negatively correlated with neutralizing activity in milk of HIV-1 infected women, indicating that the amount of TNC in mucosal fluids is not adequate to impede HIV-1 transmission. Moreover, the presence of polyclonal IgG from milk of HIV-1 infected women, but not other HIV-1 envelope-binding milk proteins or monoclonal antibodies, blocked the neutralizing activity of TNC. Finally, as exogenous administration of TNC would be necessary for it to mediate measurable HIV-1 neutralizing activity in mucosal compartments, we established that recombinantly produced TNC has neutralizing activity against transmitted/founder HIV-1 strains that mimic that of purified TNC. Thus, we conclude that endogenous TNC concentration in mucosal fluids is likely inadequate to block HIV-1 transmission to uninfected individuals.


Assuntos
Líquido Extracelular/imunologia , Genitália , Infecções por HIV/imunologia , HIV-1/imunologia , Proteínas do Leite/imunologia , Leite Humano/imunologia , Tenascina/imunologia , Anticorpos Neutralizantes/imunologia , Colo do Útero/imunologia , Feminino , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Imunidade Inata , Imunoglobulina G/imunologia , Masculino , Proteínas do Leite/farmacologia , Mucosa/imunologia , Mucosa/metabolismo , Testes de Neutralização , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes , Sêmen/imunologia , Tenascina/farmacologia
11.
PLoS Negl Trop Dis ; 5(8): e1288, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21886852

RESUMO

To identify parameters of Leishmania infection within a population of infected sand flies that reliably predict subsequent transmission to the mammalian host, we sampled groups of infected flies and compared infection intensity and degree of metacyclogenesis with the frequency of transmission. The percentage of parasites within the midgut that were metacyclic promastigotes had the highest correlation with the frequency of transmission. Meta-analysis of multiple transmission experiments allowed us to establish a percent-metacyclic "cutoff" value that predicted transmission competence. Sand fly infections initiated with variable doses of parasites resulted in correspondingly altered percentages of metacyclic promastigotes, resulting in altered transmission frequency and disease severity. Lastly, alteration of sand fly oviposition status and environmental conditions at the time of transmission also influenced transmission frequency. These observations have implications for transmission of Leishmania by the sand fly vector in both the laboratory and in nature, including how the number of organisms acquired by the sand fly from an infection reservoir may influence the clinical outcome of infection following transmission by bite.


Assuntos
Vetores de Doenças , Leishmania major/isolamento & purificação , Leishmaniose/transmissão , Psychodidae/parasitologia , Animais , Feminino , Trato Gastrointestinal/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
12.
Res Microbiol ; 160(3): 179-86, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19284970

RESUMO

The tricarballylate utilization locus (tcuRABC) of Salmonella enterica serovar Typhimurium is comprised of a 3-gene operon (tcuABC) that encodes functions that allow this bacterium to use tricarballylate as a source of carbon and energy, and the tcuR gene, which encodes a putative LysR-type transcriptional regulator. In our studies, transcription of the tcuABC operon peaked at mid-log phase, and declined moderately during stationary phase. This pattern was not due to a change in the amount of TcuR in the cell, as tcuR expression did not change under the conditions tested, and TcuR did not control tcuR expression. Tricarballylate was the co-inducer. tcuABC expression was negatively affected by the cAMP receptor protein (Crp). Expression of tcuABC was one order of magnitude higher in a crp mutant strain than in the crp(+) strain; derepression of tcuABC expression was also observed in a strain lacking adenylate cyclase (Cya). At present, it is unclear whether the effect of Crp is direct or indirect. Studies with molecular mimics of tricarballylate showed that the co-inducer site restricts binding of structural mimics that contain a hydroxyl group. Two classes of TcuR constitutive variants were isolated. Class I variants responded to tricarballylate, while Class II did not.


Assuntos
Regulação Bacteriana da Expressão Gênica , Redes e Vias Metabólicas/genética , Óperon , Salmonella typhimurium/fisiologia , Ácidos Tricarboxílicos/metabolismo , Adenilil Ciclases/genética , Adenilil Ciclases/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Proteína Receptora de AMP Cíclico/genética , Proteína Receptora de AMP Cíclico/fisiologia , Deleção de Genes , Perfilação da Expressão Gênica , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA