Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Elife ; 102021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34783654

RESUMO

Adiponectin-mediated pathways contribute to mammalian homeostasis; however, little is known about adiponectin and adiponectin receptor signaling in arthropods. In this study, we demonstrate that Ixodes scapularis ticks have an adiponectin receptor-like protein (ISARL) but lack adiponectin, suggesting activation by alternative pathways. ISARL expression is significantly upregulated in the tick gut after Borrelia burgdorferi infection, suggesting that ISARL signaling may be co-opted by the Lyme disease agent. Consistent with this, RNA interference (RNAi)-mediated silencing of ISARL significantly reduced the B. burgdorferi burden in the tick. RNA-seq-based transcriptomics and RNAi assays demonstrate that ISARL-mediated phospholipid metabolism by phosphatidylserine synthase I is associated with B. burgdorferi survival. Furthermore, the tick complement C1q-like protein 3 interacts with ISARL, and B. burgdorferi facilitates this process. This study identifies a new tick metabolic pathway that is connected to the life cycle of the Lyme disease spirochete.


Many countries around the world are seeing an increase in the number of patients diagnosed with Lyme disease, with often serious joint, heart, and neurologic complications. This illness is caused by species of 'spirochete' bacteria that live and multiply inside black-legged ticks, and get injected into mammals upon a bite. Ticks are not simply 'syringes' however, and a complex relationship is established between spirochetes and their host. This is particularly true since Lyme disease-causing bacteria such as Borrelia burgdorferi rely on ticks to obtain energy and nutrients. Tang, Cao et al. delved into these complex interactions by focusing on the molecular cascades (or pathways) involving adiponectin, a hormone essential for regulating sugar levels and processing fats. Analyses of gene and protein databases highlighted that ticks carry a receptor-like protein for adiponectin but not the hormone itself, suggesting that an alternative pathway is at play. This may involve B. burgdorferi, which gets its fats and sugars from its host. And indeed, experiments showed that ticks produced more of the adiponectin receptor-like protein when they carried B. burgdorferi; conversely, silencing the receptor reduced the number of surviving spirochetes inside the tick. Further exploration showed that the receptor mediates molecular cascades that help to process fat molecules; these are associated with spirochete survival. In addition, the receptor-like protein was activated by C1QL3, a 'complement 1q domain-contained' molecule which might be part of the tick energy-making or immune systems. Larger quantities of C1QL3 were found in ticks upon B. burgdorferi infection, suggesting that the spirochete facilitates an interaction that boosts activity of the adiponectin receptor-like protein. Overall, the work by Tang and Cao et al. revealed a new pathway which B. burgdorferi takes advantage of to infect their host and multiply. Targeting this molecular cascade could help to interfere with the life cycle of the spirochete, as well as fight Lyme disease and other insect-borne conditions.


Assuntos
Borrelia burgdorferi/metabolismo , Ixodes/metabolismo , Ixodes/microbiologia , Receptores de Adiponectina/metabolismo , Animais , Proteínas de Artrópodes/metabolismo , Vetores Artrópodes/metabolismo , Vetores Artrópodes/microbiologia , Doença de Lyme/metabolismo , Doença de Lyme/microbiologia , Fosfolipídeos/metabolismo , Interferência de RNA , Receptores de Adiponectina/genética , Transcriptoma
3.
Commun Biol ; 3(1): 556, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-33033362

RESUMO

Macrophage scavenger receptor 1 (MSR1) mediates the endocytosis of modified low-density lipoproteins and plays an important antiviral role. However, the molecular mechanism underlying MSR1 antiviral actions remains elusive. We report that MSR1 activates autophagy to restrict infection of Chikungunya virus (CHIKV), an arthritogenic alphavirus that causes acute and chronic crippling arthralgia. Msr1 expression was rapidly upregulated after CHIKV infection in mice. Msr1 knockout mice had elevated viral loads and increased susceptibility to CHIKV arthritis along with a normal type I IFN response. Induction of LC3 lipidation by CHIKV, a marker of autophagy, was reduced in Msr1-/- cells. Mechanistically, MSR1 interacted with ATG12 through its cytoplasmic tail and this interaction was enhanced by CHIKV nsP1 protein. MSR1 repressed CHIKV replication through ATG5-ATG12-ATG16L1 and this was dependent on the FIP200-and-WIPI2-binding domain, but not the WD40 domain of ATG16L1. Our results elucidate an antiviral role for MSR1 involving the autophagic function of ATG5-ATG12-ATG16L1.


Assuntos
Autofagia , Febre de Chikungunya/imunologia , Vírus Chikungunya/metabolismo , Receptores Depuradores Classe A/fisiologia , Animais , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Edição de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Depuradores Classe A/metabolismo
4.
Viruses ; 12(5)2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32397176

RESUMO

Mosquito-borne Zika virus (ZIKV) can cause congenital microcephaly and Guillain-Barré syndrome, among other symptoms. Specific treatments and vaccines for ZIKV are not currently available. To further understand the host factors that support ZIKV replication, we used mass spectrometry to characterize mammalian proteins that associate with the ZIKV NS1 protein and identified the TRiC/CCT complex as an interacting partner. Furthermore, the suppression of CCT2, one of the critical components of the TRiC/CCT complex, inhibited ZIKV replication in both mammalian cells and mosquitoes. These results highlight an important role for the TRiC/CCT complex in ZIKV infection, suggesting that the TRiC/CCT complex may be a promising therapeutic target.


Assuntos
Aedes/virologia , Chaperonina com TCP-1/metabolismo , Proteínas de Insetos/metabolismo , Mosquitos Vetores/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Infecção por Zika virus/virologia , Zika virus/fisiologia , Aedes/genética , Aedes/metabolismo , Animais , Chaperonina com TCP-1/genética , Feminino , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Insetos/genética , Mosquitos Vetores/genética , Mosquitos Vetores/metabolismo , Ligação Proteica , Proteínas não Estruturais Virais/genética , Zika virus/genética , Infecção por Zika virus/genética , Infecção por Zika virus/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-31307983

RESUMO

The mosquito-borne chikungunya virus (CHIKV) causes acute pain and joint inflammation, and in recent years the virus has caused large epidemics in previously CHIKV-free geographic areas. To advance the understanding of host factors that antagonize CHIKV, we show that synthetic agonist of liver X receptor (LXR-623) inhibits CHIKV replication by upregulating the cholesterol exporter ABCA1 and that endogenous and pharmacological activation of interferon signaling pathway partners with LXR-623 to generate a superior antiviral state.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/genética , Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Indazóis/farmacologia , Receptores X do Fígado/genética , Transportador 1 de Cassete de Ligação de ATP/antagonistas & inibidores , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/metabolismo , Colesterol/metabolismo , Colesterol/farmacologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica , Humanos , Interferons/genética , Interferons/metabolismo , Receptores X do Fígado/antagonistas & inibidores , Receptores X do Fígado/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Replicação Viral/efeitos dos fármacos
6.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30971475

RESUMO

Saliva from the mosquito vector of flaviviruses is capable of changing the local immune environment, leading to an increase in flavivirus-susceptible cells at the infected bite site. In addition, an antibody response to specific salivary gland (SG) components changes the pathogenesis of flaviviruses in human populations. To investigate whether antigenic SG proteins are capable of enhancing infection with Zika virus (ZIKV), a reemerging flavivirus primarily transmitted by the Aedes aegypti mosquito, we screened for antigenic SG proteins using a yeast display library and demonstrate that a previously undescribed SG protein we term neutrophil stimulating factor 1 (NeSt1) activates primary mouse neutrophils ex vivo Passive immunization against NeSt1 decreases pro-interleukin-1ß and CXCL2 expression, prevents macrophages from infiltrating the bite site, protects susceptible IFNAR-/- IFNGR-/- (AG129) mice from early ZIKV replication, and ameliorates virus-induced pathogenesis. These findings indicate that NeSt1 stimulates neutrophils at the mosquito bite site to change the immune microenvironment, allowing a higher level of early viral replication and enhancing ZIKV pathogenesis.IMPORTANCE When a Zika virus-infected mosquito bites a person, mosquito saliva is injected into the skin along with the virus. Molecules in this saliva can make virus infection more severe by changing the immune system to make the skin a better place for the virus to replicate. We identified a molecule that activates immune cells, called neutrophils, to recruit other immune cells, called macrophages, that the virus can infect. We named this molecule neutrophil-stimulating factor 1 (NeSt1). When we used antibodies to block NeSt1 in mice and then allowed Zika virus-infected mosquitoes to feed on these mice, they survived much better than mice that do not have antibodies against NeSt1. These findings give us more information about how mosquito saliva enhances virus infection, and it is possible that a vaccine against NeSt1 might protect people against severe Zika virus infection.


Assuntos
Aedes/virologia , Neutrófilos/metabolismo , Neutrófilos/virologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Aedes/imunologia , Animais , Arbovírus , Quimiocina CCL2 , Quimiocina CXCL2/metabolismo , Modelos Animais de Doenças , Feminino , Imunidade , Interleucina-1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mosquitos Vetores/virologia , Precursores de Proteínas/metabolismo , Células RAW 264.7 , Saliva/virologia , Glândulas Salivares/virologia , Replicação Viral , Zika virus/patogenicidade , Infecção por Zika virus/virologia
7.
iScience ; 13: 339-350, 2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-30884311

RESUMO

The TAM receptor, Axl, has been implicated as a candidate entry receptor for Zika virus (ZIKV) infection but has been shown as inessential for virus infection in mice. To probe the role of Axl in murine ZIKV infection, we developed a mouse model lacking the Axl receptor and the interferon alpha/beta receptor (Ifnar-/-Axl-/-), conferring susceptibility to ZIKV. This model validated that Axl is not required for murine ZIKV infection and that mice lacking Axl are resistant to ZIKV pathogenesis. This resistance correlates to lower pro-interleukin-1ß production and less apoptosis in microglia of ZIKV-infected mice. This apoptosis occurs through both intrinsic (caspase 9) and extrinsic (caspase 8) manners, and is age dependent, as younger Axl-deficient mice are susceptible to ZIKV pathogenesis. These findings suggest that Axl plays an important role in pathogenesis in the brain during ZIKV infection and indicates a potential role for Axl inhibitors as therapeutics during viral infection.

8.
Antiviral Res ; 161: 163-168, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30521835

RESUMO

Arboviruses represent a group of pathogens that can spread efficiently throughout human populations by hematophagous arthropod vectors. The mosquito-borne (re)emerging Chikungunya and Dengue viruses belong to the alphavirus and flavivirus genus, respectively, with no approved therapeutics or safe vaccines for humans. Transmitted by the same vector Aedes spp., these viruses cause significant morbidity and mortality in endemic areas. Due to the increasing likelihood of co-circulation and co-infection with viruses, we aimed to identify a pharmacologically targetable host factor that can inhibit multiple viruses and show that a potent antagonist of prolyl tRNA synthetase (halofuginone) suppresses both Chikungunya and Dengue viruses. Host tRNA synthetase inhibition may signify an additional approach to combat present and future epidemic pathogens.


Assuntos
Aedes/enzimologia , Aminoacil-tRNA Sintetases/antagonistas & inibidores , Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Piperidinas/farmacologia , Quinazolinonas/farmacologia , Aedes/virologia , Animais , Células Cultivadas , Febre de Chikungunya/virologia , Dengue/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Prepúcio do Pênis/citologia , Interações entre Hospedeiro e Microrganismos , Humanos , Proteínas de Insetos/antagonistas & inibidores , Masculino , Mosquitos Vetores , Receptor de Interferon alfa e beta/agonistas
9.
Open Forum Infect Dis ; 5(12): ofy315, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30568983

RESUMO

Chikungunya virus (CHIKV), an alphavirus spread by Aedes spp. mosquitos, causes severe inflammation and joint pain, progressing to a chronic arthralgic state in a subset of patients. Due to recent global epidemics of CHIKV and the potential for related viruses to cause outbreaks, multiple approaches to combat these pathogens are of interest. We report that SR9009, a synthetic agonist of nuclear receptors Rev-erb α/ß, inhibits replication of multiple alphaviruses (CHIKV and O'nyong'nyong virus) mainly by suppressing structural protein synthesis, although viral RNA accumulation is relatively unimpeded. Furthermore, SR9009 reduces the inflammatory response in cultured murine macrophages exposed to alphavirus-infected cells.

10.
J Infect Dis ; 215(11): 1720-1724, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28472297

RESUMO

Zika virus (ZIKV) can be transmitted by mosquito bite or sexual contact. Using mice that lack the type I interferon receptor, we examined sexual transmission of ZIKV. Electron microscopy analyses showed association of virions with developing sperm within testes as well as with mature sperm within epididymis. When ZIKV-infected male mice were mated with naive female mice, the weight of fetuses at embryonic day 18.5 was significantly reduced compared with the control group. Additionally, we found ocular deformities in a minority of the fetuses. These results suggest that ZIKV causes fetal abnormalities after female mating with an infected male.


Assuntos
Retardo do Crescimento Fetal/virologia , Complicações Infecciosas na Gravidez/virologia , Doenças Virais Sexualmente Transmissíveis/transmissão , Infecção por Zika virus/transmissão , Zika virus , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Gravidez , Doenças Virais Sexualmente Transmissíveis/virologia , Infecção por Zika virus/virologia
11.
Cell Rep ; 19(3): 558-568, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423319

RESUMO

Tyro3, Axl, and Mertk (TAM) receptors are candidate entry receptors for infection with the Zika virus (ZIKV), an emerging flavivirus of global public health concern. To investigate the requirement of TAM receptors for ZIKV infection, we used several routes of viral inoculation and compared viral replication in wild-type versus Axl-/-, Mertk-/-, Axl-/-Mertk-/-, and Axl-/-Tyro3-/- mice in various organs. Pregnant and non-pregnant mice treated with interferon-α-receptor (IFNAR)-blocking (MAR1-5A3) antibody and infected subcutaneously with ZIKV showed no reliance on TAMs for infection. In the absence of IFNAR-blocking antibody, adult female mice challenged intravaginally with ZIKV showed no difference in mucosal viral titers. Similarly, in young mice that were infected with ZIKV intracranially or intraperitoneally, ZIKV replication occurred in the absence of TAM receptors, and no differences in cell tropism were observed. These findings indicate that, in mice, TAM receptors are not required for ZIKV entry and infection.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Infecção por Zika virus/metabolismo , Zika virus/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Feto/virologia , Injeções Intraperitoneais , Camundongos , Placenta/virologia , Gravidez , Tropismo , Vagina/virologia , Replicação Viral , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
12.
Sci Adv ; 3(2): e1602899, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28261663

RESUMO

Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that has recently been found to cause fetal infection and neonatal abnormalities, including microcephaly and neurological dysfunction. ZIKV persists in the semen months after the acute viremic phase in humans. To further understand the consequences of ZIKV persistence in males, we infected Ifnar1-/- mice via subcutaneous injection of a pathogenic but nonlethal ZIKV strain. ZIKV replication persists within the testes even after clearance from the blood, with interstitial, testosterone-producing Leydig cells supporting virus replication. We found high levels of viral RNA and antigen within the epididymal lumen, where sperm is stored, and within surrounding epithelial cells. Unexpectedly, at 21 days post-infection, the testes of the ZIKV-infected mice were significantly smaller compared to those of mock-infected mice, indicating progressive testicular atrophy. ZIKV infection caused a reduction in serum testosterone, suggesting that male fertility can be affected. Our findings have important implications for nonvector-borne vertical transmission, as well as long-term potential reproductive deficiencies, in ZIKV-infected males.


Assuntos
RNA Viral/biossíntese , Testículo , Testosterona/sangue , Replicação Viral/fisiologia , Infecção por Zika virus , Zika virus/fisiologia , Animais , Atrofia , Masculino , Camundongos , Camundongos Knockout , RNA Viral/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Testículo/metabolismo , Testículo/virologia , Infecção por Zika virus/sangue , Infecção por Zika virus/genética , Infecção por Zika virus/patologia
13.
JCI Insight ; 1(13)2016 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-27595140

RESUMO

The strong association of Zika virus infection with congenital defects has led to questions of how a flavivirus is capable of crossing the placental barrier to reach the fetal brain. Here, we demonstrate permissive Zika virus infection of primary human placental macrophages, commonly referred to as Hofbauer cells, and placental villous fibroblasts. We also demonstrate Zika virus infection of Hofbauer cells within the context of the tissue ex vivo using term placental villous explants. In addition to amplifying infectious virus within a usually inaccessible area, the putative migratory activities of Hofbauer cells may aid in dissemination of Zika virus to the fetal brain. Understanding the susceptibility of placenta-specific cell types will aid future work around and understanding of Zika virus-associated pregnancy complications.

14.
Cell Host Microbe ; 19(5): 583-7, 2016 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-27173926

RESUMO

Arboviruses have made unexpected reappearances in recent years. Unlike viruses that undergo direct transmission, arboviruses utilize an arthropod vector (e.g., mosquitos, sandflies, and ticks) to spread throughout human populations. Here, we provide a snapshot of mosquito susceptibility to viral infection using flaviviruses, alphaviruses, and bunyaviruses as examples of emerging pathogens of global health relevance.


Assuntos
Alphavirus/genética , Culicidae/genética , Culicidae/virologia , Flavivirus/genética , Insetos Vetores/genética , Insetos Vetores/virologia , Orthobunyavirus/genética , Alphavirus/patogenicidade , Animais , Flavivirus/patogenicidade , Humanos , Orthobunyavirus/patogenicidade
15.
Proc Natl Acad Sci U S A ; 111(52): E5706-15, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512541

RESUMO

An shRNA-mediated screen of the 48 human nuclear receptor genes identified multiple candidates likely to influence the production of human cytomegalovirus in cultured human fibroblasts, including the estrogen-related receptor α (ERRα), an orphan nuclear receptor. The 50-kDa receptor and a 76-kDa variant were induced posttranscriptionally following infection. Genetic and pharmacological suppression of the receptor reduced viral RNA, protein, and DNA accumulation, as well as the yield of infectious progeny. In addition, RNAs encoding multiple metabolic enzymes, including enzymes sponsoring glycolysis (enolase 1, triosephosphate isomerase 1, and hexokinase 2), were reduced when the function of ERRα was inhibited in infected cells. Consistent with the effect on RNAs, a substantial number of metabolites, which are normally induced by infection, were either not increased or were increased to a reduced extent in the absence of normal ERRα activity. We conclude that ERRα is needed for the efficient production of cytomegalovirus progeny, and we propose that the nuclear receptor contributes importantly to the induction of a metabolic environment that supports optimal cytomegalovirus replication.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Receptor alfa de Estrogênio/metabolismo , Replicação Viral/fisiologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Glicólise/genética , Hexoquinase/genética , Hexoquinase/metabolismo , Humanos , Fosfopiruvato Hidratase/genética , Fosfopiruvato Hidratase/metabolismo , Biossíntese de Proteínas/genética , RNA Viral/genética , RNA Viral/metabolismo , Triose-Fosfato Isomerase/genética , Triose-Fosfato Isomerase/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
16.
J Virol ; 86(15): 8259-68, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623791

RESUMO

Herpes simplex virus 1 infection triggers multiple changes in the metabolism of host cells, including a dramatic decrease in the levels of NAD(+). In addition to its role as a cofactor in reduction-oxidation reactions, NAD(+) is required for certain posttranslational modifications. Members of the poly(ADP-ribose) polymerase (PARP) family of enzymes are major consumers of NAD(+), which they utilize to form poly(ADP-ribose) (PAR) chains on protein substrates in response to DNA damage. PAR chains can subsequently be removed by the enzyme poly(ADP-ribose) glycohydrolase (PARG). We report here that the HSV-1 infection-induced drop in NAD(+) levels required viral DNA replication, was associated with an increase in protein poly(ADP-ribosyl)ation (PARylation), and was blocked by pharmacological inhibition of PARP-1/PARP-2 (PARP-1/2). Neither virus yield nor the cellular metabolic reprogramming observed during HSV-1 infection was altered by the rescue or further depletion of NAD(+) levels. Expression of the viral protein ICP0, which possesses E3 ubiquitin ligase activity, was both necessary and sufficient for the degradation of the 111-kDa PARG isoform. This work demonstrates that HSV-1 infection results in changes to NAD(+) metabolism by PARP-1/2 and PARG, and as PAR chain accumulation can induce caspase-independent apoptosis, we speculate that the decrease in PARG levels enhances the auto-PARylation-mediated inhibition of PARP, thereby avoiding premature death of the infected cell.


Assuntos
Glicosídeo Hidrolases/metabolismo , Herpes Simples/enzimologia , Herpesvirus Humano 1/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteólise , Células Cultivadas , Replicação do DNA/genética , DNA Viral/genética , DNA Viral/metabolismo , Glicosídeo Hidrolases/genética , Herpes Simples/genética , Herpesvirus Humano 1/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , NAD/genética , NAD/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
17.
J Biol Chem ; 283(18): 12188-201, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18308719

RESUMO

Cysteine dioxygenase (CDO) catalyzes the conversion of cysteine to cysteinesulfinic acid and is important in the regulation of intracellular cysteine levels in mammals and in the provision of oxidized cysteine metabolites such as sulfate and taurine. Several crystal structure studies of mammalian CDO have shown that there is a cross-linked cofactor present in the active site of the enzyme. The cofactor consists of a thioether bond between the gamma-sulfur of residue cysteine 93 and the aromatic side chain of residue tyrosine 157. The exact requirements for cofactor synthesis and the contribution of the cofactor to the catalytic activity of the enzyme have yet to be fully described. In this study, therefore, we explored the factors necessary for cofactor biogenesis in vitro and in vivo and examined what effect cofactor formation had on activity in vitro. Like other cross-linked cofactor-containing enzymes, formation of the Cys-Tyr cofactor in CDO required a transition metal cofactor (Fe(2+)) and O(2). Unlike other enzymes, however, biogenesis was also strictly dependent upon the presence of substrate. Cofactor formation was also appreciably slower than the rates reported for other enzymes and, indeed, took hundreds of catalytic turnover cycles to occur. In the absence of the Cys-Tyr cofactor, CDO possessed appreciable catalytic activity, suggesting that the cofactor was not essential for catalysis. Nevertheless, at physiologically relevant cysteine concentrations, cofactor formation increased CDO catalytic efficiency by approximately 10-fold. Overall, the regulation of Cys-Tyr cofactor formation in CDO by ambient cysteine levels represents an unusual form of substrate-mediated feed-forward activation of enzyme activity with important physiological consequences.


Assuntos
Aminoácidos/metabolismo , Coenzimas/biossíntese , Cisteína Dioxigenase/metabolismo , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Catálise , Linhagem Celular , Cisteína Dioxigenase/química , Cisteína Dioxigenase/isolamento & purificação , Eletroforese em Gel de Poliacrilamida , Meia-Vida , Humanos , Fígado/enzimologia , Espectrometria de Massas , Dados de Sequência Molecular , Proteínas Mutantes/isolamento & purificação , Fragmentos de Peptídeos/química , Mutação Puntual/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Especificidade por Substrato , Compostos de Sulfidrila/metabolismo , Fatores de Tempo
18.
J Biol Chem ; 282(35): 25189-98, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17581819

RESUMO

There are only two known thiol dioxygenase activities in mammals, and they are ascribed to the enzymes cysteine dioxygenase (CDO) and cysteamine (2-aminoethanethiol) dioxygenase (ADO). Although many studies have been dedicated to CDO, resulting in the identification of its gene and even characterization of the tertiary structure of the protein, relatively little is known about cysteamine dioxygenase. The failure to identify the gene for this protein has significantly hampered our understanding of the metabolism of cysteamine, a product of the constitutive degradation of coenzyme A, and the synthesis of taurine, the final product of cysteamine oxidation and the second most abundant amino acid in mammalian tissues. In this study we identified a hypothetical murine protein homolog of CDO (hereafter called ADO) that is encoded by the gene Gm237 and belongs to the DUF1637 protein family. When expressed as a recombinant protein, ADO exhibited significant cysteamine dioxygenase activity in vitro. The reaction was highly specific for cysteamine; cysteine was not oxidized by the enzyme, and structurally related compounds were not competitive inhibitors of the reaction. When overexpressed in HepG2/C3A cells, ADO increased the production of hypotaurine from cysteamine. Similarly, when endogenous expression of the human ADO ortholog C10orf22 in HepG2/C3A cells was reduced by RNA-mediated interference, hypotaurine production decreased. Western blots of murine tissues with an antibody developed against ADO showed that the protein is ubiquitously expressed with the highest levels in brain, heart, and skeletal muscle. Overall, these data suggest that ADO is responsible for endogenous cysteamine dioxygenase activity.


Assuntos
Coenzima A/metabolismo , Cisteamina/metabolismo , Dioxigenases/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Taurina/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Coenzima A/genética , Cisteína Dioxigenase/genética , Cisteína Dioxigenase/metabolismo , Dioxigenases/genética , Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos/fisiologia , Oxirredução , Interferência de RNA , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato/fisiologia , Taurina/análogos & derivados , Taurina/genética
19.
Am J Physiol Endocrinol Metab ; 293(1): E62-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17327371

RESUMO

Cysteine levels are carefully regulated in mammals to balance metabolic needs against the potential for cytotoxicity. It has been postulated that one of the major regulators of intracellular cysteine levels in mammals is cysteine dioxygenase (CDO). Hepatic expression of this catabolic enzyme increases dramatically in response to increased cysteine availability and may therefore be part of a homeostatic response to shunt excess toxic cysteine to more benign metabolites such as sulfate or taurine. Direct experimental evidence, however, is lacking to support the hypothesis that CDO is capable of altering steady-state intracellular cysteine levels. In this study, we expressed either the wild-type (WT) or a catalytically inactivated mutant (H86A) isoform of CDO in HepG2/C3A cells (which do not express endogenous CDO protein) and cultured them in different concentrations of extracellular cysteine. WT CDO, but not H86A CDO, was capable of reducing intracellular cysteine levels in cells incubated in physiologically relevant concentrations of cysteine. WT CDO also decreased the glutathione pool and potentiated the toxicity of CdCl(2). These results demonstrate that CDO is capable of altering intracellular cysteine levels as well as glutathione levels.


Assuntos
Cisteína Dioxigenase/genética , Cisteína/metabolismo , Glutationa/metabolismo , Cloreto de Cádmio/toxicidade , Linhagem Celular Tumoral , Citotoxinas/toxicidade , Humanos , Isoenzimas/genética , Modelos Biológicos , Taurina/análogos & derivados , Taurina/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA