Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transpl Infect Dis ; 24(5): e13971, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36254507

RESUMO

BACKGROUND: Antimicrobial stewardship in solid organ transplant (SOT) recipients is important to prevent antimicrobial-associated complications, but traditional stewardship principles are challenging to implement for SOT patients. Newer methodologies to optimize stewardship efforts are needed. METHODS: PubMed was searched using the keywords "cell free DNA," "metagenomic sequencing," "host biomarker," "antimicrobial stewardship," and "SOT." RESULTS: Metagenomic sequencing of cell free DNA has the potential to be a stewardship tool for SOT recipients. Various studies have shown its use for antimicrobial de-escalation and duration shortening. Host gene expression profiles can differentiate between infectious and noninfectious syndromes and may assist in stewardship efforts. However, information in immunocompromised hosts is conflicting. CONCLUSION: Microbial cell free DNA sequencing and host gene expression profiling show promise as stewardship tools in SOT recipients. Future studies on antimicrobial stewardship in SOT recipients should focus on their clinical use and feasibility.


Assuntos
Anti-Infecciosos , Gestão de Antimicrobianos , Ácidos Nucleicos Livres , Transplante de Órgãos , Antibacterianos/uso terapêutico , Anti-Infecciosos/uso terapêutico , Gestão de Antimicrobianos/métodos , Biomarcadores , Humanos , Transplante de Órgãos/efeitos adversos , Transplante de Órgãos/métodos , Transplantados
2.
Transpl Infect Dis ; 24(3): e13822, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35253965

RESUMO

BACKGROUND: Mycoplasma hominis can cause significant infections after solid organ transplantation (SOT). Treatment should be guided by susceptibility testing, but conventional lab methods are laborious with prolonged turnaround time (TAT). This case series compares the phenotypic and genotypic susceptibility profiles of M. hominis isolates identified from SOT patients. METHODS: This is a single-center retrospective study evaluating SOT recipients with confirmed M. hominis infections. Patients' demographic, clinical, microbiological, and radiographic data were collected. Culture of M. hominis isolates was performed according to current Clinical and Laboratory Standards Institute guidelines. Phenotypic susceptibility testing was performed by University of Alabama Diagnostic Mycoplasma Laboratory. Whole genome sequencing (WGS) was performed followed by bioinformatic analysis of known genetic determinants of resistance. RESULTS: Seven SOT recipients with M. hominis infections were identified. Two out of seven (28.5%) patients had resistance detected by phenotypic susceptibility testing (Case 5 to levofloxacin and Case 7 to tetracycline). Genomic analyses confirmed the presence of mutations in the parC and parE topoisomerase genes at positions conferring to fluoroquinolone resistance in the isolate from Case 5, while the tetracycline-resistant isolate from Case 7 harbored the tetM gene. The median TAT from the date of specimen collection was 24 days for phenotypic susceptibility testing and 14 days for genotypic susceptibility testing. All seven patients received antimicrobials directed toward M. hominis and recovered with complete resolution of infection. CONCLUSIONS: WGS may offer a novel and more rapid methodology for M. hominis susceptibility testing to help optimize antimicrobial usage, but more data are needed.


Assuntos
Anti-Infecciosos , Infecções por Mycoplasma , Transplante de Órgãos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anti-Infecciosos/uso terapêutico , Humanos , Testes de Sensibilidade Microbiana , Infecções por Mycoplasma/diagnóstico , Infecções por Mycoplasma/tratamento farmacológico , Infecções por Mycoplasma/microbiologia , Mycoplasma hominis/genética , Transplante de Órgãos/efeitos adversos , Estudos Retrospectivos , Tetraciclina/uso terapêutico , Resultado do Tratamento
3.
Open Forum Infect Dis ; 8(8): ofab326, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34377725

RESUMO

Measles is a worldwide viral disease that can cause fatal complications in immunocompromised hosts such as hematopoietic cell transplant (HCT) recipients. The live attenuated measles, mumps, and rubella (MMR) vaccine is generally contraindicated post-HCT due to the risk for vaccine-associated measles. This, combined with decreasing vaccination rates due to vaccine hesitancy and the coronavirus disease 2019 pandemic, raises significant concerns for a measles resurgence that could portend devastating consequences for immunocompromised hosts. Multiple guidelines have included criteria to determine which HCT recipients can safely receive the MMR vaccine. Here, we report a case of vaccine-associated measles in a HCT recipient who met guideline-recommended criteria for MMR vaccination. The objective of this article is to query these criteria, highlight the importance of MMR vaccination, and comprehensively review the literature.

4.
J Clin Lipidol ; 11(4): 964-971, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28666713

RESUMO

BACKGROUND: GPIHBP1, a glycolipid-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) in the interstitial spaces and transports it to the capillary lumen. GPIHBP1 deficiency prevents LPL from reaching the capillary lumen, resulting in low intravascular LPL levels, impaired intravascular triglyceride processing, and severe hypertriglyceridemia (chylomicronemia). A recent study showed that some cases of hypertriglyceridemia are caused by autoantibodies against GPIHBP1 ("GPIHBP1 autoantibody syndrome"). OBJECTIVE: Our objective was to gain additional insights into the frequency of the GPIHBP1 autoantibody syndrome in patients with unexplained chylomicronemia. METHODS: We used enzyme-linked immunosorbent assays to screen for GPIHBP1 autoantibodies in 33 patients with unexplained chylomicronemia and then used Western blots and immunocytochemistry studies to characterize the GPIHBP1 autoantibodies. RESULTS: The plasma of 1 patient, a 36-year-old man with severe hypertriglyceridemia, contained GPIHBP1 autoantibodies. The autoantibodies, which were easily detectable by Western blot, blocked the ability of GPIHBP1 to bind LPL. The plasma levels of LPL mass and activity were low. The patient had no history of autoimmune disease, but his plasma was positive for antinuclear antibodies. CONCLUSIONS: One of 33 patients with unexplained chylomicronemia had the GPIHBP1 autoantibody syndrome. Additional studies in large lipid clinics will be helpful for better defining the frequency of this syndrome and for exploring the best strategies for treatment.


Assuntos
Autoanticorpos/sangue , Autoanticorpos/imunologia , Hiperlipoproteinemia Tipo I/sangue , Hiperlipoproteinemia Tipo I/imunologia , Receptores de Lipoproteínas/imunologia , Adulto , Animais , Linhagem Celular , Humanos , Hiperlipoproteinemia Tipo I/complicações , Hiperlipoproteinemia Tipo I/genética , Hipertrigliceridemia/complicações , Masculino , Mutação
5.
J Lipid Res ; 53(6): 1176-82, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22448028

RESUMO

Protein farnesyltransferase (FTase) inhibitors, generally called "FTIs," block the farnesylation of prelamin A, inhibiting the biogenesis of mature lamin A and leading to an accumulation of prelamin A within cells. A recent report found that a GGTI, an inhibitor of protein geranylgeranyltransferase-I (GGTase-I), caused an exaggerated accumulation of prelamin A in the presence of low amounts of an FTI. This finding was interpreted as indicating that prelamin A can be alternately prenylated by GGTase-I and that inhibiting both protein prenyltransferases leads to more prelamin A accumulation than blocking FTase alone. Here, we tested an alternative hypothesis-GGTIs are not specific for GGTase-I, and they lead to prelamin A accumulation by inhibiting ZMPSTE24 (a zinc metalloprotease that converts farnesyl-prelamin A to mature lamin A). In our studies, commonly used GGTIs caused prelamin A accumulation in human fibroblasts, but the prelamin A in GGTI-treated cells exhibited a more rapid electrophoretic mobility than prelamin A from FTI-treated cells. The latter finding suggested that the prelamin A in GGTI-treated cells might be farnesylated (which would be consistent with the notion that GGTIs inhibit ZMPSTE24). Indeed, metabolic labeling studies revealed that the prelamin A in GGTI-treated fibroblasts is farnesylated. Moreover, biochemical assays of ZMPSTE24 activity showed that ZMPSTE24 is potently inhibited by a GGTI. Our studies show that GGTIs inhibit ZMPSTE24, leading to an accumulation of farnesyl-prelamin A. Thus, caution is required when interpreting the effects of GGTIs on prelamin A processing.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Proteínas de Membrana/antagonistas & inibidores , Metaloendopeptidases/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Peptidomiméticos/farmacologia , Inibidores de Proteases/farmacologia , Precursores de Proteínas/metabolismo , Animais , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Lamina Tipo A , Camundongos
6.
J Lipid Res ; 53(1): 77-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22039581

RESUMO

Protein farnesyltransferase (FTase) and protein geranylgeranyltransferase-I (GGTase-I) add 15- or 20-carbon lipids, respectively, to proteins that terminate with a CaaX motif. These posttranslational modifications of proteins with lipids promote protein interactions with membrane surfaces in cells, but the in vivo importance of the CaaX prenyltransferases and the protein lipidation reactions they catalyze remain incompletely defined. One study concluded that a deficiency of FTase was inconsequential in adult mice and led to little or no tissue pathology. To assess the physiologic importance of the CaaX prenyltransferases, we used conditional knockout alleles and an albumin-Cre transgene to produce mice lacking FTase, GGTase-I, or both enzymes in hepatocytes. The hepatocyte-specific FTase knockout mice survived but exhibited hepatocellular disease and elevated transaminases. Mice lacking GGTase-I not only had elevated transaminases but also had dilated bile cannaliculi, hyperbilirubinemia, hepatosplenomegaly, and reduced survival. Of note, GGTase-I-deficient hepatocytes had a rounded shape and markedly reduced numbers of actin stress fibers. Hepatocyte-specific FTase/GGTase-I double-knockout mice closely resembled mice lacking GGTase-I alone, but the disease was slightly more severe. Our studies refute the notion that FTase is dispensable and demonstrate that GGTase-I is crucial for the vitality of hepatocytes.


Assuntos
Alquil e Aril Transferases/deficiência , Dimetilaliltranstransferase/deficiência , Farnesiltranstransferase/deficiência , Hepatócitos/enzimologia , Hepatopatias/fisiopatologia , Prenilação de Proteína/efeitos dos fármacos , Animais , Fígado/patologia , Fígado/fisiopatologia , Hepatopatias/patologia , Camundongos , Camundongos Knockout
7.
Hum Mol Genet ; 20(18): 3537-44, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21659336

RESUMO

Nuclear lamins are usually classified as A-type (lamins A and C) or B-type (lamins B1 and B2). A-type lamins have been implicated in multiple genetic diseases but are not required for cell growth or development. In contrast, B-type lamins have been considered essential in eukaryotic cells, with crucial roles in DNA replication and in the formation of the mitotic spindle. Knocking down the genes for B-type lamins (LMNB1, LMNB2) in HeLa cells has been reported to cause apoptosis. In the current study, we created conditional knockout alleles for mouse Lmnb1 and Lmnb2, with the goal of testing the hypothesis that B-type lamins are crucial for the growth and viability of mammalian cells in vivo. Using the keratin 14-Cre transgene, we bred mice lacking the expression of both Lmnb1 and Lmnb2 in skin keratinocytes (Lmnb1(Δ/Δ)Lmnb2(Δ/Δ)). Lmnb1 and Lmnb2 transcripts were absent in keratinocytes of Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice, and lamin B1 and lamin B2 proteins were undetectable. But despite an absence of B-type lamins in keratinocytes, the skin and hair of Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice developed normally and were free of histological abnormalities, even in 2-year-old mice. After an intraperitoneal injection of bromodeoxyuridine (BrdU), similar numbers of BrdU-positive keratinocytes were observed in the skin of wild-type and Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice. Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) keratinocytes did not exhibit aneuploidy, and their growth rate was normal in culture. These studies challenge the concept that B-type lamins are essential for proliferation and vitality of eukaryotic cells.


Assuntos
Proliferação de Células , Cabelo/crescimento & desenvolvimento , Queratinócitos/citologia , Lamina Tipo B/deficiência , Pele/crescimento & desenvolvimento , Células 3T3 , Animais , Células Cultivadas , Feminino , Cabelo/metabolismo , Células HeLa , Humanos , Queratinócitos/metabolismo , Lamina Tipo B/genética , Masculino , Camundongos , Camundongos Knockout , Pele/metabolismo
8.
Hum Mol Genet ; 20(3): 436-44, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21088111

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is caused by a mutant prelamin A, progerin, that terminates with a farnesylcysteine. HGPS knock-in mice (Lmna(HG/+)) develop severe progeria-like disease phenotypes. These phenotypes can be ameliorated with a protein farnesyltransferase inhibitor (FTI), suggesting that progerin's farnesyl lipid is important for disease pathogenesis and raising the possibility that FTIs could be useful for treating humans with HGPS. Subsequent studies showed that mice expressing non-farnesylated progerin (Lmna(nHG/+) mice, in which progerin's carboxyl-terminal -CSIM motif was changed to -SSIM) also develop severe progeria, raising doubts about whether any treatment targeting protein prenylation would be particularly effective. We suspected that those doubts might be premature and hypothesized that the persistent disease in Lmna(nHG/+) mice could be an unanticipated consequence of the cysteine-to-serine substitution that was used to eliminate farnesylation. To test this hypothesis, we generated a second knock-in allele yielding non-farnesylated progerin (Lmna(csmHG)) in which the carboxyl-terminal -CSIM motif was changed to -CSM. We then compared disease phenotypes in mice harboring the Lmna(nHG) or Lmna(csmHG) allele. As expected, Lmna(nHG/+) and Lmna(nHG/nHG) mice developed severe progeria-like disease phenotypes, including osteolytic lesions and rib fractures, osteoporosis, slow growth and reduced survival. In contrast, Lmna(csmHG/+) and Lmna(csmHG/csmHG) mice exhibited no bone disease and displayed entirely normal body weights and survival. The frequencies of misshapen cell nuclei were lower in Lmna(csmHG/+) and Lmna(csmHG/csmHG) fibroblasts. These studies show that the ability of non-farnesylated progerin to elicit disease depends on the carboxyl-terminal mutation used to eliminate protein prenylation.


Assuntos
Farnesiltranstransferase/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progéria/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Substituição de Aminoácidos , Animais , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Feminino , Técnicas de Introdução de Genes , Imidazóis/farmacologia , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Camundongos , Mutação , Proteínas Nucleares/química , Fenótipo , Progéria/metabolismo , Progéria/patologia , Progéria/fisiopatologia , Precursores de Proteínas/química , Prenilação de Proteína
9.
Proc Natl Acad Sci U S A ; 107(11): 5076-81, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20145110

RESUMO

Nuclear lamins are components of the nuclear lamina, a structural scaffolding for the cell nucleus. Defects in lamins A and C cause an array of human diseases, including muscular dystrophy, lipodystrophy, and progeria, but no diseases have been linked to the loss of lamins B1 or B2. To explore the functional relevance of lamin B2, we generated lamin B2-deficient mice and found that they have severe brain abnormalities resembling lissencephaly, with abnormal layering of neurons in the cerebral cortex and cerebellum. This neuronal layering abnormality is due to defective neuronal migration, a process that is dependent on the organized movement of the nucleus within the cell. These studies establish an essential function for lamin B2 in neuronal migration and brain development.


Assuntos
Cerebelo/anormalidades , Cerebelo/embriologia , Córtex Cerebral/anormalidades , Córtex Cerebral/embriologia , Lamina Tipo B/deficiência , Animais , Movimento Celular , Cerebelo/patologia , Córtex Cerebral/patologia , Inativação Gênica , Lamina Tipo B/metabolismo , Camundongos , Neurônios/patologia
10.
Hum Mol Genet ; 19(8): 1603-17, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20106865

RESUMO

The modification of proteins with farnesyl or geranylgeranyl lipids, a process called protein prenylation, facilitates interactions of proteins with membrane surfaces. Protein prenylation is carried out by a pair of cytosolic enzymes, protein farnesyltransferase (FTase) and protein geranylgeranyltransferase type I (GGTase-I). FTase and GGTase-I have attracted interest as therapeutic targets for both cancer and progeria, but very little information exists on the importance of these enzymes for homeostasis of normal tissues. One study actually suggested that FTase is entirely dispensable. To explore the importance of the protein prenyltransferases for normal tissues, we used conditional knockout alleles for Fntb and Pggt1b (which encode the beta-subunits of FTase and GGTase-I, respectively) and a keratin 14-Cre transgene to create mice lacking FTase or GGTase-I in skin keratinocytes. Keratinocyte-specific Fntb knockout mice were viable but developed severe alopecia. Although hair follicles appeared normal during development, they were morphologically abnormal after birth, and ultrastructural and immunohistochemical studies revealed many apoptotic cells. The interfollicular epidermis of Fntb-deficient mice appeared normal; however, keratinocytes from these mice could not proliferate in culture. As expected, non-farnesylated prelamin A and non-farnesylated DNAJA1 accumulated in Fntb-deficient keratinocytes. Keratinocyte-specific Pggt1b knockout mice survived development but died shortly after birth. Like Fntb-deficient keratinocytes, Pggt1b-deficient keratinocytes did not proliferate in culture. Thus, both FTase and GGTase-I are required for the homeostasis of skin keratinocytes.


Assuntos
Alquil e Aril Transferases/metabolismo , Farnesiltranstransferase/metabolismo , Queratinócitos/enzimologia , Pele/enzimologia , Alquil e Aril Transferases/genética , Animais , Células Cultivadas , Farnesiltranstransferase/genética , Feminino , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Prenilação de Proteína , Pele/crescimento & desenvolvimento , Pele/metabolismo
11.
J Lipid Res ; 51(2): 400-5, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19965595

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is caused by the accumulation of a farnesylated form of prelamin A (progerin). Previously, we showed that blocking protein farnesylation with a farnesyltransferase inhibitor (FTI) ameliorates the disease phenotypes in mouse model of HGPS (Lmna(HG/+)). However, the interpretation of the FTI treatment studies is open to question in light of recent studies showing that mice expressing a nonfarnesylated version of progerin (Lmna(nHG/+)) develop progeria-like disease phenotypes. The fact that Lmna(nHG/+) mice manifest disease raised the possibility that the beneficial effects of an FTI in Lmna(HG/+) mice were not due to the effects of the drug on the farnesylation of progerin, but may have been due to unanticipated secondary effects of the drug on other farnesylated proteins. To address this issue, we compared the ability of an FTI to improve progeria-like disease phenotypes in both Lmna(HG/+) and Lmna(nHG/+) mice. In Lmna(HG/+) mice, the FTI reduced disease phenotypes in a highly significant manner, but the drug had no effect in Lmna(nHG/+) mice. The failure of the FTI to ameliorate disease in Lmna(nHG/+) mice supports the idea that the beneficial effects of an FTI in Lmna(HG/+) mice are due to the effect of drug on the farnesylation of progerin.


Assuntos
Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Progéria/tratamento farmacológico , Progéria/enzimologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Farnesiltranstransferase/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Masculino , Camundongos , Fenótipo , Prenilação/efeitos dos fármacos , Progéria/metabolismo , Progéria/patologia , Análise de Sobrevida , Fatores de Tempo
12.
J Biol Chem ; 283(11): 7094-9, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18178963

RESUMO

Hutchinson-Gilford progeria syndrome is caused by the synthesis of a mutant form of prelamin A, which is generally called progerin. Progerin is targeted to the nuclear rim, where it interferes with the integrity of the nuclear lamina, causes misshapen cell nuclei, and leads to multiple aging-like disease phenotypes. We created a gene-targeted allele yielding exclusively progerin (Lmna HG) and found that heterozygous mice (Lmna HG/+) exhibit many phenotypes of progeria. In this study, we tested the hypothesis that the phenotypes elicited by the Lmna HG allele might be modulated by compositional changes in the nuclear lamina. To explore this hypothesis, we bred mice harboring one Lmna HG allele and one Lmna LCO allele (a mutant allele that produces lamin C but no lamin A). We then compared the phenotypes of Lmna HG/LCO mice (which produce progerin and lamin C) with littermate Lmna HG/+ mice (which produce lamin A, lamin C, and progerin). Lmna HG/LCO mice exhibited improved HG/LCO fibroblasts had fewer misshapen nuclei than Lmna HG/+ fibroblasts (p < 0.0001). A likely explanation for these differences was uncovered; the amount of progerin in Lmna HG/LCO fibroblasts and tissues was lower than in Lmna HG/+ fibroblasts and tissues. These studies suggest that compositional changes in the nuclear lamina can influence both the steady-state levels of progerin and the severity of progeria-like disease phenotypes.


Assuntos
Regulação da Expressão Gênica , Lamina Tipo A/biossíntese , Lamina Tipo A/genética , Progéria/genética , Alelos , Animais , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fraturas Ósseas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Fenótipo , Precursores de Proteínas/genética
13.
Proc Natl Acad Sci U S A ; 104(33): 13432-7, 2007 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-17652517

RESUMO

HIV protease inhibitors (HIV-PIs) target the HIV aspartyl protease, which cleaves the HIV gag-pol polyprotein into shorter proteins required for the production of new virions. HIV-PIs are a cornerstone of treatment for HIV but have been associated with lipodystrophy and other side effects. In both human and mouse fibroblasts, we show that HIV-PIs caused an accumulation of prelamin A. The prelamin A in HIV-PI-treated fibroblasts migrated more rapidly than nonfarnesylated prelamin A, comigrating with the farnesylated form of prelamin A that accumulates in ZMPSTE24-deficient fibroblasts. The accumulation of farnesyl-prelamin A in response to HIV-PI treatment was exaggerated in fibroblasts heterozygous for Zmpste24 deficiency. HIV-PIs inhibited the endoproteolytic processing of a GFP-prelamin A fusion protein. The HIV-PIs did not affect the farnesylation of HDJ-2, nor did they inhibit protein farnesyltransferase in vitro. HIV-PIs also did not inhibit the activities of the isoprenyl-cysteine carboxyl methyltransferase ICMT or the prenylprotein endoprotease RCE1 in vitro, but they did inhibit ZMPSTE24 (IC(50): lopinavir, 18.4 +/- 4.6 microM; tipranavir, 1.2 +/- 0.4 microM). We conclude that the HIV-PIs inhibit ZMPSTE24, leading to an accumulation of farnesyl-prelamin A. The inhibition of ZMPSTE24 by HIV-PIs could play a role in the side effects of these drugs.


Assuntos
Inibidores da Protease de HIV/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Metaloendopeptidases/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Precursores de Proteínas/metabolismo , Sequência de Bases , Primers do DNA , Humanos , Lamina Tipo A
14.
J Clin Invest ; 116(3): 743-52, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16511604

RESUMO

Lamin A and lamin C, both products of Lmna, are key components of the nuclear lamina. In the mouse, a deficiency in both lamin A and lamin C leads to slow growth, muscle weakness, and death by 6 weeks of age. Fibroblasts deficient in lamins A and C contain misshapen and structurally weakened nuclei, and emerin is mislocalized away from the nuclear envelope. The physiologic rationale for the existence of the 2 different Lmna products lamin A and lamin C is unclear, although several reports have suggested that lamin A may have particularly important functions, for example in the targeting of emerin and lamin C to the nuclear envelope. Here we report the development of lamin C-only mice (Lmna(LCO/LCO)), which produce lamin C but no lamin A or prelamin A (the precursor to lamin A). Lmna(LCO/LCO) mice were entirely healthy, and Lmna(LCO/LCO) cells displayed normal emerin targeting and exhibited only very minimal alterations in nuclear shape and nuclear deformability. Thus, at least in the mouse, prelamin A and lamin A appear to be dispensable. Nevertheless, an accumulation of farnesyl-prelamin A (as occurs with a deficiency in the prelamin A processing enzyme Zmpste24) caused dramatically misshapen nuclei and progeria-like disease phenotypes. The apparent dispensability of prelamin A suggested that lamin A-related progeroid syndromes might be treated with impunity by reducing prelamin A synthesis. Remarkably, the presence of a single Lmna(LCO) allele eliminated the nuclear shape abnormalities and progeria-like disease phenotypes in Zmpste24-/- mice. Moreover, treating Zmpste24-/- cells with a prelamin A-specific antisense oligonucleotide reduced prelamin A levels and significantly reduced the frequency of misshapen nuclei. These studies suggest a new therapeutic strategy for treating progeria and other lamin A diseases.


Assuntos
Lamina Tipo A/fisiologia , Lâmina Nuclear/fisiologia , Proteínas Nucleares/fisiologia , Precursores de Proteínas/fisiologia , Animais , Linhagem Celular Transformada , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Debilidade Muscular/genética , Proteínas Nucleares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/genética , RNA Mensageiro/metabolismo , Crânio/anormalidades , Coluna Vertebral/anormalidades
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...