Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Circ Res ; 134(4): 351-370, 2024 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-38299369

RESUMO

BACKGROUND: Pulmonary hypertension (PH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and elevated mean pulmonary arterial pressure, resulting in right heart failure. METHODS: Here, we show that direct targeting of the endothelium to uncouple eNOS (endothelial nitric oxide synthase) with DAHP (2,4-diamino 6-hydroxypyrimidine; an inhibitor of GTP cyclohydrolase 1, the rate-limiting synthetic enzyme for the critical eNOS cofactor tetrahydrobiopterin) induces human-like, time-dependent progression of PH phenotypes in mice. RESULTS: Critical phenotypic features include progressive elevation in mean pulmonary arterial pressure, right ventricular systolic blood pressure, and right ventricle (RV)/left ventricle plus septum (LV+S) weight ratio; extensive vascular remodeling of pulmonary arterioles with increased medial thickness/perivascular collagen deposition and increased expression of PCNA (proliferative cell nuclear antigen) and alpha-actin; markedly increased total and mitochondrial superoxide production, substantially reduced tetrahydrobiopterin and nitric oxide bioavailabilities; and formation of an array of human-like vascular lesions. Intriguingly, novel in-house generated endothelial-specific dihydrofolate reductase (DHFR) transgenic mice (tg-EC-DHFR) were completely protected from the pathophysiological and molecular features of PH upon DAHP treatment or hypoxia exposure. Furthermore, DHFR overexpression with a pCMV-DHFR plasmid transfection in mice after initiation of DAHP treatment completely reversed PH phenotypes. DHFR knockout mice spontaneously developed PH at baseline and had no additional deterioration in response to hypoxia, indicating an intrinsic role of DHFR deficiency in causing PH. RNA-sequencing experiments indicated great similarity in gene regulation profiles between the DAHP model and human patients with PH. CONCLUSIONS: Taken together, these results establish a novel human-like murine model of PH that has long been lacking in the field, which can be broadly used for future mechanistic and translational studies. These data also indicate that targeting endothelial DHFR deficiency represents a novel and robust therapeutic strategy for the treatment of PH.


Assuntos
Hipertensão Pulmonar , Tetra-Hidrofolato Desidrogenase , Animais , Humanos , Camundongos , Endotélio/metabolismo , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/genética , Hipóxia , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Tetra-Hidrofolato Desidrogenase/deficiência , Hipoxantinas , Modelos Animais de Doenças
2.
Elife ; 82019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31573512

RESUMO

The mechanisms of adaptation to inactivation of essential genes remain unknown. Here we inactivate E. coli dihydrofolate reductase (DHFR) by introducing D27G,N,F chromosomal mutations in a key catalytic residue with subsequent adaptation by an automated serial transfer protocol. The partial reversal G27- > C occurred in three evolutionary trajectories. Conversely, in one trajectory for D27G and in all trajectories for D27F,N strains adapted to grow at very low metabolic supplement (folAmix) concentrations but did not escape entirely from supplement auxotrophy. Major global shifts in metabolome and proteome occurred upon DHFR inactivation, which were partially reversed in adapted strains. Loss-of-function mutations in two genes, thyA and deoB, ensured adaptation to low folAmix by rerouting the 2-Deoxy-D-ribose-phosphate metabolism from glycolysis towards synthesis of dTMP. Multiple evolutionary pathways of adaptation converged to a suboptimal solution due to the high accessibility to loss-of-function mutations that block the path to the highest, yet least accessible, fitness peak.


Assuntos
Adaptação Biológica , Escherichia coli/enzimologia , Escherichia coli/crescimento & desenvolvimento , Genes Essenciais , Tetra-Hidrofolato Desidrogenase/deficiência , Escherichia coli/genética , Evolução Molecular , Metaboloma , Mutação de Sentido Incorreto , Proteoma , Inoculações Seriadas , Tetra-Hidrofolato Desidrogenase/genética
3.
Redox Biol ; 24: 101185, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30954686

RESUMO

Hypertension and abdominal aortic aneurysm (AAA) are severe cardiovascular diseases with incompletely defined molecular mechanisms. In the current study we generated dihydrofolate reductase (DHFR) knockout mice for the first time to examine its potential contribution to the development of hypertension and AAA, as well as the underlying molecular mechanisms. Whereas the homozygote knockout mice were embryonically lethal, the heterozygote knockout mice had global reduction in DHFR protein expression and activity. Angiotensin II infusion into these animals resulted in substantially exaggerated elevation in blood pressure and development of AAA, which was accompanied by excessive eNOS uncoupling activity (featured by significantly impaired tetrahydrobiopterin and nitric oxide bioavailability), vascular remodeling (MMP2 activation, medial elastin breakdown and adventitial fibrosis) and inflammation (macrophage infiltration). Importantly, scavenging of mitochondrial reactive oxygen species with Mito-Tempo in vivo completely abrogated development of hypertension and AAA in DHFR knockout mice, indicating a novel role of mitochondria in mediating hypertension and AAA downstream of DHFR deficiency-dependent eNOS uncoupling. These data for the first time demonstrate that targeting DHFR-deficiency driven mitochondrial dysfunction may represent an innovative therapeutic option for the treatment of AAA and hypertension.


Assuntos
Anemia Megaloblástica/complicações , Aneurisma da Aorta Abdominal/etiologia , Aneurisma da Aorta Abdominal/metabolismo , Hipertensão/etiologia , Hipertensão/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Tetra-Hidrofolato Desidrogenase/deficiência , Angiotensina II/metabolismo , Animais , Aneurisma da Aorta Abdominal/patologia , Pressão Sanguínea , Modelos Animais de Doenças , Loci Gênicos , Hipertensão/diagnóstico , Hipertensão/fisiopatologia , Macrófagos/metabolismo , Macrófagos/patologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Ultrassonografia
4.
J Biosci Bioeng ; 128(4): 487-494, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31031194

RESUMO

Since the use of protein therapeutics is effective for treating intractable human diseases, the production of biologic therapeutic agents has dramatically increased over the past three decades. The Chinese hamster ovary (CHO) cell lines are the most commonly used host cell expression system for recombinant protein production. High productive and stable clonal cell lines for recombinant protein production have been established from the DHFR-deficient CHO cell using the dihydrofolate reductase/methotrexate (DHFR/MTX) selection methods. Human embryonic kidney 293 (HEK293) cells are alternative host cells widely used for protein production. In most case, however, the cells are used for the transient expression, and there is no gene amplification system in HEK293 cells. In this study, we established a DHFR-deficient HEK293 cell line for the high yield of recombinant proteins. We doubly knocked out DHFR and DHFR2 in the MAN1A1/A2/B1/C1-quadruple knockout HEK293 (QD-KO) cells, using the CRISPR/Cas9 system. The DHFR-deficient QD-KO cells were used to overexpress two proteins, lysosomal acid lipase and the constant fragment of human immunoglobulin G1 by the DHFR/MTX gene-amplification method. This method resulted in a dramatic increase in the two protein expressions in the DHFR-deficient QD-KO cells by increasing MTX concentration. Our system could be adopted in the production of several recombinant proteins including therapeutic proteins.


Assuntos
Glicoproteínas/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Animais , Técnicas de Cultura de Células , Cricetinae , Cricetulus , Amplificação de Genes , Glicoproteínas/genética , Células HEK293 , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tetra-Hidrofolato Desidrogenase/deficiência
5.
Redox Biol ; 11: 118-125, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27912196

RESUMO

Oxidative stress plays an important role in the formation of abdominal aortic aneurysm (AAA), and we have recently established a causal role of uncoupled eNOS in this severe human disease. We have also shown that activation of NADPH oxidase (NOX) lies upstream of uncoupled eNOS. Therefore, identification of the specific NOX isoforms that are required for eNOS uncoupling and AAA formation would ultimately lead to novel therapies for AAA. In the present study, we used the Ang II infused hph-1 mice to examine the roles of NOX isoforms in the development of AAA. We generated double mutants of hph-1-NOX1, hph-1-NOX2, hph-1-p47phox, and hph-1-NOX4. After two weeks of Ang II infusion, the incidence rate of AAA substantially dropped from 76.5% in Ang II infused hph-1 mice (n=34) to 11.1%, 15.0%, 9.5% and 0% in hph-1-NOX1 (n=27), hph-1-NOX2 (n=40), hph-1-p47phox (n=21), and hph-1-NOX4 (n=33) double mutant mice, respectively. The size of abdominal aortas of the four double mutant mice, determined by ultrasound analyses, was significantly smaller than the hph-1 mice. Aortic nitric oxide and H4B bioavailabilities were markedly improved in the double mutants, while superoxide production and eNOS uncoupling activity were substantially diminished. These effects seemed attributed to an endothelial specific restoration of dihydrofolate reductase expression and activity, deficiency of which has been shown to induce eNOS uncoupling and AAA formation in both Ang II-infused hph-1 and apoE null animals. In addition, over-expression of human NOX4 N129S or T555S mutant newly identified in aneurysm patients increased hydrogen peroxide production, further implicating a relationship between NOX and human aneurysm. Taken together, these data indicate that NOX isoforms 1, 2 or 4 lies upstream of dihydrofolate reductase deficiency and eNOS uncoupling to induce AAA formation. These findings may promote development of novel therapeutics for the treatment of the disease by inhibiting NOX signaling.


Assuntos
Aneurisma da Aorta Abdominal/genética , NADPH Oxidase 4/genética , NADPH Oxidases/genética , Óxido Nítrico Sintase Tipo III/genética , Complexo Repressor Polycomb 1/genética , Angiotensina II/metabolismo , Animais , Aneurisma da Aorta Abdominal/fisiopatologia , Apolipoproteínas E/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Mutação , Estresse Oxidativo/genética , Isoformas de Proteínas/genética , Superóxidos/metabolismo , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolato Desidrogenase/genética
6.
Biotechnol Prog ; 32(6): 1539-1546, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27578320

RESUMO

Adaptation of dihydrofolate reductase (DHFR)-deficient Chinese hamster ovary (CHO) DG44 cells to chemically defined suspension culture conditions is a time-consuming and labor-intensive process because nonadapted DHFR-deficient CHO DG44 cells normally show poor growth in chemically defined medium (CDM). We examined the effects of folate derivatives, ribonucleotides, and nucleobases on the growth of suspension-adapted DHFR-deficient CHO DG44 cells in CDM. Among the tested additives, tetrahydrofolate (THF) was identified as an effective component for increasing cell growth. THF supplementation in the range of 0.2-359 µM enhanced cell growth in in-house CDM. Addition of 3.6 µM THF to in-house CDM resulted in a more than 2.5-fold increase in maximum viable cell density. Moreover, supplementation of six different commercial CDMs with 3.6 µM THF yielded up to 2.9-fold enhancement of maximum viable cell density. An anchorage- and serum-dependent DHFR-deficient CHO DG44 cell line was adapted within two consecutive passages to suspension growth in in-house CDM supplemented with 3.6 µM THF. These data indicate that supplementation of chemically defined cell culture media with greater than 0.2 µM THF can help achieve a high density of suspension-adapted DHFR-deficient CHO DG44 cells and may facilitate rapid adaptation of nonadapted DHFR-deficient CHO DG44 cells to suspension culture. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1539-1546, 2016.


Assuntos
Meios de Cultura/farmacologia , Tetra-Hidrofolato Desidrogenase/metabolismo , Tetra-Hidrofolatos/farmacologia , Animais , Células CHO , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cricetulus , Meios de Cultura/química , Relação Dose-Resposta a Droga , Relação Estrutura-Atividade , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolatos/química
7.
Biotechnol Prog ; 29(4): 980-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23606666

RESUMO

Therapeutic monoclonal antibodies (mAb) are often produced in Chinese hamster ovary (CHO) cells. Three commonly used CHO host cells for generating stable cell lines to produce therapeutic proteins are dihydrofolate reductase (DHFR) positive CHOK1, DHFR-deficient DG44, and DUXB11-based DHFR deficient CHO. Current Genentech commercial full-length antibody products have all been produced in the DUXB11-derived DHFR-deficient CHO host. However, it has been challenging to develop stable cell lines producing an appreciable amount of antibody proteins in the DUXB11-derived DHFR-deficient CHO host for some antibody molecules and the CHOK1 host has been explored as an alternative approach. In this work, stable cell lines were developed for three antibody molecules in both DUXB11-based and CHOK1 hosts. Results have shown that the best CHOK1 clones produce about 1 g/l for an antibody mAb1 and about 4 g/l for an antibody mAb2 in 14-day fed batch cultures in shake flasks. In contrast, the DUXB11-based host produced ∼0.1 g/l for both antibodies in the same 14-day fed batch shake flask production experiments. For an antibody mAb3, both CHOK1 and DUXB11 host cells can generate stable cell lines with the best clone in each host producing ∼2.5 g/l. Additionally, studies have shown that the CHOK1 host cell has a larger endoplasmic reticulum and higher mitochondrial mass.


Assuntos
Anticorpos Monoclonais/biossíntese , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolato Desidrogenase/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetulus , Retículo Endoplasmático/metabolismo , Citometria de Fluxo , Mitocôndrias/metabolismo , Tetra-Hidrofolato Desidrogenase/genética
8.
Appl Microbiol Biotechnol ; 92(4): 779-90, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21792592

RESUMO

In Chinese hamster ovary (CHO) cells, rapid glucose metabolism normally leads to inefficient use of glucose, most of which is converted to lactate during cell cultures. Since lactate accumulation during the culture often exerts a negative effect on cell growth and valuable product formation, several genetic engineering approaches have been developed to suppress lactate dehydrogenase-A (LDH-A), the enzyme converting pyruvate into lactate. However, despite the reduced lactate accumulation, such cell cultures are eventually terminated in the late period of the culture, mainly due to apoptosis. Therefore, we developed an apoptosis-resistant, less lactate-producing dhfr(-) CHO cell line (CHO-Bcl2-LDHAsi) by overexpressing Bcl-2, one of the most well-known anti-apoptotic proteins, and by downregulating LDH-A in a dhfr(-) CHO cell line. When the dhfr(-) CHO-Bcl2-LDHAsi cell line was used as a host cell line for the development of recombinant CHO (rCHO) cells producing an Fc-fusion protein, the culture longevity of the rCHO cells was extended without any detrimental effect of genetic engineering on specific protein productivity. Simultaneously, the specific lactate production rate and apparent yield of lactate from glucose were reduced to 21-65% and 37-78% of the control cells, respectively. Taken together, these results show that the use of an apoptosis-resistant, less lactate-producing dhfr(-) CHO cell line as a host cell line saves the time and the effort of establishing an apoptosis-resistant, less lactate-producing rCHO cells for producing therapeutic proteins.


Assuntos
L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Tetra-Hidrofolato Desidrogenase/deficiência , Animais , Apoptose , Células CHO , Sobrevivência Celular , Cricetinae , Cricetulus , Expressão Gênica , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Lactato Desidrogenase 5 , Oxirredução , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
9.
Am J Hum Genet ; 88(2): 216-25, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21310276

RESUMO

Dihydrofolate reductase (DHFR) is a critical enzyme in folate metabolism and an important target of antineoplastic, antimicrobial, and antiinflammatory drugs. We describe three individuals from two families with a recessive inborn error of metabolism, characterized by megaloblastic anemia and/or pancytopenia, severe cerebral folate deficiency, and cerebral tetrahydrobiopterin deficiency due to a germline missense mutation in DHFR, resulting in profound enzyme deficiency. We show that cerebral folate levels, anemia, and pancytopenia of DHFR deficiency can be corrected by treatment with folinic acid. The characterization of this disorder provides evidence for the link between DHFR and metabolism of cerebral tetrahydrobiopterin, which is required for the formation of dopamine, serotonin, and norepinephrine and for the hydroxylation of aromatic amino acids. Moreover, this relationship provides insight into the role of folates in neurological conditions, including depression, Alzheimer disease, and Parkinson disease.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Anemia Megaloblástica/genética , Pancitopenia/genética , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolato Desidrogenase/genética , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Sequência de Aminoácidos , Anemia Megaloblástica/tratamento farmacológico , Anemia Megaloblástica/patologia , Sequência de Bases , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Encéfalo/enzimologia , Encéfalo/patologia , Feminino , Ácido Fólico/líquido cefalorraquidiano , Deficiência de Ácido Fólico/líquido cefalorraquidiano , Humanos , Lactente , Leucovorina/uso terapêutico , Imageamento por Ressonância Magnética , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Pancitopenia/tratamento farmacológico , Pancitopenia/patologia , Linhagem , Conformação Proteica , Homologia de Sequência de Aminoácidos , Tetra-Hidrofolato Desidrogenase/química
10.
Am J Hum Genet ; 88(2): 226-31, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21310277

RESUMO

The importance of intracellular folate metabolism is illustrated by the severity of symptoms and complications caused by inborn disorders of folate metabolism or by folate deficiency. We examined three children of healthy, distantly related parents presenting with megaloblastic anemia and cerebral folate deficiency causing neurologic disease with atypical childhood absence epilepsy. Genome-wide homozygosity mapping revealed a candidate region on chromosome 5 including the dihydrofolate reductase (DHFR) locus. DHFR sequencing revealed a homozygous DHFR mutation, c.458A>T (p.Asp153Val), in all siblings. The patients' folate profile in red blood cells (RBC), plasma, and cerebrospinal fluid (CSF), analyzed by liquid chromatography tandem mass spectrometry, was compatible with DHFR deficiency. DHFR activity and fluorescein-labeled methotrexate (FMTX) binding were severely reduced in EBV-immortalized lymphoblastoid cells of all patients. Heterozygous cells displayed intermediate DHFR activity and FMTX binding. RT-PCR of DHFR mRNA revealed no differences between wild-type and DHFR mutation-carrying cells, whereas protein expression was reduced in cells with the DHFR mutation. Treatment with folinic acid resulted in the resolution of hematological abnormalities, normalization of CSF folate levels, and improvement of neurological symptoms. In conclusion, the homozygous DHFR mutation p.Asp153Val causes DHFR deficiency and leads to a complex hematological and neurological disease that can be successfully treated with folinic acid. DHFR is necessary for maintaining sufficient CSF and RBC folate levels, even in the presence of adequate nutritional folate supply and normal plasma folate.


Assuntos
Anemia Megaloblástica/genética , Deficiência de Ácido Fólico/diagnóstico , Mutação/genética , Doenças do Sistema Nervoso/genética , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolato Desidrogenase/genética , Anemia Megaloblástica/diagnóstico , Criança , Pré-Escolar , Eritrócitos/metabolismo , Feminino , Fluoresceínas/metabolismo , Ácido Fólico/sangue , Ácido Fólico/líquido cefalorraquidiano , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/líquido cefalorraquidiano , Homozigoto , Humanos , Masculino , Metotrexato/análogos & derivados , Metotrexato/metabolismo , Modelos Moleculares , Doenças do Sistema Nervoso/diagnóstico , Linhagem , Conformação Proteica , Tetra-Hidrofolato Desidrogenase/química
11.
J Biotechnol ; 152(4): 189-93, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21345355

RESUMO

DHFR-deficient CHO cells are the most commonly used host cells in the biopharmaceutical industry and over the years, individual substrains have evolved, some have been engineered with improved properties and platform technologies have been designed around them. Unexpectedly, we have observed that different DHFR-deficient CHO cells show only poor growth in fed-batch cultures even in HT supplemented medium, whereas antibody producer cells derived from these hosts achieved least 2-3 fold higher peak cell densities. Using a set of different expression vectors, we were able to show that this impaired growth performance was not due to the selection procedure possibly favouring fast growing clones, but a direct consequence of DHFR deficiency. Re-introduction of the DHFR gene reproducibly restored the growth phenotype to the level of wild-type CHO cells or even beyond which seemed to be dose-dependent. The requirement for a functional DHFR gene to achieve optimal growth under production conditions has direct implications for cell line generation since it suggests that changing to a selection system other than DHFR would require another CHO host which - especially for transgenic CHO strains and tailor-suited process platforms - this could mean significant investments and potential changes in product quality. In these cases, DHFR engineering of the current CHO-DG44 or DuxB11-based host could be an attractive alternative.


Assuntos
Biofarmácia/métodos , Células CHO/fisiologia , Proliferação de Células/efeitos dos fármacos , Meios de Cultura Livres de Soro/química , Hipoxantina/farmacologia , Tetra-Hidrofolato Desidrogenase/deficiência , Timidina/farmacologia , Animais , Células CHO/efeitos dos fármacos , Cricetinae , Cricetulus , Primers do DNA/genética , Relação Dose-Resposta a Droga , Técnicas de Inativação de Genes , Reação em Cadeia da Polimerase , Tetra-Hidrofolato Desidrogenase/genética
12.
Proteomics ; 10(12): 2292-302, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20391528

RESUMO

Chinese hamster ovary (CHO) cells are the major mammalian host for producing various therapeutic proteins. Among CHO cells, the dihydrofolate reductase-deficient CHO DG44 cell line has been used as a popular mammalian host because of the availability of a well-characterized genetic selection and amplification system. However, this cell line has not been studied at the proteome level. Here, the first detailed proteome analysis of the CHO DG44 cell line is described. A protein reference map of the CHO DG44 cell line was established by analyzing whole cellular proteins using 2-DE with various immobilized pH gradients (pHs 3-10, 5-8, and 3-6) in the first dimension and a 12% acrylamide gel in the second dimension. The map is composed of over 1400 silver-stained protein spots. Among them, 179 protein spots, which represent proteins associated with various biological processes and cellular compartments, were identified based on MALDI-TOF-MS and MS/MS. This proteome database should be valuable for better understanding of CHO cell physiology and protein expression patterns which may lead to efficient therapeutic protein production.


Assuntos
Eletroforese em Gel Bidimensional , Tetra-Hidrofolato Desidrogenase/deficiência , Animais , Células CHO , Cricetinae , Cricetulus , Eletroforese em Gel de Poliacrilamida , Concentração de Íons de Hidrogênio , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Tetra-Hidrofolato Desidrogenase/genética
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(11): 1002-4, 2009 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-19900367

RESUMO

AIM: To obtain high level expression of recombinant human truncated osteoprotegerin (TOPG) with higher bioactivity in CHO-DHFR(-) cells. METHODS: The recombinant vector pcDNA3.1/DHFR-TOPG was constructed and transfected into CHO-DHFR(-) cells by the directions of LipofectAMINE 2000 for stable expression. The stable expression cell strains were screened by selective medium IMDM with 50 mL/L FCS, then serially passed in methotraxate (MTX) for gene amplification. The expression were analyzed by ELISA and RT-PCR. At last, the bioactivity analysis was performed in vitro. RESULTS: The expression level of recombinant truncated human OPG was up to 6 mg/L x 72 h, and it had significant suppression effect on the formation of OLC (P<0.05). CONCLUSION: Recombinant truncated human OPG has high expression and bioactivity. The results make it possible for further studying and clinical implying of OPG.


Assuntos
Osteoprotegerina/química , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/farmacologia , Tetra-Hidrofolato Desidrogenase/deficiência , Animais , Células CHO , Clonagem Molecular , Cricetinae , Cricetulus , DNA Complementar/genética , Expressão Gênica , Vetores Genéticos/genética , Humanos , Camundongos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/isolamento & purificação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Biologicals ; 37(5): 277-81, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19457681

RESUMO

DHFR-deficient Chinese hamster ovary (CHO DHFR(-)) cells are the most popular mammalian expression system for inducible amplification of transgene. In order to obtain more stable transfected CHO DHFR(-) cell clones, transfection efficiency of electroporation under different conditions were systemically investigated using plasmid pSV-beta-Gal as reporter gene. Transfection efficiency was proportionally increased with pulse duration and number of pulse applied. In addition, higher transfection efficiency was found in high salt extracellular solution (Berg's and Hank's buffers) than in intracellular solution (cytomix buffer) under the same electroporation condition. The highest transfection efficiency in examined conditions was about 1 in 350 cells (or 0.289%) when cells were electroporated with twice pulses at 400V, 375microF. The present study offers an optimized guideline for introducing exogenous DNA into CHO DHFR(-) cells by electroporation.


Assuntos
Células CHO , Eletroporação , Expressão Gênica , Tetra-Hidrofolato Desidrogenase/genética , Transfecção/métodos , Animais , Soluções Tampão , Cricetinae , Cricetulus , Eficiência , Eletroporação/métodos , Genes , Genes Reporter , Vetores Genéticos , Tetra-Hidrofolato Desidrogenase/deficiência
16.
Proc Natl Acad Sci U S A ; 105(15): 5809-14, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18359850

RESUMO

Gene knockout is the most powerful tool for determining gene function or permanently modifying the phenotypic characteristics of a cell. Existing methods for gene disruption are limited by their efficiency, time to completion, and/or the potential for confounding off-target effects. Here, we demonstrate a rapid single-step approach to targeted gene knockout in mammalian cells, using engineered zinc-finger nucleases (ZFNs). ZFNs can be designed to target a chosen locus with high specificity. Upon transient expression of these nucleases the target gene is first cleaved by the ZFNs and then repaired by a natural-but imperfect-DNA repair process, nonhomologous end joining. This often results in the generation of mutant (null) alleles. As proof of concept for this approach we designed ZFNs to target the dihydrofolate reductase (DHFR) gene in a Chinese hamster ovary (CHO) cell line. We observed biallelic gene disruption at frequencies >1%, thus obviating the need for selection markers. Three new genetically distinct DHFR(-/-) cell lines were generated. Each new line exhibited growth and functional properties consistent with the specific knockout of the DHFR gene. Importantly, target gene disruption is complete within 2-3 days of transient ZFN delivery, thus enabling the isolation of the resultant DHFR(-/-) cell lines within 1 month. These data demonstrate further the utility of ZFNs for rapid mammalian cell line engineering and establish a new method for gene knockout with application to reverse genetics, functional genomics, drug discovery, and therapeutic recombinant protein production.


Assuntos
Desoxirribonucleases/metabolismo , Deleção de Genes , Técnicas Genéticas , Animais , Linhagem Celular , Inativação Gênica , Métodos , Mutagênese Sítio-Dirigida , Engenharia de Proteínas , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolato Desidrogenase/genética , Dedos de Zinco
17.
G Ital Nefrol ; 25(1): 32-41, 2008.
Artigo em Italiano | MEDLINE | ID: mdl-18264916

RESUMO

This review examines the history of folate, its metabolism and its relationship to drugs and diseases. The scientific interest in folate has been increasing in recent years because of new findings related to its important role in many diseases like macrocytic anemia, congenital malformations, vascular thrombosis, atherosclerotic disease and cancer. The fascinating puzzle of folate is analyzed and the most recent news about folate treatment in patients with chronic renal failure is reported.


Assuntos
Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Carbono/metabolismo , Doenças Cardiovasculares/metabolismo , Anormalidades Congênitas/genética , Anormalidades Congênitas/metabolismo , Endotélio Vascular/fisiopatologia , Ácido Fólico/fisiologia , Ácido Fólico/uso terapêutico , Antagonistas do Ácido Fólico/uso terapêutico , Deficiência de Ácido Fólico/complicações , Deficiência de Ácido Fólico/genética , Homocisteína/metabolismo , Humanos , Falência Renal Crônica/metabolismo , Metionina/metabolismo , Metilação , Metilenotetra-Hidrofolato Redutase (NADPH2)/deficiência , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Tetra-Hidrofolato Desidrogenase/deficiência , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/fisiologia , Tetra-Hidrofolatos/metabolismo
18.
Rev Neurol (Paris) ; 163(10): 904-10, 2007 Oct.
Artigo em Francês | MEDLINE | ID: mdl-18033026

RESUMO

INTRODUCTION: Homocysteine lies at an important metabolic branch point; it may be either converted to cystathionine through the transsulfuration pathway, or methylated to form methionine. Hyperhomocysteinemia may result from hereditary defects affecting one of these reactions. STATE OF ART: Cystathionine beta synthase or 5,10-methylenetetrahydrofolate deficiency can both result in homocystinuria. Current knowledge about biochemical mechanisms leading to hyperhomocysteinemia, clinical and radiological features, pathogenesis and treatment are reviewed, focusing on late onset forms of these diseases which can be diagnosed in adulthood. CBS deficiency is characterized by lens dislocation, skeletal abnormalities, neurologic disturbances and thromboembolism. MTHFR deficiency leads to various neurological symptoms, ranging from developmental delay to encephalopathy, including motor and gait abnormalities, seizures, psychiatric manifestations and rarely strokes. The treatment of CBS deficiency depends on vitamin B6, whereas MTHFR deficiency can be efficiently treated by vitamin B12, folic acid, and betaine. PERSPECTIVES: Homocysteinemia should be measured in patients with unexplained neurological manifestations or thromboembolism.


Assuntos
Homocistinúria/terapia , Tetra-Hidrofolato Desidrogenase/deficiência , Animais , Ácido Fólico/sangue , Homocisteína/sangue , Homocisteína/metabolismo , Homocistinúria/diagnóstico , Homocistinúria/genética , Homocistinúria/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Doenças do Sistema Nervoso/diagnóstico , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/terapia , Tetra-Hidrofolato Desidrogenase/genética
19.
Vaccine ; 25(20): 4103-11, 2007 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-17428585

RESUMO

Chinese hamster ovary (CHO) cells and dihydrofolate reductase (dhfr)/methotrexate (MTX) gene amplification system are routinely used to generate stable producer CHO cell clones in biopharmaceutical industries. The present study proposes a novel method by the co-amplification of the silencing vector targeted to dhfr gene for improvements of selecting high-producing clones in dhfr-deficient and wild-type CHO cells. Using the silencing vector also resulted in improving the stability of the recombinant protein expression in the absence of MTX in the CHO/dhFr(-) and wild-type CHO cells. This new method is proposed to generate highly expressed stable cell clones of both dhfr-deficient and wild-type CHO cells for recombinant antigen production. Utilization of the silencing vector designed in this study can improve antigen expression through dhfr-directed gene amplification in other dhfr-competent cell lines for vaccine development.


Assuntos
Antígenos/biossíntese , Vetores Genéticos/genética , Interferência de RNA , Tetra-Hidrofolato Desidrogenase/genética , Animais , Antígenos/genética , Sequência de Bases , Células CHO , Células Clonais , Cricetinae , Cricetulus , Amplificação de Genes , Proteínas de Fluorescência Verde , Metotrexato/farmacologia , Camundongos , Dados de Sequência Molecular , Tetra-Hidrofolato Desidrogenase/biossíntese , Tetra-Hidrofolato Desidrogenase/deficiência , Transfecção
20.
Malar J ; 6: 35, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17378942

RESUMO

BACKGROUND: Plasmodium falciparum is the predominant human malaria species in Mozambique and a lead cause of mortality among children and pregnant women nationwide. Sulphadoxine/pyrimethamine (S/P) is used as first line antimalarial treatment as a partner drug in combination with artesunate. METHODS: A total of 92 P. falciparum-infected blood samples, from children with uncomplicated malaria attending the Centro de Saude de Bagamoyo in the Province of Maputo-Mozambique, were screened for S/P resistance-conferring mutations in the pfdhfr and pfdhps genes using a nested mutation-specific polymerase chain reaction and restriction digestion (PCR-RFLP). The panel of genetic polymorphisms analysed included the pfdhfr 164L mutation, previously reported to be absent or rare in Africa. RESULTS: The frequency of the S/P resistance-associated pfdhfr triple mutants (51I/59R/108N) and of pfdhfr/pfdhps quintuple mutants (51I/59R/108N + 437G/540E) was 93% and 47%, respectively. However, no pfdhfr 164L mutants were detected. CONCLUSION: The observation that a considerably high percentage of P. falciparum parasites contained S/P resistance-associated mutations raises concerns about the validity of this drug as first-choice treatment in Mozambique. On the other hand, no pfdhfr 164L mutant was disclosed, corroborating the view that that this allele is still rare in Africa.


Assuntos
Antimaláricos/farmacologia , Malária Falciparum/parasitologia , Mutação , Plasmodium falciparum/genética , Pirimetamina/farmacologia , Sulfadoxina/farmacologia , Tetra-Hidrofolato Desidrogenase/deficiência , Adolescente , Animais , Artemisininas/farmacologia , Artesunato , Criança , Pré-Escolar , Códon , Di-Hidropteroato Sintase/genética , Di-Hidropteroato Sintase/metabolismo , Combinação de Medicamentos , Resistência a Múltiplos Medicamentos/genética , Feminino , Humanos , Lactente , Malária Falciparum/tratamento farmacológico , Masculino , Moçambique , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/enzimologia , Sesquiterpenos/farmacologia , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...