Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 150(2): 465-76, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26481854

RESUMO

BACKGROUND & AIMS: Crohn's disease (CD) is associated with a dysregulated immune response to commensal micro-organisms in the intestine. Mice deficient in inositol polyphosphate 5'-phosphatase D (INPP5D, also known as SHIP) develop intestinal inflammation resembling that of patients with CD. SHIP is a negative regulator of PI3Kp110α activity. We investigated mechanisms of intestinal inflammation in Inpp5d(-/-) mice (SHIP-null mice), and SHIP levels and activity in intestinal tissues of subjects with CD. METHODS: We collected intestines from SHIP-null mice, as well as Inpp5d(+/+) mice (controls), and measured levels of cytokines of the interleukin 1 (IL1) family (IL1α, IL1ß, IL1ra, and IL6) by enzyme-linked immunosorbent assay. Macrophages were isolated from lamina propria cells of mice, IL1ß production was measured, and mechanisms of increased IL1ß production were investigated. Macrophages were incubated with pan-phosphatidylinositol 3-kinase inhibitors or PI3Kp110α-specific inhibitors. Some mice were given an antagonist of the IL1 receptor; macrophages were depleted from ilea of mice using clodronate-containing liposomes. We obtained ileal biopsies from sites of inflammation and peripheral blood mononuclear cells (PBMCs) from treatment-naïve subjects with CD or without CD (controls), and measured SHIP levels and activity. PBMCs were incubated with lipopolysaccharide and adenosine triphosphate, and levels of IL1ß production were measured. RESULTS: Inflamed intestinal tissues and intestinal macrophages from SHIP-null mice produced higher levels of IL1B and IL18 than intestinal tissues from control mice. We found PI3Kp110α to be required for macrophage transcription of Il1b. Macrophage depletion or injection of an IL1 receptor antagonist reduced ileal inflammation in SHIP-null mice. Inflamed ileal tissues and PBMCs from patients with CD had lower levels of SHIP protein than controls (P < .0001 and P < .0002, respectively). There was an inverse correlation between levels of SHIP activity in PBMCs and induction of IL1ß production by lipopolysaccharide and adenosine triphosphate (R(2) = .88). CONCLUSIONS: Macrophages from SHIP-deficient mice have increased PI3Kp110α-mediated transcription of Il1b, which contributes to spontaneous ileal inflammation. SHIP levels and activity are lower in intestinal tissues and peripheral blood samples from patients with CD than controls. There is an inverse correlation between SHIP activity and induction of IL1ß production by lipopolysaccharide and adenosine triphosphate in PBMCs. Strategies to reduce IL1B might be developed to treat patients with CD found to have low SHIP activity.


Assuntos
Doença de Crohn/enzimologia , Ileíte/enzimologia , Íleo/enzimologia , Interleucina-1beta/metabolismo , Macrófagos/enzimologia , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases , Doença de Crohn/diagnóstico , Doença de Crohn/genética , Doença de Crohn/imunologia , Modelos Animais de Doenças , Humanos , Ileíte/diagnóstico , Ileíte/genética , Ileíte/imunologia , Íleo/imunologia , Íleo/patologia , Inositol Polifosfato 5-Fosfatases , Interleucina-18/metabolismo , Interleucina-1beta/genética , Leucócitos Mononucleares/enzimologia , Leucócitos Mononucleares/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Inibidores de Fosfoinositídeo-3 Quinase , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Inibidores de Proteínas Quinases/farmacologia , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-1/metabolismo , Transcrição Gênica , Regulação para Cima
2.
Methods Mol Biol ; 946: 225-39, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23179835

RESUMO

Macrophages play a key role in the innate immune response and help to direct the acquired immune response. Early in the innate immune response, they produce reactive oxygen species and pro-inflammatory cytokines and chemokines to drive inflammation and are referred to as "classically activated" or "killer" macrophages (M1). During the resolution phase of inflammation, they switch to what is known as an "alternatively activated" phenotype or "healer" macrophage (M2) and contribute to debris scavenging, angiogenesis, and wound healing. M1 macrophages are activated by treatment with IFNγ or LPS and M2 macrophages are activated by treatment with Th2 cytokines IL-4 or IL-13 and the M2 phenotype switch can be enhanced by IL-10. Macrophages can also be skewed during differentiation in vitro, and the resultant phenotype depends upon the cytokine provided to support their differentiation. In murine macrophages, MCSF promotes differentiation to an M1 phenotype, GM-CSF promotes differentiation to an M2 phenotype and IL-3 promotes differentiation into a profoundly M2 skewed phenotype. A defining feature of the phenotype of murine M1 versus M2 macrophages is how they metabolize L-arginine. In response to an inflammatory stimulus like LPS, M1 macrophages produce inducible nitric oxide synthase (iNOS) which uses L-arginine as a substrate to produce nitric oxide (NO). M2 macrophages constitutively produce the enzyme arginase I (argI), which sequesters L-arginine from iNOS and results in the production of ornithine and downstream polyamines and L-proline. M1 macrophages also produce relatively higher levels of pro-inflammatory IL-12 and lower levels of anti-inflammatory IL-10 relative to M2 macrophages. In this chapter, we describe in vitro derivation of polarized bone marrow macrophages and methods to analyze the resulting phenotype including Q-PCR, Western blotting, and enzyme assays to determine argI and iNOS expression and activity, as well as production of IL-12p40 and IL-10 and determination of IL-12/IL-10 ratios. Production of iNOS, NO, IL-12p40, and IL-10 are measured after treatment with LPS.


Assuntos
Macrófagos/citologia , Técnicas de Cultura de Tecidos/métodos , Animais , Arginase/metabolismo , Western Blotting , Células da Medula Óssea/citologia , Diferenciação Celular , Citocinas/biossíntese , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Reação em Cadeia da Polimerase
3.
J Mol Histol ; 43(1): 95-106, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22113624

RESUMO

NAD(P) steroid dehydrogenase-like (NSDHL) is an X-linked gene that encodes a 3ß-hydroxysteroid dehydrogenase in the cholesterol biosynthetic pathway. Loss-of-function mutations in NSDHL cause Congenital Hemidysplasia with Ichthyosiform erythroderma and Limb Defects (CHILD) and CK syndromes. CHILD syndrome is a male lethal X-linked dominant disorder characterized by asymmetric skin and limb anomalies in affected females. CK syndrome is an intellectual disability disorder characterized by disproportionate short stature, brain malformations, and dysmorphic features in affected males. To understand better the relationship of the expression of mRNA and protein encoded by human NSDHL to the peripheral malformations of these disorders, we characterized the peripheral expression of the mRNA and protein by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), immunoblotting and immunohistochemistry. We also profiled the mRNA expression of mouse Nsdhl by in situ hybridization. Expression of the mRNA and protein encoded by human NSDHL parallels that of mouse Nsdhl mRNA for most but not all tissues. Furthermore, human NSDHL protein and mouse Nsdhl mRNA were expressed in tissues synthesizing cholesterol and steroids and in all peripheral tissues affected by CHILD or CK syndromes.


Assuntos
3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Perfilação da Expressão Gênica , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Adolescente , Animais , Criança , Pré-Escolar , Feminino , Feto , Humanos , Eritrodermia Ictiosiforme Congênita/genética , Eritrodermia Ictiosiforme Congênita/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Lactente , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/metabolismo , Masculino , Camundongos , Especificidade de Órgãos , Síndrome
4.
Am J Pathol ; 179(1): 180-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21640975

RESUMO

Intestinal fibrosis is a serious complication of Crohn's disease (CD) that can lead to stricture formation, which requires surgery. Mechanisms underlying intestinal fibrosis remain elusive because of a lack of suitable mouse models. Herein, we describe a spontaneous mouse model of intestinal inflammation with fibrosis and the profibrotic role of arginase I. The Src homology 2 domain-containing inositol polyphosphate 5'-phosphatase-deficient (SHIP(-/-)) mice developed spontaneous discontinuous intestinal inflammation restricted to the distal ileum starting at the age of 4 weeks. Mice developed several key features resembling CD, including inflammation and fibrosis. Inflammation was characterized by abundant infiltrating Gr-1-positive immune cells, granuloma-like immune cell aggregates that contained multinucleated giant cells, and a mixed type 2 and type 17 helper T-cell cytokine profile. Fibrosis was characterized by a thickened ileal muscle layer, collagen deposition, and increased fibroblasts at the sites of collagen deposition. SHIP(-/-) ilea had increased arginase activity and arginase I expression that was inversely proportional to nitrotyrosine staining. SHIP(-/-) mice were treated with the arginase inhibitor S-(2-boronoethyl)-l-cysteine, and changes in the disease phenotype were measured. Arginase inhibition did not affect the number of immune cell infiltrates in the SHIP(-/-) mouse ilea; rather, it reduced collagen deposition and muscle hyperplasia. These findings suggest that arginase activity is a potential target to limit intestinal fibrosis in patients with CD.


Assuntos
Arginase/metabolismo , Fibrose/enzimologia , Fibrose/patologia , Inflamação/patologia , Enteropatias/patologia , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Arginase/antagonistas & inibidores , Ácidos Borônicos/farmacologia , Citocinas/metabolismo , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/etiologia , Hiperplasia/metabolismo , Hiperplasia/patologia , Íleo/metabolismo , Íleo/patologia , Técnicas Imunoenzimáticas , Inflamação/enzimologia , Inflamação/etiologia , Inositol Polifosfato 5-Fosfatases , Enteropatias/enzimologia , Enteropatias/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Receptores de Quimiocinas/metabolismo , Células Th17
5.
J Leukoc Biol ; 90(3): 483-92, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21685246

RESUMO

mϕ are heterogeneous in their functions, and although it is clear that inflammatory mϕ contribute to inflammation in IBDs, multiple lines of evidence suggest that M2a mϕ may offer protection during intestinal inflammation. In vivo SHIP-deficient mouse mϕ are M2a so SHIP-deficient mice provide a unique genetic model of M2a mϕ. Based on this, this study tested the hypothesis that SHIP-deficient, M2a mϕ protect mice from intestinal inflammation. The objectives were to compare the susceptibility of SHIP+/+ and SHIP-/- littermates with DSS-induced intestinal inflammation and to determine whether protection was mϕ-mediated and whether protection could be transferred to a susceptible host. We have found that SHIP-/- mice are protected during DSS-induced intestinal inflammation. SHIP-/- mice have delayed rectal bleeding and reduced weight loss, disruption of intestinal architecture, and immune cell infiltration during DSS-induced colitis relative to their WT littermates. Using liposome depletion of mϕ, we found that SHIP-/- mouse protection was indeed mϕ-mediated. Finally, we determined that SHIP-/- mϕ-mediated protection could be conferred to susceptible WT mice by adoptive transfer of M2a mϕ derived ex vivo. This study supports our hypothesis by demonstrating that SHIP-deficient, M2a mϕ are protective in this murine model of acute intestinal inflammation. Adoptive transfer of M2a mϕ to patients with IBDs offers a promising, new strategy for treatment that may be particularly useful in patients who are otherwise refractory to conventional therapies.


Assuntos
Colite/patologia , Colite/prevenção & controle , Sulfato de Dextrana/toxicidade , Macrófagos/metabolismo , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Western Blotting , Células Cultivadas , Colite/induzido quimicamente , Colo/citologia , Colo/efeitos dos fármacos , Citocinas/metabolismo , Feminino , Técnicas Imunoenzimáticas , Inflamação/induzido quimicamente , Inflamação/patologia , Inflamação/prevenção & controle , Inositol Polifosfato 5-Fosfatases , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Eur J Immunol ; 41(6): 1742-53, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21469115

RESUMO

Alternatively activated macrophages are critical in host defense against parasites and are protective in inflammatory bowel disease, but contribute to pathology in asthma and solid tumors. The mechanisms underlying alternative activation of macrophages are only partially understood and little is known about their amenability to manipulation in pathophysiological conditions. Herein, we demonstrate that Src homology 2-domain-containing inositol-5'-phosphatase (SHIP)-deficient murine macrophages are more sensitive to IL-4-mediated skewing to an alternatively activated phenotype. Moreover, SHIP levels are decreased in macrophages treated with IL-4 and in murine GM-CSF-derived and tumor-associated macrophages. Loss of SHIP and induction of alternatively activated macrophage markers, Ym1 and arginase I (argI), were dependent on phosphatidylinositol 3-kinase (PI3K) activity and argI induction was dependent on the class IA PI3Kp110δ isoform. STAT6 was required to reduce SHIP protein levels, but reduced SHIP levels did not increase STAT6 phosphorylation. STAT6 transcription was inhibited by PI3K inhibitors and enhanced when SHIP was reduced using siRNA. Importantly, reducing SHIP levels enhanced, whereas SHIP overexpression or blocking SHIP degradation reduced, IL-4-induced argI activity. These findings identify SHIP and the PI3K pathway as critical regulators of alternative macrophage activation and SHIP as a target for manipulation in diseases where macrophage phenotype contributes to pathology.


Assuntos
Ativação de Macrófagos , Macrófagos/metabolismo , Neoplasias/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Arginase/genética , Arginase/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Inositol Polifosfato 5-Fosfatases , Interleucina-4/imunologia , Interleucina-4/metabolismo , Lectinas/genética , Lectinas/metabolismo , Ativação de Macrófagos/genética , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/imunologia , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/imunologia , RNA Interferente Pequeno/genética , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais/genética , Ativação Transcricional/genética , Transgenes/genética , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo , Domínios de Homologia de src/genética
7.
Am J Hum Genet ; 87(6): 905-14, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21129721

RESUMO

CK syndrome (CKS) is an X-linked recessive intellectual disability syndrome characterized by dysmorphism, cortical brain malformations, and an asthenic build. Through an X chromosome single-nucleotide variant scan in the first reported family, we identified linkage to a 5 Mb region on Xq28. Sequencing of this region detected a segregating 3 bp deletion (c.696_698del [p.Lys232del]) in exon 7 of NAD(P) dependent steroid dehydrogenase-like (NSDHL), a gene that encodes an enzyme in the cholesterol biosynthesis pathway. We also found that males with intellectual disability in another reported family with an NSDHL mutation (c.1098 dup [p.Arg367SerfsX33]) have CKS. These two mutations, which alter protein folding, show temperature-sensitive protein stability and complementation in Erg26-deficient yeast. As described for the allelic disorder CHILD syndrome, cells and cerebrospinal fluid from CKS patients have increased methyl sterol levels. We hypothesize that methyl sterol accumulation, not only cholesterol deficiency, causes CKS, given that cerebrospinal fluid cholesterol, plasma cholesterol, and plasma 24S-hydroxycholesterol levels are normal in males with CKS. In summary, CKS expands the spectrum of cholesterol-related disorders and insight into the role of cholesterol in human development.


Assuntos
3-Hidroxiesteroide Desidrogenases/genética , Anormalidades Múltiplas/genética , Alelos , Doenças Genéticas Ligadas ao Cromossomo X/genética , Temperatura , Adolescente , Adulto , Sequência de Aminoácidos , Animais , Éxons , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Homologia de Sequência de Aminoácidos , Adulto Jovem
8.
Mol Genet Metab ; 98(4): 356-66, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19631568

RESUMO

NSDHL (NAD(P)H sterol dehydrogenase-like), is a 3beta-hydroxysterol dehydrogenase thought to function in the demethylation of sterol precursors in one of the later steps of cholesterol biosynthesis. Mutations in the X-linked NSDHL gene cause CHILD syndrome in humans, and the male-lethal bare patches (Bpa) phenotype in mice. The relative level of NSDHL expression among different mouse tissues at several stages of embryogenesis and postnatal development was analyzed by immunohistochemistry. In wild type (WT) embryos, the highest levels of expression were seen in the liver, dorsal root ganglia, central nervous system, retina, adrenal gland and testis. Heterozygous Bpa(1H) females are mosaic for NSDHL expression due to normal random X-inactivation. NSDHL-deficient cells were detected in the developing cerebral cortex and retina of Bpa(1H) female embryos. In postnatal WT and Bpa(1H) animals, we compared the expression pattern of NSDHL in skin, an affected tissue; liver, a main site of cholesterol synthesis; and brain, a tissue dependent on endogenous synthesis of cholesterol due to lack of transport across the blood-brain barrier. Clonal populations of mutant cells were visible in the brain, skin and liver of Bpa(1H) pups. In the liver, the proportion of NSDHL negative cells dropped from approximately 50% at postnatal day 6 to approximately 20% at one year of age. In the brain, which showed the highest expression in cerebral cortical and hippocampal neurons, the proportion of NSDHL negative cells also dropped dramatically over the first year of life. Our results suggest that while NSDHL-deficient cells in the mosaic Bpa(1H) female are able to survive and differentiate during embryonic development, they are subject to negative selection over the life of the animal.


Assuntos
3-Hidroxiesteroide Desidrogenases/deficiência , 3-Hidroxiesteroide Desidrogenases/metabolismo , Colesterol/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , 3-Hidroxiesteroide Desidrogenases/genética , Animais , Animais Recém-Nascidos , Anticorpos/imunologia , Especificidade de Anticorpos , Western Blotting , Extratos Celulares , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes , Especificidade de Órgãos
9.
Dev Biol ; 259(1): 34-47, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12812786

RESUMO

The neural crest and sensory placodes arise from a region of the embryonic ectoderm that lies between the neural plate and future epidermis. While some of the signalling pathways that are involved in cell fate determination at the border of the neural plate have been characterised, it is still unclear how different signals are integrated. Transcription factors of the DLX gene family that may mediate such cell fate decisions are expressed at the border of the neural plate. Here, we demonstrate that DLX5 is involved in positioning this border by repressing neural properties and simultaneously by promoting the formation of border-like cells that express the neural fold markers MSX1 and BMP4 and the preplacodal region marker SIX4. However, DLX5 is not sufficient to impart epidermal character or to specify cell fates that arise at the border of the neural plate, like neural crest or fully formed sensory placodes, in a cell-autonomous manner. Additional signals are generated when mature neural plate and epidermis interact and these are required for neural crest formation. We propose that patterning of the embryonic ectoderm is a multistep process that sequentially subdivides the ectoderm into regions with defined cell fates.


Assuntos
Ectoderma/fisiologia , Estruturas Embrionárias/fisiologia , Proteínas de Homeodomínio/fisiologia , Crista Neural/citologia , Animais , Adesão Celular , Galinhas , Feminino , Morfogênese
10.
Mol Cell Biol ; 22(2): 389-99, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11756536

RESUMO

Transcriptional corepressors of the Groucho/transducin-like Enhancer of split (Gro/TLE) family regulate a number of developmental pathways in both invertebrates and vertebrates. They form transcription repression complexes with members of several DNA-binding protein families and participate in the regulation of the expression of numerous genes. Despite their pleiotropic roles, little is known about the mechanisms that regulate the functions of Gro/TLE proteins. It is shown here that Gro/TLEs become hyperphosphorylated in response to neural cell differentiation and interaction with the DNA-binding cofactor Hairy/Enhancer of split 1 (Hes1). Hyperphosphorylation of Gro/TLEs is correlated with a tight association with the nuclear compartment through interaction with chromatin, suggesting that hyperphosphorylated Gro/TLEs may mediate transcriptional repression via chromatin remodeling mechanisms. Pharmacological inhibition of protein kinase CK2 reduces the Hes1-induced hyperphosphorylation of Gro/TLEs and causes a decrease in the chromatin association of the latter. Moreover, the transcription repression activity of Gro/TLEs is reduced by protein kinase CK2 inhibition. Consistent with these observations, Gro/TLEs are phosphorylated in vitro by purified protein kinase CK2. Taken together, these results implicate protein kinase CK2 in Gro/TLE functions. They suggest further that this kinase is involved in a hyperphosphorylation mechanism activated by Hes1 that promotes the transcription repression functions of Hes1-Gro/TLE protein complexes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Repressoras/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Caseína Quinase II , Diferenciação Celular , Núcleo Celular/metabolismo , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição HES-1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...