Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de estudo
Tipo de documento
Intervalo de ano de publicação
1.
Prostaglandins Other Lipid Mediat ; 153: 106522, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33358892

RESUMO

Platelet Activating Factor (PAF) is a known phospholipid mediator of inflammation. Since its first description in 1972, it has emerged as a key regulator of vital cellular signaling functions, as proliferation, cell adhesion, and apoptosis. Evidence suggests that interactions between PAF and its receptor (PAFR) play a critical role in nervous system tissues, including the retina. The retina is a very important constituent of the visual system, along with the cornea, sclera, choroid, iris, and ciliary body, that acts synergistically to provide vision and to maintain optical homeostasis. There is evidence that PAF may regulate a wide range of physiological functions in the visual system tissues, such as eye development, inflammation, epithelial wound healing, and synapsis. Due to their multiple functions, PAF and PAFR also have important pathological and clinical implications in ocular disorders such as Choroidal Neovascularization (CNV), Age Macular Degeneration, (AMD), Diabetic Retinopathy (DR), transplant responses, and pharmacological interactions. Studies with PAFR antagonists have shown promising results such as inhibition of neovascularization and chloroquine-induced retinopathies, as well as reducing inflammation and retinal cell death. Due to the importance of PAFR signaling in the visual system and ophthalmology research, this review aims to provide a general overview of current and future perspectives about PAF in eye biology.


Assuntos
Fator de Ativação de Plaquetas , Apoptose , Humanos , Receptores Acoplados a Proteínas G , Retina
2.
Front Cell Neurosci ; 17: 1211446, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37545879

RESUMO

Background: Toxoplasmosis affects one third of the world population and has the protozoan Toxoplasma gondii as etiological agent. Congenital toxoplasmosis (CT) can cause severe damage to the fetus, including miscarriages, intracranial calcification, hydrocephalus and retinochoroiditis. Severity of CT depends on the gestational period in which infection occurs, and alterations at the cellular level during retinal development have been reported. In this study, we proposed a mouse CT model to investigate the impact of infection on retinal development. Methods: Pregnant females of pigmented C57BL/6 strain mice were infected intragastrically with two T. gondii cysts (ME49 strain) at embryonic day 10 (E10), and the offspring were analyzed at E18. Results: Infected embryos had significantly smaller body sizes and weights than the PBS-treated controls, indicating that embryonic development was affected. In the retina, a significant increase in the number of Ki-67-positive cells (marker of proliferating cells) was found in the apical region of the NBL of infected mice compared to the control. Supporting this, cell cycle proteins Cyclin D3, Cdk6 and pChK2 were significantly altered in infected retinas. Interestingly, the immunohistochemical analysis showed a significant increase in the population of ß-III-tubulin-positive cells, one of the earliest markers of neuronal differentiation. Conclusions: Our data suggests that CT affects cell cycle progression in retinal progenitor cells, possibly inducing the arrest of these cells at G2/M phase. Such alterations could influence the differentiation, anticipating/increasing neuronal maturation, and therefore leading to abnormal retinal formation. Our model mimics important events observed in ocular CT.

3.
Cell Physiol Biochem ; 28(2): 267-78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21865734

RESUMO

BACKGROUND/AIMS: Renal tubular cells are the main target of ischemic insult associated with acute renal injury. Low oxygen and nutrient supplies result in ATP depletion, leading to cell death and loss of renal function. A possible mechanism by which bone marrow-derived cells support renal tissue regeneration relies on the capacity of mononuclear cells (BMMC), particularly mesenchymal stem cells (MSC), to secrete paracrine factors that mediate support for kidney regeneration. METHODS: BMMC/MSC and renal cells (LLC-PK(1) from pig and IRPTC from rat) were co-cultured under stressful conditions (ATP depletion and/or serum free starvation), physically separated by a microporous membrane (0.4 µm), was used to determine whether bone marrow-derived cells can interact with renal cells in a paracrine manner. RESULTS: This interaction resulted in stimulation of renal cell proliferation and the arrest of cell death. MSC elicit effective responses in renal cells in terms of stimulating proliferation and protection. Such effects are observed in renal cells co-cultured with rat BMMC/MSC, an indication that paracrine mechanisms are not entirely species-specific. CONCLUSION: The paracrine action of BMMC/MSC was influenced by a renal cell stimulus released during stress, indicating that cross-talk with injured cells is required for renal regeneration supported by bone marrow-derived cells.


Assuntos
Células da Medula Óssea/citologia , Células Epiteliais/citologia , Túbulos Renais Proximais/citologia , Células-Tronco Mesenquimais/citologia , Comunicação Parácrina/fisiologia , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura Livres de Soro , Masculino , Ratos , Ratos Wistar , Suínos
4.
Mol Neurobiol ; 55(11): 8612-8624, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29574630

RESUMO

Adenine nucleotides through P2Y1 receptor stimulation are known to control retinal progenitor cell (RPC) proliferation by modulating expression of the p57KIP2, a cell cycle regulator. However, the role of Gi protein-coupled P2Y12 and P2Y13 receptors also activated by adenine nucleotides in RPC proliferation is still unknown. Gene expression of the purinergic P2Y12 subtype was detected in rat retina during early postnatal days (P0 to P5), while expression levels of P2Y13 were low. Immunohistochemistry assays performed with rat retina on P3 revealed P2Y12 receptor expression in both Ki-67-positive cells in the neuroblastic layer and Ki-67-negative cells in the ganglion cell layer and inner nuclear layer. Nonetheless, P2Y13 receptor expression could not be detected in any stratum of rat retina. Intravitreal injection of PSB 0739 or clopidogrel, both selective P2Y12 receptor antagonists, increased by 20 and 15%, respectively, the number of Ki-67-positive cells following 24 h of exposure. Moreover, the P2Y12 receptor inhibition increased cyclin D1 and decreased p57KIP2 expression. However, there were no changes in the S phase of the cell cycle (BrdU-positive cells) or in mitosis (phospho-histone-H3-positive cells). Interestingly, an increase in the number of cyclin D1/TUNEL-positive cells after treatment with PSB 0739 was observed. These data suggest that activation of P2Y12 receptors is required for the successful exit of RPCs from cell cycle in the postnatal rat retina.


Assuntos
Organogênese , Receptores Purinérgicos P2Y12/metabolismo , Receptores Purinérgicos P2/metabolismo , Retina/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Ratos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y12/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
5.
Int J Dev Neurosci ; 25(8): 499-508, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17981424

RESUMO

ATP and ADP induce retinal cell proliferation through activation of PKC and extracellular signal-regulated kinases (ERKs). Here, we characterized the effect of purinergic agonists on the turnover of phosphoinositides and activation of ERKs during development of the chick embryo retina. When intact retinas were incubated with ATP, ADP or UTP, a dose-dependent accumulation of [(3)H]-phosphoinositides was observed (% of control, EC(50): 548+/-20.5%, 0.18 mM; 314+/-53.8%, 0.51 mM; 704+/-139.9%, 0.018 mM, respectively). Only the response promoted by ADP was completely inhibited by the P2 receptor antagonists, PPADS and suramin. All the responses decreased with the progression of retinal development. Western blot assays revealed that ATP, ADP and UTP stimulated the phosphorylation of ERKs in the chick embryo retina very early during development (% of control: 174+/-16; 199+/-16.4 and 206+/-37, respectively). The responses to ADP and UTP were transient and dose-dependent, showing EC(50) values of 0.12 mM and 0.009 mM. The response to ADP was inhibited by the antagonists PPADS and suramin and by U73122 and chelerythrine chloride, which block PLC and PKC, respectively. Conversely, chelerythrine chloride did not block the response induced by UTP. Immunohistochemical analysis revealed that ATP and ADP induced the phosphorylation of ERKs in cells of the neuroblastic layer of retinas from embryos at E8. Our data showed that ATP, ADP and UTP stimulate the turnover of InsPs and promoted the activation of ERKs in the chick embryo retina. ADP, through activation of P2Y(1) receptors, activated ERK pathway through PLC and PKC and UTP, via P2Y(4)-like receptors, induced the phosphorylation of ERKs through a pathway that did not involve PKC.


Assuntos
Trifosfato de Adenosina/farmacologia , Retina/citologia , Retina/embriologia , Transdução de Sinais/fisiologia , Difosfato de Adenosina/farmacologia , Animais , Western Blotting , Bromodesoxiuridina , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , DNA/biossíntese , Imuno-Histoquímica , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Técnicas de Cultura de Órgãos , Fosfatidilinositóis/metabolismo , Receptores Purinérgicos P2/fisiologia , Receptores Purinérgicos P2Y1 , Retina/metabolismo , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Timidina/metabolismo , Fixação de Tecidos , Uridina Trifosfato/farmacologia
6.
Mol Neurobiol ; 54(7): 5142-5155, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27558237

RESUMO

Previous studies demonstrated that exogenous ATP is able to regulate proliferation of retinal progenitor cells (RPCs) in vitro possibly via P2Y1 receptor, a G protein-coupled receptor. Here, we evaluated the function of adenine nucleotides in vivo during retinal development of newborn rats. Intravitreal injection of apyrase, an enzyme that hydrolyzes nucleotides, reduced cell proliferation in retinas at postnatal day 2 (P2). This decrease was reversed when retinas were treated together with ATPγ-S or ADPß-S, two hydrolysis-resistant analogs of ATP and ADP, respectively. During early postnatal days (P0 to P5), an increase in ectonucleotidase (E-NTPDase) activity was observed in the retina, suggesting a decrease in the availability of adenine nucleotides, coinciding with the end of proliferation. Interestingly, intravitreal injection of the E-NTPDase inhibitor ARL67156 increased proliferation by around 60 % at P5 rats. Furthermore, immunolabeling against P2Y1 receptor was observed overall in retina layers from P2 rats, including proliferating Ki-67-positive cells in the neuroblastic layer (NBL), suggesting that this receptor could be responsible for the action of adenine nucleotides upon proliferation of RPCs. Accordingly, intravitreal injection of MRS2179, a selective antagonist of P2Y1 receptors, reduced cell proliferation by approximately 20 % in P2 rats. Moreover, treatment with MRS 2179 caused an increase in p57KIP2 and cyclin D1 expression, a reduction in cyclin E and Rb phosphorylated expression and in BrdU-positive cell number. These data suggest that the adenine nucleotides modulate the proliferation of rat RPCs via activation of P2Y1 receptors regulating transition from G1 to S phase of the cell cycle.


Assuntos
Difosfato de Adenosina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Receptores Purinérgicos P2Y1/metabolismo , Retina/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Difosfato de Adenosina/farmacologia , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Bromodesoxiuridina/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ratos , Retina/metabolismo , Células-Tronco/citologia
7.
Brain Res ; 947(1): 78-83, 2002 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-12144855

RESUMO

Pharmacological blockers of cyclin-dependent kinases (CDKs) can inhibit cell cycle progression. Deferoxamine (DFO) and mimosine (MIMO) arrest cells reversibly at the G1/S transition and olomoucine (OLO) inhibits the cell cycle at both G1/S and G2/M. We investigated the effect of these drugs upon cell death in histotypical explants taken from the retina of neonatal rats. Degeneration of retinal ganglions cells (RGC) induced by axotomy was inhibited by OLO (100 microM) but not by DFO (up to 2 mM) or MIMO (up to 1 mM). On the other hand, after 1 day in vitro, all cell cycle inhibitors induced cell death in the neuroblastic layer (NBL) of the explants. DFO and MIMO induced cell death only of proliferating cells, identified either by their incorporation of bromodeoxyuridine or by immunolabeling the proliferating cell nuclear antigen. In turn, OLO induced cell death of both proliferating and post-mitotic cells. However, the post-mitotic cells were unlabeled with markers of retinal differentiation. Our results indicate that cyclin-dependent kinases are involved in the control of sensitivity to cell death in the retina, and that retinal cells present differentiation-dependent responses to modulation of CDK activity.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Retina/citologia , Retina/crescimento & desenvolvimento , Adenilil Ciclases/metabolismo , Animais , Anisomicina/farmacologia , Antivirais , Axotomia , Bromodesoxiuridina , Contagem de Células , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Colforsina/farmacologia , Técnicas de Cultura , DNA/biossíntese , DNA/genética , Ativação Enzimática/efeitos dos fármacos , Imuno-Histoquímica , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Retina/efeitos dos fármacos , Degeneração Retiniana/patologia
8.
PLoS One ; 8(1): e53517, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23301080

RESUMO

PI3K/Akt is an important pathway implicated in the proliferation and survival of cells in the CNS. Here we investigated the participation of the PI3K/Akt signal pathway in cell cycle of developing retinal progenitors. Immunofluorescence assays performed in cultures of chick embryo retinal cells and intact tissues revealed the presence of phosphorylated Akt and 4E-BP1 in cells with typical mitotic profiles. Blockade of PI3K activity with the chemical inhibitor LY 294002 (LY) in retinal explants blocked the progression of proliferating cells through G2/M transition, indicated by an expressive increase in the number of cells labeled for phosphorylated histone H3 in the ventricular margin of the retina. No significant level of cell death could be detected at this region. Retinal explants treated with LY for 24 h also showed a significant decrease in the expression of phospho-Akt, phospho-GSK-3 and the hyperphosphorylated form of 4E-BP1. Although no change in the expression of cyclin B1 was detected, a significant decrease in CDK1 expression was noticed after 24 h of LY treatment both in retinal explants and monolayer cultures. Our results suggest that PI3K/Akt is an active pathway during proliferation of retinal progenitors and its activity appears to be required for proper CDK1 expression levels and mitosis progression of these cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/embriologia , Retina/metabolismo , Células-Tronco/citologia , Animais , Ciclo Celular , Divisão Celular , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Cromonas/farmacologia , Ciclina B1/metabolismo , Fase G2 , Histonas/metabolismo , Microscopia de Fluorescência , Mitose , Morfolinas/farmacologia , Fosforilação , Retina/citologia , Transdução de Sinais , Fatores de Tempo
9.
PLoS One ; 6(1): e16058, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21298035

RESUMO

Nuclear migration is regulated by the LIS1 protein, which is the regulatory subunit of platelet activating factor (PAF) acetyl-hydrolase, an enzyme complex that inactivates the lipid mediator PAF. Among other functions, PAF modulates cell proliferation, but its effects upon mechanisms of the cell cycle are unknown. Here we show that PAF inhibited interkinetic nuclear migration (IKNM) in retinal proliferating progenitors. The lipid did not, however, affect the velocity of nuclear migration in cells that escaped IKNM blockade. The effect depended on the PAF receptor, Erk and p38 pathways and Chk1. PAF induced no cell death, nor a reduction in nucleotide incorporation, which rules out an intra-S checkpoint. Notwithstanding, the expected increase in cyclin B1 content during G2-phase was prevented in the proliferating cells. We conclude that PAF blocks interkinetic nuclear migration in retinal progenitor cells through an unusual arrest of the cell cycle at the transition from S to G2 phases. These data suggest the operation, in the developing retina, of a checkpoint that monitors the transition from S to G2 phases of the cell cycle.


Assuntos
Núcleo Celular/fisiologia , Fase G2 , Fator de Ativação de Plaquetas/fisiologia , Glicoproteínas da Membrana de Plaquetas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Fase S , Animais , Transporte Biológico , Proliferação de Células , Quinase 1 do Ponto de Checagem , MAP Quinases Reguladas por Sinal Extracelular , Proteínas Quinases , Ratos , Retina/citologia , Células-Tronco , Proteínas Quinases p38 Ativadas por Mitógeno
10.
Int J Dev Neurosci ; 28(1): 63-73, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19799993

RESUMO

Previous data suggest that nucleotides are important mitogens in the developing chick retina. Here, we extended the study on the mitogenic effect of ATP to newborn mouse retinal explants. Our results showed that P2Y(1) receptors were widely distributed in C57bl/6 mice retina and that the majority of PCNA positive cells co-localized with P2Y(1) receptor. To evaluate proliferation, retinal explants obtained from newborn mice were incubated with 0.5 microCi [(3)H]-thymidine or 3 microM BrDU 1h before the end of culture. Our data showed that ATP induced a dose-dependent increase in [(3)H]-thymidine incorporation, an effect that was mimicked by ADP but not by UTP and was blocked by the P2 antagonist PPADS in a dose-dependent manner. The increase in [(3)H]-thymidine incorporation induced by ATP was only observed in explants cultured for 3 days or less and was mimicked by the ectoapyrase inhibitor ARL 67156. It corresponded to an increase in the number of BrdU(+) cells in the neuroblastic layer (NL) of the tissue, suggesting that ATP, through activation of P2Y(1) receptors, induced proliferation of late developing progenitors in retinal explants of newborn mice. The increase in the number of BrdU(+) cells was observed across the whole NL when explants were incubated with ATP for 24h and no increase in the number of p-histone H3 labeled cells could be noticed at this time point. In longer incubations of 48h with ATP or 24h with ATP followed by a period of 24h in fresh medium, an increase in the number of BrdU(+) cells promoted by ATP was observed only in the middle and outer, but not in the inner NL. In these conditions, an increase in the number of p-histone H3 labeled cells was detected in the outer NL, suggesting that ATP induced cells to enter S and progress to G2 phase of the cell cycle in the first 24h period of incubation. ATP also induced an increase and a decrease in the expression of cyclin D1 and p27(kip1), respectively, in retinal progenitors of the NL. While the increase in the expression of cyclin D1 was observed when retinal explants were incubated for 3h or longer periods of time, the decrease in the expression of p27(kip1) was noticed only after 6h incubation with ATP. Both effects were blocked by the P2 receptor antagonist PPADS. These data suggest that ATP induces cell proliferation in retinal explants by inducing late developing progenitors to progress from G1 to S phase of cell cycle.


Assuntos
Trifosfato de Adenosina/metabolismo , Ciclo Celular/fisiologia , Proliferação de Células , Retina/crescimento & desenvolvimento , Retina/fisiologia , Animais , Animais Recém-Nascidos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Histonas/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Antígeno Nuclear de Célula em Proliferação/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Retina/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Fatores de Tempo
11.
Dev Neurobiol ; 68(5): 620-31, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18278803

RESUMO

In developing retina, the nucleus of the elongated neuroepithelial cells undergoes interkinetic nuclear migration (INM), that is it migrates back and forth across the proliferative layer during the cell cycle. S-phase occurs at the basal side, while M-phase occurs at the apical margin of the retinal progenitors. G1 and G2-phases occur along the nuclear migration pathway. We tested whether this feature of the retinal cell cycle is controlled by CK2, which, among its many substrates, phosphorylates both molecular motors and cytoskeletal components. Double immunolabeling showed that CK2 is contained in BrdU-labeled retinal progenitors. INM was examined after pulse labeling the retina of newborn rats with BrdU, by plotting nuclear movement from basal to apical sides of the retinal progenitors during G2. The CK2 specific inhibitor 4,5,6,7-tetrabromobenzotriazole inhibited the activity of rat retinal CK2, and blocked nuclear movement proper in a dose-dependent way. No apoptosis was detected, and total numbers of BrdU-labeled nuclei remained constant following treatment. Immunohistochemistry showed that, following inhibition of CK2, the tubulin cytoskeleton is disorganized, with reduced acetylated and increased tyrosinated tubulin. This indicates a reduction in stable microtubules, with accumulation of free tubulin dimers. The results show that CK2 activity is required for INM in retinal progenitor cells.


Assuntos
Caseína Quinase II/fisiologia , Ciclo Celular/fisiologia , Núcleo Celular/enzimologia , Retina/citologia , Retina/fisiologia , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Antimetabólitos , Western Blotting , Bromodesoxiuridina , Caseína Quinase II/genética , Núcleo Celular/fisiologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Imunofluorescência , Imuno-Histoquímica , Nucleotídeos/metabolismo , Ratos , Retina/enzimologia , Células-Tronco/enzimologia , Triazóis , Tubulina (Proteína)/biossíntese , Tubulina (Proteína)/genética
12.
Biochemistry ; 46(4): 987-96, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17240982

RESUMO

Plant defensins, components of the plant innate immune system, are cationic cysteine-rich antifungal peptides. Evidence from the literature [Thevissen, K., et al. (2003) Peptides 24, 1705-1712] has demonstrated that patches of fungi membrane containing mannosyldiinositolphosphorylceramide and glucosylceramides are selective binding sites for the plant defensins isolated from Dahlia merckii and Raphanus sativus, respectively. Whether plant defensins interact directly or indirectly with fungus intracellular targets is unknown. To identify physical protein-protein interactions, a GAL4-based yeast two-hybrid system was performed using the antifungal plant peptide Pisum sativum defensin 1 (Psd1) as the bait. Target proteins were screened within a Neurospora crassa cDNA library. Nine out of 11 two-hybrid candidates were nuclear proteins. One clone, detected with high frequency per screening, presented sequence similarity to a cyclin-like protein, with F-box and WD-repeat domains, related to the cell cycle control. GST pull-down assay corroborated in vitro this two-hybrid interaction. Fluorescence microscopy analysis of FITC-conjugated Psd1 and DAPI-stained fungal nuclei showed in vivo the colocalization of the plant peptide Psd1 and the nucleus. Analysis of the DNA content of N. crassa conidia using flow cytometry suggested that Psd1 directed cell cycle impairment and caused conidia to undergo endoreduplication. The developing retina of neonatal rats was used as a model to observe the interkinetic nuclear migration during proliferation of an organized tissue from the S toward the M phase of the cell cycle in the presence of Psd1. The results demonstrated that the plant defensin Psd1 regulates interkinetic nuclear migration in retinal neuroblasts.


Assuntos
Ciclinas/metabolismo , Defensinas/metabolismo , Proteínas Fúngicas/metabolismo , Neurospora crassa/metabolismo , Proteínas de Plantas/metabolismo , Animais , Animais Recém-Nascidos , Antifúngicos/metabolismo , Sequência de Bases , Sítios de Ligação , Ciclo Celular , Ciclinas/genética , DNA Fúngico/genética , DNA de Plantas/genética , Defensinas/genética , Defensinas/farmacologia , Proteínas Fúngicas/genética , Neurospora crassa/citologia , Neurospora crassa/genética , Pisum sativum/genética , Pisum sativum/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/farmacologia , Ligação Proteica , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Retina/citologia , Retina/efeitos dos fármacos , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA